Special Issue


Synthetic and Systems Biotechnology in Food Sciences

Mostafa Rezaei-Tavirani, Masoumeh Farahani

Applied Food Biotechnology, Vol. 11 No. 2 (2024), 26 Aban 2024, Page e1
https://doi.org/10.22037/afb.v11i2.47151

Background and Objective: Food biotechnology significantly impacts the food industry and human health. Many advanced investigations are involved in the ongoing development of food quality and safety. Introducing the critical cutting-edge features of research in food biotechnology is the main aim of this special issue. Application of nanotechnology as a multidisciplinary field to develop food safety and production, using microbiota as the modulators of body immunity against stresses, introducing bioinformatics tools to evaluate food microbial contamination, using systems biology assessment to explore the efficient and safe food preservative agents, evaluating food bioprocess protocols such as fermentation, presentation of advanced bioremediation techniques for detoxification of environmental toxins heavy metals, and highlighting significant progress in engineered edible probiotic vaccines are presented and discussed to provide new perspective of food biotechnology. Understanding and using the close relationship between technology development and food quality and safety improvement is accentuated as a continuous effort in food biotechnology research.

Results and Conclusion: The special issue illustrates the impact of multi-disciplinary research and highlights the need to develop new technologies in the food industry, especially multi-omics systems biology technologies and synthetic biotechnology. The major three topics presented based on systems biology include role of biopreservative agents in food safety. Collaboration between traditional knowledge and newly introduced approaches will accelerate future food biotechnology achievements.

Conflict of interest: The authors declare no conflict of interest.

  1. Introduction

 

The food industry and biotechnology development are associated closely with human health and lifestyle investigations. Indeed, different roles of biotech-nology may lead to increased immunity and safety in food sectors, e.g., by incorporating microalgae and/ or probiotics into food and microbial metabolites with medicinal and industrial applications [1-10]. So, evaluating the latest technological trends and advan-cements is critical in food biotechnology. Advanced efficient production, quality maintenance during storage, and achieving food safety standards require alterations and progressions in the related technol-ogies. The human body's response to the intake of certain foods in the short and long term is the best tool to display the quality and safety of diet and the applied technology. The high throughput methods such as genomics, transcriptomics, proteomics, and metabol-omics accompanied by bioinformatics have attracted the attention of experts in biology, medicine, pharmacology, and food sciences and technologies. Application of artificial intelligence, introduction of advanced software and databases, and representation of different models; have provided a new window to solve the complex problems in the various fields of human health. Today, the systems biology approach is a well-known trend for investigating the unsolved complications in the mentioned arenas. It is expected that the mass production of data and optimum organization and interpretation of findings could create a new perspective on human needs and environmental sources such as food. Collaboration between traditional knowledge and newly introduced approaches will accelerate future food biotechnology achievements. The articles presented in this research topic provide an overview of the impact of multi-disciplinary research and highlight the need to develop new technologies in the food industry, especially multi-omics systems biology technologies and syn-thetic biotechnology.

Three systems biology-based articles have been presented on this research topic.

Optimizing preservative agents using biotech-nology plays a crucial role in food safety. Evaluation of nisin effect on human health versus sodium benzoate (a chemical preservative) depicted in the research by Bandarian et al. [11]. The biological impact of yoghurt fermentation on human health is discussed in the study by Rostami Nejad et al. [12]. It is planned to find a suitable method to decrease the harmful effects of aflatoxin on the human body by Hamzehloo-Moghadam et al. [13].

A review article on this research topic addresses the issue of probiotics as dietary supplements and oral vaccines, which are an effective factor in human health. The potential of edible vaccines has been determined as safe vaccines in possible pathogenicity, good stimulation of mucosal and systematic immunity, and reasonable cost due to the lack of complex purification processes. Therefore, investigating the effectiveness of engineered probiotics based on novel genome editing tools and their challenges in genera-ting edible probiotic vaccines is to be considered [14]. Moreover, probiotics as live microorganisms can improve the gut microbiome to expand the optimistic role of the gut microbiome against the hostile side effects of chemotherapy and radiotherapy [15]. Chemotherapy and radiotherapy, the two common cancer treatment methods are accompanied by considerable side effects. Investigations have demon-strated substantial gastrointestinal disorders associated with chemotherapy and radiotherapy [16,17]. Evidence has pointed out the positive roles of microbiota in modifying these adverse effects and their severity in treated patients [18,19]. Today, probiotics are highlighted in food biotechnology and industry as an essential compound of several foods.

The environment contains essential elements for life, but industrialization has worsened pollution, especially from heavy metals, which threaten human health and ecosystems. The discussion presented by Fateminasab et al. highlights the rise of environmental toxins and the limitations of traditional pollution control, emphasizing the urgent need for advanced bioremediation techniques [20]. Toxic heavy metals like arsenic, cadmium, chromium, mercury, and lead pose serious health risks, particularly to children, with exposure occurring through contaminated food, water, and air. Bioremediation uses living organisms to detoxify pollutants. Probiotics can effectively elimin-ate these toxins from food. The report showcases microbes' potential in detoxifying heavy metals and notes the success of engineered microorganisms like recombinant E. coli in degrading waste and pollutants. Environmental factors such as humidity and temper-ature also impact microbial degradation. Additionally, various microbial species play a crucial role in detoxif-ying contaminants in water, soil, and air, with biofilm-mediated bioremediation emerging as a promising strategy for effective pollutant removal [20-24].

Finally, a review by Koushki et al. focuses on nanotechnology applications in the food industry [25]. They highlighted the considerable role of linking food science and nanotechnology as a multidisciplinary scientific field in protecting food safety, production, processing, and quality control.

  1. Conclusion

The special issue illustrates the impact of multi-disciplinary research and highlights the need to develop new technologies in the food industry, especially multi-omics systems biology technologies, and synthetic biotechnology. The major three topics presented based on systems biology include the role of bio-preservative agents in food safety. Collaboration between traditional knowledge and newly introduced approaches will accelerate future food biotechnology achievements.

  1. Acknowledgements

The authors appreciate all scientific support of the Research Vice-chancellor at Shahid Beheshti Univer-sity of Medical Sciences and Proteomics Research Center.

  1. Conflict of Interest

The authors declare no conflict of interest.

References

  1. Toranposhti TH, Hosseini FS, Rezaei M, Tahamtani Y. Bioactivity assay of microalgae: antioxidant and antidiabetic potentials in a transgenic zebrafish model. Appl Food Biotechnol. 2024; 11(1): e25.

https://doi.org/10.22037/afb.v11i1.45488

  1. Lestari SD, Hussain N, Hussin AS, Mustafa S, Sew YS. Bacillus multi-strain from malaysian fish sauces demonstrating proteolytic, lipolytic, esterolytic and glutamic-acid production activities. Appl Food Biotechnol. 2024; 11(1): e26.

https://doi.org/10.22037/afb.v11i1.45507

  1. Evelyn E, Chairul C, Sriningsih S, Aulia Y. Pulsed electric-field processing of orange juice containing paecilomyces variotii spores: comparisons to power ultrasound and thermal treatments. Appl Food Biotechnol. 2024; 11(1): e27.

https://doi.org/10.22037/afb.v11i1.45741

  1. Salimi F, Ahari H, Anvar SA, Moghanjoghi AM. Investigating antibacterial effects of nano-Ag/titanium dioxide on polylactic acid nanocomposites produced using extrusion method. Appl Food Biotechnol. 2024; 11(1): e28.

https://doi.org/10.22037/afb.v11i1.45932

  1. Shaikh SS, Joshi C, Malek F, Malik A, Gandhi M. Food Storage, Processing and genetic stability studies of Bacillus (Heyndrickxia) coagulans BCP92 (MTCC 25460). Appl Food Biotechnol. 2024; 11(1): e22.

https://doi.org/10.22037/afb.v11i1.44919

  1. Puspita AE, Pangastuti A, Listyawati S, Sari SL. Investigating untapped potentials: velvet beans as novel prebiotic sources and their effects on gut microbiota and short-chain fatty acid level. Appl Food Biotechnol. 2024; 11(1): e17.

https://doi.org/10.22037/afb.v11i1.44643

  1. Tsai CC, Lai ZY. Rapid identification and application of Lactobacillus plantarum, Lactobacillus paracasei and Lactobacillus pentosus using multiplex polymerase chain reaction and species-specific primers, targeting 16S ribosomal RNA and recA genes. Appl Food Biotechnol. 2024; 11(1): e24.

https://doi.org/10.22037/afb.v11i1.44863

  1. Sağlam H. Incorporation of microencapsulated and free Lactiplantibacillus plantarum to bitter chocolate: sensory and survival analyses. Appl Food Biotechnol. 2024; 11(1): e18.

https://doi.org/10.22037/afb.v11i1.44424

  1. Mazinani S, Fadaei V, Khosravi-Darani K. Impact of Spirulina platensis on physicochemical properties and viability of Lactobacillus acidophilus of probiotic UF Feta Cheese. J. Food Process Preserv. 2016; 40 (6): 1318-1324.

https://doi.org/10.1111/jfpp.12717

  1. Hooshdar P, Kermanshahi RK, Ghadam P, Khosravi-Darani K. A review on production of exopolysaccharide and biofilm in probiotics like lactobacilli and methods of analysis. Biointerface Res Appl Chem. 2020; 10(5): 6058-6075.

https://doi.org/10.33263/BRIAC105.60586075

  1. Bandarian F, Razi F, Razzaghi Z, Rostami Nejad M, Arjmand B, Rezaei-Tavirani M. The effect of sodium benzoate and nisin on human HepG2 cell gene expression. Appl Food Biotechnol. 2024; 11(2): e3.

https://doi.org/10.22037/afb.v11i2.45959

  1. Rostami Nejad M, Razi F, Razzaghi Z, Bandarian F, Arjmand B, Rezaei-Tavirani M. Assessing biological effects of yoghurt consumption against acidified milk: A System biology study. Appl Food Biotechnol. 2024; 11(2): e2.

https://doi.org/10.22037/afb.v11i2.45910

  1. Hamzehloo-Moghadam M, Asri N, Jahani Sherafat S, Mansouri V. Detection of aflatoxin contamination through biomarker discovery in human intestinal Caco-2 cells. Appl Food Biotechnol. 2024; 11(2): e4.

https://doi.org/10.22037/afb.v11i2.46449

  1. Hashemi N, Shams F, Sharif E, Bakhshi Valilou M. Recent advances in engineered edible probiotic vaccines: Promising agents in the effectiveness of probiotics. Appl Food Biotechnol. 2024; 11(2): e5

https://doi.org/10.22037/afb.v11i2.46302

  1. Talaei Mashhadi MR, Mousavi M, Khalili-Tanha G, Nazari E. Probiotics as an adjunct therapy for decreasing gastrointestinal complications from chemoradiotherapy. Appl Food Biotechnol. 2024; 11(2): e6

https://doi.org/10.22037/afb.v11i2.46338

  1. Nurgali K, Jagoe RT, Abalo R. Editorial: Adverse effects of cancer chemotherapy: Anything new to improve tolerance and reduce sequelae? Front Pharmacol. 2018; 9:245.

https://doi.org/10.3389/fphar.2018.00245

  1. Sipaviciute A, Sileika E, Burneckis A, Dulskas A. Late gastrointestinal toxicity after radiotherapy for rectal cancer: A systematic review. Int J Colorectal Dis. 2020; 35(6):977-983.

https://doi.org/10.1007/s00384-020-03595-x

  1. Tonneau M, Elkrief A, Pasquier D, Paz Del Socorro T, Chamaillard M, Bahig H, Routy B. The role of the gut microbiome on radiation therapy efficacy and gastrointestinal complications: A systematic review. Radiother Oncol. 2021; 156: 1-9.

https://doi.org/10.1016/j.radonc.2020.10.033

  1. Yin B, Wang X, Yuan F, Li Y, Lu P. Research progress on the effect of gut and tumor microbiota on antitumor efficacy and adverse effects of chemo-therapy drugs. Front Microbiol. 2022; 13: 899111.

https://doi.org/10.3389/fmicb.2022.899111

  1. Fateminasab F, Koushki M, Jafarian F, Mansouri V, Ahmadzadeh A, Amiri-Dashatan N, Farahani M. evaluating the efficacy of microbial interventions in heavy metal decontamination in the environment and food production systems. Appl Food Biotechnol. 2024; 11(2) e8.

http://doi.org/10.22037/afb.v11i2.46832

  1. Zoghi A, Khosravi‐Darani K, Sohrabvandi S, Attar H. Patulin removal from synbiotic apple juice using Lactobacillus plantarum ATCC 8014. J Appl Micro-biol. 2019; 126(4): 1149-1160.

https://doi.org/10.1111/jam.14172

  1. Hadiani MR, Khosravi-Darani K, Rahimifard N, Younesi H. Assessment of mercury biosorption by Saccharomyces cerevisiae: response surface method-logy for optimization of low Hg (II) concentrations. J Environ Chem Eng. 2018; 6(4): 4980-4987.

https://doi.org/10.1016/j.jece.2018.07.034

  1. Zoghi A, Salimi M, Mirmahdi RS, Massoud R, Khosravi-Darani K, Mohammadi R, Rouhi M, Tripathy AD. Effect of pretreatments on bioremoval of metals and subsequent exposure to simulated gastrointestinal conditions. Qual Assur Saf Crops Foods. 2022; 14(3): 145-155.

https://doi.org/10.15586/qas.v14i3.1012

  1. Sarlak Z, Khosravi-Darani K, Rouhi M, Garavand F, Mohammadi R, Sobhiyeh MR. Bioremediation of organophosphorus pesticides in contaminated food-stuffs using probiotics. Food Control. 2021; 126: 108006.

https://doi.org/10.1016/j.foodcont.2021.108006

  1. Koushki M, Amiri-Dashatan N, Pourghadamyari H, Khodabandehloo H, Bagheri F, Farahani M, Tayebi L. An overview of the relationship between the food industry and nano-technology. Appl Food Biotechnol. 2024; 11(2) e7.

http://dx.doi.org/10.22037/afb.v11i2.46737

Assessing Biological Effects of Yoghurt Consumption against Acidified Milk: A System Biology Study

Mohammad Rostami Nejad, Farideh Razi, Zahra Razzaghi, Fatemeh Bandarian, Babak Arjmand, Mostafa Rezaei-Tavirani

Applied Food Biotechnology, Vol. 11 No. 2 (2024), 26 Aban 2024, Page e2
https://doi.org/10.22037/afb.v11i2.45910

Background and Objective: Yoghurt is a fermented milk product by bacteria; a process including transformation of lactose and galactose to lactic acid. In other words, milk acidification is a critical step in the industrial process to produce various dairy foods and components such as yogurt and caseinates. This study aimed to assess yoghurt effects on gene expression of human whole blood against acidified milk.

Material and Methods: Whole blood gene expression changes of yoghurt consumers against individuals that received acidified milks were retrieved from gene expression omnibus (GEO) database and pre-assessed via GEO2R program to find significant differentially expressed genes (DEG). Significant DEGs were assessed via director protein-protein interaction (PPI) network analysis and gene ontology enrichment to investigate critical genes and targeted biological processes.

Results and Conclusion: Pre-assessment analysis showed that whole blood gene expression profiles of the yoghurt group changed (characterized by 37 significant DEGs) while samples of acidified milk consumers included no significant alterations. Moreover, PPI network analyses showed that RPSA, RPS5, RPS14, PABPC1, DDX60L, FEN1, MRPL12 and KAT6A were the highlighted significant DEGs. Based on the gene ontology, enrichment biosynthesis of ceruloplasmin was addressed as the targeted biological process.

It was concluded that yoghurt included significant effects on gene expression profiles of whole blood while acidified milk did not. Downregulation of genes that were involved in ceruloplasmin production and function was highlighted as the major event in blood of yoghurt consumers.

Conflict of interest: The authors declare no conflict of interest.

 

  1. Introduction

 

Lactic acid bacteria are used to convert milk to yoghurt via the fermentation process. Lactose and galactose of milk transform to lactic acid via fermentation; a process which is accompanied with nutrient, physical and chemical modifications of the milk matrix [1]. Several factors such as starter cultures, fermentation conditions and milk characteristics can affect milk fermentation for yoghurt production. Improvement of the fermentation process leads to the production of quality products [2]. There are evidence that lactic acid bacteria modify microbiota of the yoghurt consumers through fermentation [3]. Acidified milk foods are well-known products worldwide [4]. Milk acidification is an essential step of the industrial production process of various dairy products and components such as cheeses, yogurts and caseinates [5]. Investigations indicate that glucono-δ-lactone as a milk acidifier induces coagulation of milk proteins [6]. Studies have shown advantages of fermented dairy consumption such as decreased lipid parameters, decreased circulating parameters of inflammation and regulation of glycemia [7-9]. Significant characteristics of milk fermentation that affects metabolism correspond to lactose metabolism in people worldwide, who cannot eat milk due to its lactose [10].

Liver plays critical roles in copper homeostasis. Investigations indicate that most of the copper is transferred from the liver to plasma in form of ceruloplasmin [11]. Ceruloplasmin is involved in the metabolic balance of iron. Contribution of ceruloplasmin in neurodegenerative diseases such as Alzheimer’s disease, Wilson’s disease and Parkinson’s disease is verified by the researchers. Its roles in metabolic diseases such as obesity, diabetes and hyperlipidemia are reported by the researchers as well [12].

Sample collection is a critical step in analytical experiments. Whole blood is an available source of genomic DNA, which has widely been used in services worldwide [13,14]. High-throughput technologies such as genomics, proteomics and transcriptomics can provide large numbers of data on gene products [15]. Bioinformatics as an appropriate tool is close to genomics to solve numerous problems in medicine and biology [16]. The PPI network analysis as a bioinformatics technique has been interested by the scientists to analyze genomic data. Genes or proteins are included in networks and play various roles in the networks based on their characteristics. The PPI network analysis is used in various fields of medicine and nutrition [17-19]. There are investigations about the effects of yoghurt consumption on gene expression profiles of blood cells. Roles of yoghurt and acidified milk consumptions in modulation of inflammation are documented [20]. In the present study, transcriptomic data of the whole blood of seven young men, who consumed yoghurt and acidified milk, were extracted from the GEO database and compared to each other using PPI network analysis. Critical genes were identified and assessed via gene ontology to find targeted biological terms. Findings open new windows to possible advantages of yoghurt consumption as a public nutrient.

 

  1. Materials and Methods

2.1. Data collection

To investigate effects of fermentation on the quality of milk and the consequence results on human health, GSE98645 was searched within the GEO database. Whole blood transcriptomes of seven healthy young men (mean (±SEM) age of 24.6 y ±4.7) after consuming probiotic yoghurt or acidified milk were compared with those before interventions. Data were published as an original article [21]. The yoghurt was fermented by Lactobacillus delbrueckii spp. bulgaricus, Streptococcus thermophilus and L. rhamnosus and the acidified milk was prepared using d-(+)-glucono-δ-lactone (2%). Details of the method were described by Burton et al. [21]. The acidified milk mimicked physical characteristics, pH and texture of yoghurt [22]. The dairy product was consumed once (800 g) for 15 min

(https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=gse98645). The postprandial assay was completed after an overnight fast. Venous blood sampling was completed in the fasting state and during the 6-h postprandial time following dairy intakes [21]. Then, RNA was extracted using Paxgene whole-blood samples and sequenced using Illumina HiSeq platform.

2.2. Pre-assessment analysis

Gene expression profiles of the samples linked to the consumed yoghurt and acidified milk were compared with those of control individuals using GEO2R program. Moderated t-statistic method was used in GEO2R analysis. Distribution of data was assessed via a box plot to investigate equivalence characteristics of the samples. Significant up and down-regulated DEGs were visualized using volcano plot. Significant DEGs were identified and cleaned (uncharacterized genes were removed) based on an adjusted p-value of less than 0.05.

2.3. Protein-protein interaction network analysis

Significant DEGs were included in CluePedia v.1.5.7 of Cytoscape software v.3.7.2 to form a directed PPI network to find the critical genes. Nodes were connected by the activation, inhibition, expression, reaction, catalysis and post-translation modification actions. Undirected PPI network was used to find binding actions between the recognized significant DEGs. Significant DEGs were included in the “protein query” of the STRING database to construct a PPI network using Cytoscape software. A confidence score of 0.1 was used to maximize connections between the nodes. The network was analyzed via the “Network analyzer” plugin of Cytoscape software. Element of the major connected component was visualized and sorted through the degree values. Associated biological terms were assessed via gene ontology enrichment. Significant DEGs were enriched using ClueGO v.1.5.7 plugin of Cytoscape software. Biological terms were extracted from GO_BiologicalProcess-EBI-UniProt-GOA-ACAP-ARAP_08.05.2020_00h00, GOCellularComponent-EBI-UniProt-GOA-ACAP-ARAP_08.05.2020_00h00,EACTOME_Reactions_08.05.2020 ontology sources. Clustering of biological terms was carried out using default kappa score. Corrected statistical p-values were generated via Bonferroni step-down correction method.

2.4. Statistical analysis

Significant DEGs were identified based on adjusted p-values of less than 0.05. Undirected PPI network was constructed regarding confidence scores of 0.1. Biological terms were achieved based on the corrected p-values via Bonferroni step-down correction method.

Enrichment/depletion (two-sided hypergeometric test) statistical test was used as well. A network specificity less than detailed was addressed.

  1. Results and Discussion

3.1. Pre-assessment analysis

A box plot of compared whole blood gene expression profiles of the yoghurt group (seven samples) with control group is present in Figure 1. Samples were median-centric and comparable statistically. A linked volcano plot of yoghurt-control analysis is shown in Figure 2. Significant up and down-regulated DEGs are visualized in Figure 2. The box plot of compared seven whole-blood gene expression profiles of the acidified milk group with those of the control groups is illustrated in Figure 3. Samples were matched statistically. Volcano plot of the acidified milk-control analysis is shown in Figure 4. As shown in the figure, no significant DEGs were detected. This analysis was ignored for further assessments. Assessment showed 37 significant DEGs (adjusted p-value < 0.05) from separating the yoghurt group from the controls. A list of the significant DEGs is present in Table 1.

3.2. Protein-protein interaction network analysis

Significant DEGs were assessed via a directed PPI network to find prominent relationships between DEGs. The PPI network included two subnetworks and the isolated nodes. The two subnetworks of the directed PPI network of yoghurt–control analysis including the significant DEGs are shown in Figure 5. Activation, inhibition, expression, reaction, catalysis and post-translation modification actions were used to form the network. In general, activation, reaction, catalysis and post-translation modification actions included relationships between nodes of the identified subnetworks. The constructed undirected PPI network of yoghurt-control analysis is mapped in Figure 6. The 22 nodes of the major connected component of the PPI network were connected by 51 edges. Isolated and paired nodes are shown in Figure 6. However, undirected PPI network was small and was not a scale-free network, nodes were layout based on degree values to show various centrality characteristics of the nodes.

3.3. Gene ontology enrichment

Gene ontology results for yoghurt–control analysis are present in Figure 7. A total number of 54 biological terms of biological processes, cellular components and reactions were identified as the linked biological terms. Biological terms were clustered in four groups [“formation of translation initiation complexes yielding circularized ceruloplasmin mRNA in a 'closed-loop' conformation”, “positive regulation of nuclear-transcribed mRNA poly(A) tail shortening”, “rDNA heterochromatin assembly” and “protein import into mitochondrial matrix”] were as the targeted terms. List of the biological terms is shown in Table 2.

Pre-assessment analyses, including box plots and volcano plots (Figures 1, 2), indicated that whole-blood gene expression profiles of the participants who consumed yoghurt were comparable with those of the controls. Significant DEGs were visualized in the volcano plot. Rundblad et al. reported effects of fermented dairy product intakes on gene expression responses of the peripheral blood mononuclear cells associated with less inflammation [23]. As shown in Figure 1, box plot of Figure 3 corres-ponds to a similar analysis. However, no significant DEGs were reported (Figure 4). Results indicated that yoghurt included significant effects on gene expression profiles of the whole bloods while acidified milk did not.

A list of 37 significant DEGs, including 14 upregulated and 13 downregulated genes responsible for consuming yoghurt, is present in Table 1. Importance of the biomarkers of food intake is highlighted in the literature

[24]. An investigation by Rezaei et al. showed that consumption of yoghurt led to beneficial effects on blood pressure, blood glucose, serum lipid and glycated hemoglobin. However, C-reactive protein level, high-density lipoprotein and cholesterol were not affected by yoghurt consumption [25].

To screen the 37 significant DEGs, actions between the studied genes were mapped (Figure 5). Technically, RPSA, RPS5, RPS14, PABPC1, DDX60L, FEN1, MRPL12, H4C11, H4C12 and KAT6A were addressed as the interacted DEGs. From the ten highlighted DEGs, H4C11 and H4C12 were not present in the PPI network while others were appeared as the central nodes (Figure 6). Gene ontology assessment is an appropriate tool to assess a set of genes [26]. Results of gene ontology enrichment of the 37 DEGs are illustrated in Figure 7. As depicted in Figure 7, two clusters of biological processes including “rDNA heterochromatin assembly” and “formation of translation initiation complexes yielding circularized ceruloplasmin mRNA in ‘closed-loop’ conformation” were associated to the RPSA, RPS5, RPS14, PABPC1, H4C11 and H4C12. It seems that “formation of translation initiation complexes yielding circularized ceruloplasmin mRNA in ‘closed-loop’ conformation” was the major targeted group of biological processes.

The most complex characteristics of translation in eukaryotes include the established initiation process. It needs at least  11 eukaryotic initiation factors with coordinated interactions [27]. There is a model that corresponds with the formation a closed-loop structure by mRNA in the initiation of protein synthesis in eukaryotic organisms [28]. More than 95% of plasma copper is associated with ceruloplasmin.

This serum ferroxidase belongs to the multicopper oxidase family, which needs copper for its function. Investigations indicate that malfunction of ceruloplasmin leads to aceruloplasminemia; a neurodegenerative disease [29]. As depicted in Table 2 and Figure 7, “formation of translation initiation complexes yielding circularized ceruloplasmin mRNA in ‘closed-loop’ conformation” includes 16 biological processes and is associated to RPSA, RPS5, RPS14, PABPC1 and DHX30 genes. As discussed previously, RPSA, RPS5, RPS14 and PABPC1 were highlighted as critical genes. Relationships between these four critical genes are shown in Figure 5. As present in Figure 5, RPS14 is affected directly and indirectly by RPS5, RPSA and PABPC1. Downregulation of RPSA, RPS5 and RPS14 is shown in Table 1.

It could be concluded that yoghurt consumption was accompanied by decreased yield of “formation of translation initiation complexes yielding circularized ceruloplasmin mRNA in ‘closed-loop’ conformation” biological processes group, meaning that blood ceruloplasmin level decreased in the yoghurt consumers. Studies demonstrate that ceruloplasmin is the most abundant protein in milk containing β-casein variant A1A1 [30]. Raia et al. reported effects of ceruloplasmin deficiency on the impairment of  brain iron metabolisms and behaviors in mice [31]. They showed that ceruloplasmin deficiency played roles in dysregulation of lipid metabolism in the liver and adipose tissues of mice [32]. As shown in Figure 7, “formation of translation initiation complexes yielding circularized ceruloplasmin mRNA in a 'closed-loop' conformation” was connected to five associated DEGs; hence, it could be concluded that ceruloplasmin deficiency was a specific change in gene expression profiles of the yoghurt consumers. Benefits of this finding need further studies.

  1. Conclusion

In conclusion, yoghurt includes significant effects on gene expression of whole bloods while acidified milk does not. Totally, 37 significant DEGs were highlighted as a result of yoghurt consumption. Downregulation of genes that are associated with the production and function of ceruloplasmin was highlighted in the blood of yoghurt consumers. Further studies, including detailed experiments on ceruloplasmin in bloods of yoghurt consumers, validation of the introduced critical DEGs via the associated methods such as quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and larger size samples, are needed to investigate complete molecular metabolisms associated to yoghurt consumption.

  1. Ethical Code

This study was approved via IR.SBMU.RETECH.-REC.1403.260 ethical code

  1. Acknowledgements

This study was supported by Shahid Beheshti University of Medical Sciences.

  1. Conflict of Interest

Mostafa Rezaei-Tavirani contributed to the conception and design of the study and literature review. All authors participated equally in project administration and writing of the primary draft of the manuscript, providing critical revision and editing. All authors approved the final version of the manuscript.

References

  1. Liu SQ. Practical implications of lactate and pyruvate metabolism by lactic acid bacteria in food and beverage fermentations. Int J Food Microbiol.2003; 83(2): 115-31.

https://doi.org/10.1016/S0168-1605(02)00366-5

  1. Trimigno A, Bøge Lyndgaard C, Atladóttir GA, Aru V, Balling Engelsen S, Harder Clemmensen LK. An NMR metabolomics approach to investigate factors affecting the yoghurt fermentation process and quality. Metabolites. 2020; 10(7): 293.

https://doi.org/10.3390/metabo10070293

  1. Le Roy CI, Kurilshikov A, Leeming ER, Visconti A, Bowyer RC, Menni C, Falchi M, Koutnikova H, Veiga P, Zhernakova A, Derrien M, Spector TD. Yoghurt consumption is associat-ed with changes in the composition of the human gut micr-obiome and metabolome. BMC Microbiol. 2022; 22(1): 39.

https://doi.org/10.1186/s12866-021-02364-2

  1. Czaja TP, Vickovic D, Pedersen SJ, Hougaard AB, Ahrné L. Spectroscopic characterisation of acidified milk powders. Int Dairy J. 2023;142:105664.

https://doi.org/10.1016/j.idairyj.2023.105664

  1. Liu D, Yu Y, Zhang J, Liu X, Wang M, Hemar Y, M. Regenstein J, Zhou P. Biochemical and physico-chemical changes of skim milk during acidification with glucono-δ-lactone and hydrogen chloride. Food Hydrocoll. 2017; 66: 99-109.

https://doi.org/10.1016/j.foodhyd.2016.12.030

  1. Chen YC, Chen CC, Chen ST, Hsieh JF. Proteomic profiling of the coagulation of milk proteins induced by glucono-delta-lactone. Food Hydrocoll. 2016; 52: 137-143.

https://doi.org/10.1016/j.foodhyd.2015.06.005

  1. Mohamadshahi M, Veissi M, Haidari F, Shahbazian H, Kaydani G-A, Mohammadi F. Effects of probiotic yogurt consumption on inflammatory biomarkers in patients with type 2 diabetes. BioImpacts. 2014; 4(2): 83-88.

https://doi.org/10.5681%2Fbi.2014.007

  1. Nestel PJ, Mellett N, Pally S, Wong G, Barlow CK, Croft K, Trevor A M, Peter J M. Effects of low-fat or full-fat fermented and non-fermented dairy foods on selected cardiovascular biomarkers in overweight adults. Br J Nutr. 2013; 110(12): 2242-2249.

https://doi.org/10.1017/S0007114513001621

  1. Ejtahed HS, Mohtadi-Nia J, Homayouni-Rad A, Niafar M, Asghari-Jafarabadi M, Mofid V. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutr J. 2012; 28(5): 539-543.

https://doi.org/10.1016/j.nut.2011.08.013

  1. Savaiano DA, AbouElAnouar A, Smith DE, Levitt MD. Lactose malabsorption from yogurt, pasteurized yogurt, sweet acidophilus milk and cultured milk in lactase-deficient individuals. Am J Clin Nutr. 1984; 40(6): 1219-1223.

https://doi.org/10.1093/ajcn/40.6.1219

  1. Linder M C. Ceruloplasmin and other copper binding components of blood plasma and their functions: An update: Metallomics. 2016; 8(9): 887-905.

https://doi.org/10.1039/c6mt00103c

  1. Liu Z, Wang M, Zhang C, Zhou S, Ji G. Molecular functions of ceruloplasmin in metabolic disease pathology. Diabetes Obes Metab. 2022; 15: 695-711.

https://doi.org/10.2147/DMSO.S346648

  1. Giavarina D, Lippi G. Blood venous sample collection: Recommendations overview and a checklist to improve quality. Clin Chem. 2017; 50(10-11): 568-573.

https://doi.org/10.1016/j.clinbiochem.2017.02.021

  1. Chacon-Cortes D, Griffiths LR. Methods for extracting genomic DNA from whole blood samples: Current perspectives. J Biorepos Sci Appl Med. 2014; 2: 1-9

https://doi.org/10.2147/BSAM.S46573

  1. Petrov PD, Bonet ML, Reynes B, Oliver P, Palou A, Ribot J. Whole blood RNA as a source of transcript-based nutrition-and metabolic health-related biomarkers. PloS one. 2016; 11(5): e0155361.

https://doi.org/10.1371/journal.pone.0155361

  1. Pereira R, Oliveira J, Sousa M. Bioinformatics and computational tools for next-generation sequencing analysis in clinical genetics. J Clin Med. 2020; 9(1): 132.

https://doi.org/10.3390/jcm9010132

  1. Rezaei Tavirani M, Ahmadzadeh A, Rostami Nejad M. Herbal diet and the impact of nutrition on lipogenic activity, a system biology study. Res J Pharmacogn. 2023; 10(4): 43-50.

https://doi.org/10.22127/rjp.2023.405866.2155

  1. Rezaei Tavirani M, Arjmand B, Razzaghi M, Ahmadzadeh A. 50S Ribosomal proteins family is the main target of cinnamon extract: A network analysis. Res J Pharmacogn. 2021; 8(2): 63-68.

https://doi.org/10.22127/rjp.2021.265776.1659

  1. Rezaei Tavirani M, Farahani M, Razzaghi Z, Arjmand B, Khodadoost M. Introducing Coffee as a Complementary Agent Beside Metformin Against Type 2 Diabetes. Res J Pharmacogn. 2024; 11(3): 31-40.

https://doi.org/10.22127/rjp.2024.418824.2239

  1. Sagaya FM, Hurrell RF, Vergères G. Postprandial blood cell transcriptomics in response to the ingestion of dairy products by healthy individuals. J Nutr Biochem. 2012; 23(12): 1701-1715.

https://doi.org/10.1016/j.jnutbio.2012.01.001

  1. Burton KJ, Pimentel G, Zangger N, Vionnet N, Drai J, McTernan PG, Pralong FP, Delorenzi M, Vergères G. Modulation of the peripheral blood transcriptome by the ingestion of probiotic yoghurt and acidified milk in healthy, young men. PLoS One. 2018; 13(2): e0192947.

https://doi.org/10.1371/journal.pone.0192947

  1. Burton KJ, Rosikiewicz M, Pimentel G, Bütikofer U, Von Ah U, Voirol M-J, Greub G, Pralong FP, Vergères G, Vionnet N, Croxatto. A, Aeby S, Drai J, McTernan Ph. Probiotic yogurt and acidified milk similarly reduce postprandial inflammation and both alter the gut microbiota of healthy, young men. Br J Nutr. 2017; 117(9): 1312-1322.

https://doi.org/10.1017/S0007114517000885

  1. Rundblad A, Holven KB, Øyri LK, Hansson P, Ivan IH, Gjevestad GO, Thoresen M, Ulven SM. Intake of fermented dairy products induces a less pro‐inflammatory postprandial peripheral blood mononuclear cell gene expression response than non‐fermented dairy products: A randomized controlled cross‐over trial. Mol Nutr Food Res. 2020; 64(21): e2000319.

https://doi.org/10.1002/mnfr.202000319

  1. Münger LH, Garcia-Aloy M, Vázquez-Fresno R, Gille D, Rosana ARR, Passerini A, Soria-Florido M-T , Pimentel G, Sajed T, Wishart DS, Lacueva CA, Vergères G, Praticò G. Biomarker of food intake for assessing the consumption of dairy and egg products. Genes Nutr. 2018; 13: 1-18.

https://doi.org/10.1186/s12263-018-0615-5

  1. Rezaei M, Sanagoo A, Jouybari L, Behnampoo N, Kavosi A. The effect of probiotic yogurt on blood glucose and cardiovascular biomarkers in patients with type II diabetes: A randomized controlled trial. Evid Based Care. 2017; 6(4): 26-35.

https://doi.org/10.22038/ebcj.2016.7984

  1. Razzaghi Z, Arjmand B, Hamzeloo-Moghadam M, Tavirani MR. Gene ontology assessment of indirect cold physical plasma and UV-radiation molecular mechanism at the cellular level. J Lasers Med Sci. 2023; 14: e10

https://doi.org/10.34172/jlms.2023.10

  1. Pisarev AV, Unbehaun A, Hellen CU, Pestova TV. Assembly and analysis of eukaryotic translation initiation complexes. Method Enzymol. 2007; 430: 147-77.

https://doi.org/10.1016/S0076-6879(07)30007-4

  1. Denis CL, Laue TM, Wang X. Identification of a 57S translation complex containing closed-loop factors and the 60S ribosome subunit. Sci Rep. 2018; 8(1): 11468.

https://doi.org/10.1038/s41598-018-29832-6

  1. Hellman NE, Gitlin JD. Ceruloplasmin metabolism and function. Annu Rev Nutr. 2002; 22(1): 439-458.

https://doi.org/10.1146/annurev.nutr.22.012502.114457

  1. Wang X, Yu Z, Zhao X, Han R, Huang D, Yang Y, et al. Comparative proteomic characterization of bovine milk containing β‐casein variants A1A1 and A2A2 and their hetero-zygote A1A2. J Sci Food Agric. 2021; 101(2): 718-725.

https://doi.org/10.1002/jsfa.10684

  1. Niu L, Zhou Y, Lu L, Su A, Guo X. Ceruloplasmin deficiency impaired brain iron metabolism and behavior in mice. Cell Biochem Biophys . 2022; 80(2): 385-393.

https://doi.org/10.1007/s12013-022-01061-9

  1. Raia S, Conti A, Zanardi A, Ferrini B, Scotti GM, Gilberti E, De Palma G, David S, Alessio M. Ceruloplasmin-deficient mice show dysregulation of lipid metabolism in liver and adipose tissue reduced by a protein replacement. Int J Mol Sci. 2023; 24(2): 1150.

https://doi.org/10.3390/ijms24021150.



The Effect of Sodium Benzoate and Nisin on Human HepG2 Cell Gene Expression

Fatemeh Bandarian, Farideh Razi, Zahra Razzaghi, Mohammad Rostami Nejad, Babak Arjmand, Mostafa Rezaei-Tavirani

Applied Food Biotechnology, Vol. 11 No. 2 (2024), 26 Aban 2024, Page e3
https://doi.org/10.22037/afb.v11i2.45959

Background and Objective: Sodium benzoate is known as a preservative compound with a high safety profile in the food industrial and pharmaceutical field due to its antibacterial and antifungal properties. In the other hand, nisin is a bio-preservative agent. Determining the effect of sodium benzoate on human HepG2 cell gene expression in comparison with nisin is the aim of this study.

Material and Methods: The effect of sodium benzoate on gene expression of human HepG2 cells was extracted from the Gene Expression Omnibus (GEO) database.  Pre-evaluation analysis via GEO2R confirmed valid analysis. The significant differentially expressed genes (DEGs) were investigated via protein-protein interaction (PPI) network analysis and the hubs were screened via a directed regulatory network. The critical hub genes were identified and discussed. The apoptosis-related dysregulated genes of human HepG2 from the literature were assessed among the significant DEGs of sodium benzoate analysis.

Results and Conclusion: A total number of 11521 significant DEGs were identified. The PPI network including 4095 recognized DEGs with 208 hub nodes was created. The hubs were assessed via a directed protein network and MAPK1, CCND1, MAPK14, RAF1, KRAS, MAPK3, PIK3CA, SIRT1, EGF, RBX1, FYN, and NIP7 were pointed out as the critical hub genes in response to presence of sodium benzoate. A total of 78 dysregulated genes of nisin analysis (except TNRSF25) had no common genes with significant DEGs of sodium benzoate evaluation. It can be concluded that sodium benzoate and nisin affect essential cellular functions such as cell cycle progression, cell motility and metabolism, cell proliferation, and survival in various manners. It seems wide usage of food preservative agents requires more investigation to guarantee human health maintenance.

Conflict of interest: The authors declare no conflict of interest.

 

  1. Introduction

 

Sodium benzoate is known as a preservative compound with a high safety profile in the food industry [1]. It is used widely as an antibacterial and antifungal preservative in food and pharmaceutical fields.  It has seen some tender of sodium benzoate as a therapeutic compound to treat medical disorders [2, 3]. Benzoate teratogenic and neurotoxic effects on zebrafish, chromosome abnormalities in cultured human lymphocytes, the reaction of benzoate with ascorbic acid to yield carcinogen benzene in drinks, and the effect on neurotransmission and cognitive functioning are the reported disadvantages of benzoate function [4]. Appl-ication of sodium benzoate is limited to 0 – 5 mg/kg by the World Health Organization and Food and Agriculture Organization of the United Nations [5]. Investigation indic-ates that sodium benzoate affects gene expression in the liver. Based on this study various doses of sodium benzoate silenced MAPK8 expression [6].

The growing request for fresh-like foodstuffs regarding the potential health hazards of chemical food preservative compounds led to the rise of food biotechnology features of biological antimicrobial reagents as food preservative tools. Bacteriocins are alternative food preservative substances that are small polypeptide molecules with antimicrobial activity [7]. Nisin is an antimicrobial peptide that was originally detected in milk fermentation culture. It is suggested that bacterial blockage in human foods is a safe agent [8-10]. Nisin as a natural food preservative compound is used in some food products versus many Gram-positive microorganisms. It can be applied alone or with other food preservative compounds [11].

Gene expression analysis appears as a suitable tool to explore the molecular mechanism of diseases and the function of drugs and chemical compounds [12]. Since gene expression analysis products are a set of significant up and downregulated DEGs, bioinformatics is a useful method to evaluate the findings [13]. PPI network analysis as a bioinformatics approach has attracted the attention of researchers to analyze genomic data [14]. The nodes of a PPI network are connected via the edges to form an interactome. In the scale-free PPI network, there are central nodes that are known as hubs. The hub nodes are characterized by a high value of degree. The degree is the number of the first neighbors of a node. It is reported that the hub nodes of a PPI network play critical roles in the function of the studied sample [15, 16]. The directed PPI networks are constructed from nodes that are connected by the directed edges from the causal genes to the affected
genes [17]. The activation, inhibition, and expression actions are used in the directed PPI network to explore the critical genes of the studied network [18].

In the present study, gene expression profiles of human HepG2 cells in the presence of sodium benzoate is analyzed via directed PPI network to find the critical affected genes in response to the applied chemical preservative compound. The dysregulated genes of cells in the presence of nisin as a bio-preservative agent are investigated in the literature. Comparison of gene expression alterations of human HepG2 cells in the presence of sodium benzoate and nisin is the main aim of this project.

 

  1. Materials and Methods

2.1. Data collection

The effect of sodium benzoate on gene expression of human HepG2 cells was searched in the GEO database. Information about four samples of treated cells with 10 mM sodium benzoate for 24 hours before harvesting relative to the four individual controls was found in GSE108469 (GPL16791) (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE108469). Data are retrieved via library strategy; RNA-Seq, library source; transcriptome, library selection; cDNA, and instrument model; Illumina HiSeq 2500. The effect of nisin on gene expression of human HepG2 cells was extracted from published documents of Zainodini N et al [10].

2.2. Pre-evaluation analysis

Data was assessed via the GEO2R program to find the significant DEGs. A Uniform Manifold Approximation and Projection (UMAP) plot was applied to detect the separation of treated cells from the control individuals. Significant up and downregulated DEGs were visualized via volcano plot. A number of significant DEGs relative to the unsignificant DEGs were shown via the Venn diagram.

2.3. PPI network analysis

The significant DEGs were investigated via undirected PPI network analysis.  The queried significant DEGs were included in the STRING database via “protein query” and the recognized genes were included in the PPI network by Cytoscape software v 3.7.2. The network was analyzed by the “Network analyzer” application of Cytoscape software to find the central nodes. The hubs as central nodes were selected based on degree cutoff = mean + 2(standard deviation). To explore critical genes, hub nodes were assessed via directed PPI network analysis. The hubs were connected by activation, inhibition, and expression links. Since the out-degree value is corresponded to the actor role of a node, the nodes of the network were arranged based on the out-degree value to find the controller DEGs.

2.4. Comparative study

 The dysregulated genes of the human HepG2 cells in the presence of nisin were investigated among the reported DEGs of sodium benzoate analysis.

2.5. Statistical analysis

The significant DEGs were determined based on adjusted P-value < 0.05. The PPI network was constructed based on a confidence score = 0.4. Hubs were identified via degree value cutoff = mean + 2(standard deviation).

 

 

  1. Results and Discussion

3.1. Pre-evaluation analysis

Data was assessed to find the critical significant DEGs that discriminate the treated cells with sodium benzoate from controls. UMAP plot is shown in Figure 1, the treated cells are completely separated from controls. UMAP is applied to reduce dimensions of data to 2-dimensional presentation and the amounts of two axes are not meaningful. The volcano plot (Figure 2) indicates that there are huge numbers of significant DEGs. As depicted in Figure 3, there are 11521 significant DEGs including up and downregulated genes.

3.2. PPI network analysis

The significant DEGs were included in an undirected PPI network; the interactome was formed from 4095 nodes and 63934 edges. The 208 hub nodes were determined based on degree value cutoff = 101. The hub nodes were inserted in a directed PPI network including 95 isolated genes, 2 paired DEGs, a subnetwork of 13 genes, and a main connected component of 98 nodes. The subnetwork of 13 genes and the main connected components that are connected via activation, inhibition, and expression relationships are shown in Figure 4. For better resolution, the directed PPI network including activation and expression, activation, expression, and inhibition are presented in Figures 5-8 respectively. To find a critical controller hub, the directed PPI network including a subnetwork of 13 genes, and a main connected component of 98 nodes was analyzed and arranged based on out-degree value (see Figure 9).

3.3 Nisin-related gene expression analysis

The up and down-regulated genes in the presence of nisin which are extracted from the document of Zainodini N et al [10] are shown in Table 1. As depicted in Table 1, 78 dysregulate gene are appeared as targeted genes by nisin. It should be mentioned that the dysregulated genes were identified based on the human apoptosis PCR array to find genes activated by nisin during possible apoptosis. This method has limited the presented data to apoptosis.

One of the attractive tools in biomedical investigation is transcriptomic analysis. UMAP as a non-linear method can efficiently group assorted samples in single-cell RNA sequencing analysis [19]. The clustering of the treated samples with sodium benzoate from the control cells is depicted in Figure 1. This finding corresponds to differences between gene expression profiles of the treated cells and control individuals. The other visualization method is a volcano plot which shows unstandardized signals such as log-fold-change versus noise-adjusted/standardized signals as like -log10(p-value) from the t-test. Volcano plot provides a double filtering tool to display screened data [20). Volcano plot presentation of data in Figure 2 displays a large number of significant up and downregulated DEGs (11521 DEGs (see Figure 3) in response to the presence of sodium benzoate in cell culture media.

PPI network analysis is an attractive tool to assess large numbers of genes or proteins to explore critical individuals. For instance, the down-regulation of ALB and upregulation of JUN, FOS, and MYC as critical molecular events in response to okadaic acid by human intestinal caco-2 cells is detected via PPI network analysis [21]. In the present study, 208 hub DEGs were identified as the central genes associated to the effect of sodium benzoate on the treated cells. Gene co-expression networks are a widely useful approach to detect the system-level functionality of genes. The nodes in gene co-expression networks are connected via significantly co-expressed relationships between them. Gene activation and inhibition are highlighted to interpret gene regulatory networks [22, 23].

Activation, inhibition, and expression relationship between the introduced hubs are shown in Figure 4. Complex features of the action map (see Figure 4), required the presentation of each action in a related simple map (see Figures 5-8).  As depicted in Figures 4 and 5, activation is a prominent action between the nodes of the network and NIP7 appears as a regulatory (activator) gene. Activation connections between nodes of network are so complex (see Figure 6) and understanding of relationships between the studied DEGs requires precious analysis. As shown in Figure 7, expression action appeared as the simplest network and included a minimum number of nodes.

One of the well-organized neighborhood-based metrics is degree value which has been extensively used due to its simplicity and low computation complication. The directionality of edges is not considered between the nodes, this problem limits the usage of degree centrality in a directed network. In a directed network, the kind of relationship between the nodes has significant importance. Therefore out-degree and in-degree values are appeared as the major tools to analyze the nodes of network [24). The directed network of hub nodes (see Figure 4) was analyzed and the network was arranged based on out-degree value. The result is shown in Figure 9. As depicted in Figure 9, MAPK1, CCND1, MAPK14, RAF1, KRAS, MAPK3, PIK3CA, SIRT1, EGF, RBX1, FYN, and NIP7 were pointed out as the crucial DEGs. As shown in Figure 6, all crucial genes (100%) are presented in the 3 significant subnetworks of the activation map while MAPK1, CCND1, MAPK14, KRAS, MAPK3, SIRT1, and EGF (58%) are included in the main connected component of expression map (see Figure 7). As shown in Figure 8, except for SIRT1 and NIP7 other crucial DEGs (83%) are included in the inhibition map. It can be concluded that activation, inhibition, and expression are respectively the main actions between the nodes.

Nucleolar pre-rRNA processing protein NIP7 is a downregulated hub gene that is highlighted in the activation map. Investigation indicates that downregulation of NIP7 is associated with pre-rRNA processing, leading to an inequality of the 40S/60S subunit ratio and reduction of the 34S pre-rRNA concentration and an intensification of the 26S and 21S pre-rRNA concentrations. This finding corresponds with cell proliferation alteration in human cells [25). MAPK1, MAPK3, and MAPK14 belong to mitogen-activated protein kinase (MAPK) gene family, which are involved in regulation of cytokines and proteases expres-sion, cell adherence, cell cycle progression, cell motility and metabolism. The effect of MAPKs on cell proliferation, survival, differentiation, development, and apoptosis is investigated and reported [26, 27]. There is evidence that the downregulation of Raf-1 proto-oncogene, serine/threonine kinase (RAF1) and upregulation of cyclin D1 (CCND1) is accompanied by HepG2 cells proliferation and migration induction [28]. The downregulation of RAF1 and the upregulation of CCND1 in response to the presence of sodium benzoate in human HepG2 cells are pointed out in the present study.

Investigation indicates that overexpression of miR‑30c prevents proliferation, migration and invasion of prostate cancer cell lines. This effect of miR‑30c is demonstrated via the downregulation of KRAS proto-oncogene, GTPase (KRAS) protein by miR‑30c [29]. KRAS is downregulated in the presence of sodium benzoate. EGF is another upregulated gene in response to sodium benzoate effect on the treated cells. There is evidence that EGF induces tumor cell invasion [30]. Sirtuin-1 (SIRT1) is a downregulated hub, this gene is known as a protective element against the progression and development of non-alcoholic fatty liver disease [31). It is reported that phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) upregulation is associated with several tumors [32]. PIK3CA has been upregulated in the presence of sodium benzoate. Investigation revealed a significant role of ring-box 1 (RBX1) in the development of non-small cell lung cancer [33]. It appeared as a downregulated hub in the present study. FYN proto-oncogene, Src family tyrosine kinase (FYN) is the last downregulated gene, upregulation of FYN in thyroid carcinoma is reported by Zheng et al [34].

The apoptosis-related dysregulated genes in response to the presence of nisin are shown in Table 1. The assessment showed except for TNRSF25 there are no common genes between these dysregulated genes and the significant DEGs of sodium benzoate analysis. TNRSF25 is upregulated in the presence of sodium benzoate 2-fold while is downregulated in the presence of nisin 6.6-fold. Both up and down-regulation of TNRSF25 refer to the side effects of sodium benzoate and nisin effect as food preservative agents. Findings indicate that nisin activity against apoptosis is different from sodium benzoate. It seems nisin has a potent effect on cellular apoptosis relative to sodium benzoate.

  1. Conclusion

In conclusion, the basic functional features of human HepG2 cells are affected by sodium benzoate. Discrimination of the 40S/60S subunit ratio, decrease of the 34S pre-rRNA concentration, intensification of the 26S and 21S pre-rRNA concentrations, regulation of cytokines, some cancerous processes expression, cell adherence, cell cycle progression, cell motility and metabolism, cell proliferation, survival, differentiation, development, and apoptosis, tumor cell invasion, and progression and development of non-alcoholic fatty liver disease related pathways are the highlighted targets of sodium benzoate. The mentioned alterations include positive and negative functional changes. However, the negative side effects are dominant relative to the benefits. Nisin analysis showed that many apoptotic-related genes were dysregulated in response to the presence of nisin. However, this effect was not similar to the response of cells to the presence of sodium benzoate. Based on findings, chemical and bio-preservative food additives can affect human cellular function in various manners which influence human health. Finally, it is difficult to introduce nisin as a safer food preservative compound relative to sodium benzoate.

  1. Acknowledgements

Shahid Beheshti University of Medical Sciences supported this research.

  1. Conflict of Interest

The authors report no conflicts of interest.

  1. Author contributions

Mostafa Rezaei-Tavirani is contributed to the conception and design of the work and literature review. All authors participated equally in project administration and writing of the first draft of manuscripts, providing critical revision and editing. All authors approved the final version of the manuscript.

  1. Ethical Code

This project is approved via IR.SBMU.RETECH.REC.1403.105 Ethical code.

References

  1. Walczak-Nowicka ŁJ, Herbet M. Sodium benzoate harm-fulness and potential use in therapies for disorders related to the nervous system: A review. Nutrients. 2022; 14(7): 1497.

https://doi.org/10.3390/nu14071497

  1. Khan IS, Dar KB, Ganie SA, Ali MN. Toxicological impact of sodium benzoate on inflammatory cytokines, oxidative stress and biochemical markers in male Wistar rats. Drug Chem Toxicol. 2022; 45(3): 1345-1354.

https://doi.org/10.1080/01480545.2020.1825472

  1. Olofinnade AT, Onaolapo AY, Onaolapo OJ, Olowe OA. The potential toxicity of food-added sodium benzoate in mice is concentration-dependent. Toxicol Res (Camb). 2021; 10(3): 561-569.

https://doi.org/10.1093/toxres/tfab024

  1. Piper JD, Piper PW. Benzoate and sorbate salts: a systematic review of the potential hazards of these invaluable preservatives and the expanding spectrum of clinical uses for sodium benzoate. Compr Rev Food Sci Food Saf. 2017; 16(5): 868-880.

https://doi.org/10.1111/1541-4337.12284

  1. Shahmohammadi M, Javadi M, Nassiri-Asl M. An overview on the effects of sodium benzoate as a preservative in food products. Biotech Health Sci. 2016; 3(3): 7-11.

https://doi.org/10.17795/bhs-35084

  1. Raposa B, Pónusz R, Gerencsér G, Budán F, Gyöngyi Z, Tibold A, Hegyi D, Kiss I, Koller Á, Varjas T. Food additives: Sodium benzoate, potassium sorbate, azorubine, and tartrazine modify the expression of NFκB, GADD45α, and MAPK8 genes. Physiol Int. 2016; 103(3): 334-343.

 https://doi.org/10.1556/2060.103.2016.3.6

  1. Johnson EM, Jung DY-G, Jin DY-Y, Jayabalan DR, Yang DSH, Suh JW. Bacteriocins as food preservatives: Challenges and emerging horizons. Crit Rev Food Sci Nutr. 2018; 58(16): 2743-2767.

https://doi.org/10.1080/10408398.2017.1340870

  1. Shin JM, Gwak JW, Kamarajan P, Fenno JC, Rickard AH, Kapila YL. Biomedical applications of nisin. J Appl Microbiol. 2016; 120(6): 1449-1465.

https://doi.org/10.1111/jam.13033

  1. Rai M, Pandit R, Gaikwad S, Kövics G. Antimicrobial peptides as natural bio-preservative to enhance the shelf-life of food. J Food Sci Technol. 2016; 53: 3381-94.

https://doi.org/10.1007/s13197-016-2318-5

  1. Zainodini N, Hajizadeh MR, Mirzaei MR. Evaluation of apoptotic gene expression in hepatoma cell line (HepG2) following nisin treatment. Asian Pac J Cancer Prev. 2021; 22(5): 1413.

https://doi.org/10.31557/APJCP.2021.22.5.1413

  1. Gharsallaoui A, Oulahal N, Joly C, Degraeve P. Nisin as a food preservative: part 1: physicochemical properties, antimicrobial activity, and main uses. Crit Rev Food Sci Nutr. 2016; 56(8): 1262-1274.

https://doi.org/10.1080/10408398.2013.763765

  1. Shishodia S. Molecular mechanisms of curcumin action: gene expression. Biofactors. 2013; 39(1): 37-55.

https://doi.org/10.1002/biof.1041

  1. Zhang J, Wang X, Xu L, Zhang Z, Wang F, Tang X. Investigation of potential genetic biomarkers and molecular mechanism of ulcerative colitis utilizing bioinformatics analysis. Biomed Res Int. 2020; 2020(1): 4921387.

https://doi.org/10.1155/2020/4921387

  1. Khan MM, Mohsen MT, Malik MZ, Bagabir SA, Alkhanani MF, Haque S, Serajuddin M, Bharadwaj M. Identification of potential key genes in prostate cancer with gene expression, pivotal pathways and regulatory networks analysis using integrated bioinformatics methods. Genes. 2022; 13(4): 655.

https://doi.org/10.3390/genes13040655

  1. Rezaei-Tavirani M, Rezaei-Tavirani S, Mansouri V, Rostami-Nejad M, Rezaei-Tavirani M. Protein-protein interaction network analysis for a biomarker panel related to human esophageal adenocarcinoma. Asian Pac J Cancer Prev. 2017; 18(12): 3357-3363.

https://doi.org/10.22034/APJCP.2017.18.12.3357

  1. Safari-Alighiarloo N, Taghizadeh M, Rezaei-Tavirani M, Goliaei B, Peyvandi AA. Protein-protein interaction networks (PPI) and complex diseases. Gastroenterol Hepatol Bed Bench. 2014;7(1):17-31.
  2. Silverbush D, Sharan R. A systematic approach to orient the human protein–protein interaction network. Nat Commun. 2019; 10(1): 3015.

https://doi.org/10.1038/s41467-019-10887-6

  1. Mansouri V, Arjmand B, Hamzeloo-Moghadam M, Razzaghi Z, Khodadoost M, Rezaei Tavirani M, Rezaei Tavirani M, Ahmadzadeh A. Introducing BDNF and SNAI1 as the Crucial Targeted Genes by UV Radiation. J Lasers Med Sci. 2022; 13: e76-e.

https://doi.org/10.34172/jlms.2022.76

  1. Yang Y, Sun H, Zhang Y, Zhang T, Gong J, Wei Y, Duan YG, Shu M, Yang Y, Wu D, Yu D. Dimensionality reduction by UMAP reinforces sample heterogeneity analysis in bulk transcriptomic data. Cell Rep. 2021; 36(4).

https://doi.org/10.1016/j.celrep.2021.109442

  1. Li W. Volcano plots in analyzing differential expressions with mRNA microarrays. J Bioinform Comput Biol. 2012; 10(06): 1231003.

https://doi.org/10.1142/S0219720012310038

  1. Robati RM, Razzaghi Z, Arjmand B, Tavirani MR, Nejad MR, Rezaei M, Zamanian Azodi M. The Maim Targets of Okadaic Acid Toxin in Human Intestinal Caco-2 Cells: An Investigation of Biological Systems. Int J Med Toxicol Forensic Med.2023: 13(4): E42997.

https://doi.org/10.32598/ijmtfm.v13i4.42997

  1. Mercatelli D, Scalambra L, Triboli L, Ray F, Giorgi FM. Gene regulatory network inference resources: A practical overview. Biochim Biophys Acta Gene Regul Mech. 2020; 1863(6): 194430.

https://doi.org/10.1016/j.bbagrm.2019.194430

  1. Pratapa A, Jalihal AP, Law JN, Bharadwaj A, Murali T. Benchmarking algorithms for gene regulatory network inference from single-cell transcriptomic data. Nat Methods. 2020; 17(2): 147-154.

https://doi.org/10.1038/s41592-019-0690-6

  1. Jia P, Liu J, Huang C, Liu L, Xu C. An improvement method for degree and its extending centralities in directed networks. Physica A Stat. 2019; 532: 121891.

https://doi.org/10.1016/j.physa.2019.121891

  1. Morello LG, Hesling C, Coltri PP, Castilho BA, Rimokh R, Zanchin NI. The NIP7 protein is required for accurate pre-rRNA processing in human cells. Nucleic Acids Res. 2011; 39(2): 648-665.

https://doi.org/10.1093/nar/gkq758

  1. Ronkina N, Gaestel M. MAPK-activated protein kinases: servant or partner? Annu Rev Biochem. 2022;91(1): 505-540.

https://doi.org/10.1146/annurev-biochem-081720-114505

  1. Tsvetankova R, Tsvetkova I, Hayrabedyan S, Todorova K. Restoring mitophagy in prostate cancer cells: the role of miR-141 rescue in counteracting MAPK1/ERK2-dependent autophagy suppression. Biotech Biotechnol Equip. 2023; 37(1): 2293055.

https://doi.org/10.1080/13102818.2023.2293055

  1. Xie H, Jing R, Liao X, Chen H, Xie X, Dai H, Pan L. Arecoline promotes proliferation and migration of human HepG2 cells through activation of the PI3K/AKT/mTOR pathway. Hereditas. 2022; 159(1): 29.

https://doi.org/10.1186/s41065-022-00241-0

  1. Zhang J, Wang X, Wang Y, Peng R, Lin Z, Wang Y, Hu B, Wang J, Shi G. Low expression of microRNA‑30c promotes prostate cancer cells invasion involved in downregulation of KRAS protein. Oncol Lett. 2017; 14(1): 363-368.

https://doi.org/10.3892/ol.2017.6163

  1. Huang K, Gao N, Bian D, Zhai Q, Yang P, Li M, Wang X. Correlation between FAK and EGF‐Induced EMT in Colorectal Cancer Cells. J Oncol. 2020; 2020(1): 5428920.

https://doi.org/10.1155/2020/5428920

  1. Niu B, He K, Li P, Gong J, Zhu X, Ye S, Ou Z, Ren G. SIRT1 upregulation protects against liver injury induced by a HFD through inhibiting CD36 and the NF‑κB pathway in mouse kupffer cells. Mol Med Rep. 2018; 18(2): 1609-1615.

https://doi.org/10.3892/mmr.2018.9088

  1. Jin X, Wang D, Lei M, Guo Y, Cui Y, Chen F, Sun W, Chen X. TPI1 activates the PI3K/AKT/mTOR signaling pathway to induce breast cancer progression by stabilizing CDCA5. J Transl Med. 2022; 20(1): 191.

https://doi.org/10.1186/s12967-022-03370-2

  1. Xing R, Chen K-B, Xuan Y, Feng C, Xue M, Zeng Y-C. RBX1 expression is an unfavorable prognostic factor in patients with non-small cell lung cancer. Surg Oncol. 2016; 25(3): 147-151.

https://doi.org/10.1016/j.suronc.2016.05.006

  1. Zheng J, Li H, Xu D, Zhu H. Upregulation of tyrosine kinase FYN in human thyroid carcinoma: role in modulating tumor cell proliferation, invasion, and migration. Cancer Biother Radiopharm. 2017; 32(9): 320-326.

https://doi.org/10.1089/cbr.2017.2218



Detection of Aflatoxin Contamination through Biomarker Discovery in Human Intestinal Caco-2 Cells

Maryam Hamzehloo-Moghadam, Nastaran Asri, Somayeh Jahani Sherafat, Vahid Mansouri

Applied Food Biotechnology, Vol. 11 No. 2 (2024), 26 Aban 2024, Page e4
https://doi.org/10.22037/afb.v11i2.46449

Background and Objective: Aflatoxin includes four toxin types of B1, B2, G1 and G2. Due to the widespread contamination of food and agricultural products and the harmful effects of aflatoxin on the human body, investigating biological effects of aflatoxin and preventing its harmful effects on human health are important for the experts. The present study aimed to find a complementary method to modify standard detection methods of harmful levels of aflatoxin based on biomarker discovery.

Material and Methods: Data regarding changes in the gene expression profile of human intestinal Caco-2 cells were retrieved from the gene expression omnibus database, specifically concerning effects of aflatoxin B1 (AB1) on treated cells. Data were assessed via directed protein-protein interaction network analysis and gene ontology enrichment to identify the critical differentially expressed genes and the associated biochemical pathways.

Results and Conclusion: From the 934 differentially expressed genes, 623 were investigated through protein-protein interaction network analysis. Two directed protein-protein interaction networks were constructed from the significantly upregulated and downregulated differen-tially expressed genes. The COL4A6, IRF1, SLC2A2, CITED2, SULT2A1, RRAS and PCYT1B emerged as critical target genes affected by AB1. Various cellular functions, including cell proliferation, migration, apoptosis and metabolism, were identified as the key biochemical pathways that were affected. It was concluded that the levels of these critical target genes could be addressed as appropriate criteria for modifying standard methods of aflatoxin contamination detection.

Conflict of interest: The authors declare no conflict of interest.

  1. Introduction

 

Aspergillus flavus (AF) is a fungus with saprotrophic and pathogenic characteristics [1]. Wheat grains, legumes and tree nuts are best known for AF colonization [2]. The AF is used in production of fermented foods [3]. Aflatoxin is a dangerous mycotoxin in the food industry produced by AF [4]. Aflatoxin is classified as a Group 1 carcinogen [5] with four types of B1, B2, G1 and G2. The AF produces carcinogenic aflatoxins B1 and B2 [6]. More than 80% of aflatoxin B1 (AB1) absorption occur in the proximal part of the gastrointestinal tract [GIT) via passive transport after ingestion [7]. The cytochrome P450 enzyme can metabolize AB1 in the liver [8]; However, AB1 metabolism to its toxic metabolites may occur in the digestive tract [9]. The intestinal epithelia serve as the first line of defense against contaminants, protecting against toxic components entering through thigh junction proteins and are primary targets for mycotoxins such as AB1 [10]. The human Caco-2 intestinal cell line expresses typical intestinal nutrient transporters and is a widely used in-vitro model for drug delivery and metabolism studies [11]. Genome profile affected by AB1 as a biological system is investigated [12]. The AB1 increases lactate dehydro-genase (LDH) cytosolic enzyme levels, promotes dose-dependent accumulation of reactive oxygen species (ROS) and induces DNA damages in vitro [13]. The cytochrome P450 enzyme plays a significant role in bioactivation of AB1 to AB1-8,9-epoxide, indicating a high level of oxidative stress in intestinal cells in vitro [14].  There is a significant diversity in aflatoxin metabolism in various species [15]. Metabolism of AB1 in humans varies across regions and is affected by age, which affects individual resistance to AB1 as well [14]. Genome instability and cancer risk due to A-T mutations resulting in diminished DNA repair capability have been discussed by Engin et al. [16]. The AB1 exposure is associated with synergetic point mutations in the human P53 gene [17]. Diversity of metabolic pathways and various biomarkers involved in AB1 toxicity necessitate finding appropriate biomarkers for early detection of intestine cancers associated to AB1 exposure. Use of genomic data banks and network analysis software provides powerful tools to analyze data and identify hub genes associated with aflatoxin carcinogen characteristics [18]. Since effects of aflatoxin on gene profiles are not well-investigated in colon cancer, this study aimed to assess effects of AB1 on genes of Caco2 cells using information from valid data banks to carry out genes network analysis, introducing key biomarkers affected by AB1. This advancement holds significant implications for improving food safety protocols and early detection strategies of aflatoxin-associated health risks.

  1. Materials and Methods

2.1. Data collection

In this study, GSE75934 was investigated using toxicity, aflatoxin B1 and human intestinal keywords in gene expression omnibus (GEO) database

(https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=gse75934). The gene expression profiles of five sets of cells; control and treated cells with PATULIN, ASCLADIOL, AB1, VERSICOLORIN A and STRIGMATOCYSTIN. Gene expression profiles of the treated cells with AB1 toxin and control cells were selected for analysis. Total RNA from the studied cells was extracted using EXTRACT-ALL® reagent (Eurobio, Les Ulis, France) following the manufacturer’s manual. As described in the recorded data, for each sample, cyanine-3 (Cy3) labeled cRNA was prepared from 200 ng of total RNA using the One-Color Quick Amp Labeling kit (Agilent, USA) according to the manufacturer's instructions, followed by Agencourt RNAClean XP (Agencourt Bioscience Corpor-ation, Beverly, Massachusetts). Dye incorporation and cRNA yield were checked with the Biospec nano spectro-photometer (Shimadzu). More details of experiments are present in the following address.

(https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSM1970394).

2.2. Pre-assessment analysis

Human intestinal Caco-2 cells were exposed in vitro to 1 µM of AB1 for 24 h. Then, RNA was extracted and compared with that of the control cells (cells in presence of DMSO). Details of the methods were described in a published document by Gauthier et al. [19]. The GEO2R analysis was carried out with default options to find significant differentially expressed genes (DEGs). The GEO2R program was used to assess comparability of the samples via box plot visualization and volcano plot prese-ntation of the dysregulated genes. Significant DEGs were highlighted based on adjusted p-value. Samples matching was carried out and visualized using box plot analysis and GEO2R program. Presence of significant DEGs was assessed using volcano plot. Significant DEGs were addressed based on adjusted p-values less than 0.05. Repeated and uncharacterized DEGs were excluded from further analysis.

2.3. Gene ontology enrichment and directed protein-protein interaction network analysis

Significant up and downregulated DEGs were enriched separately to find the associated biochemical pathways using ClueGO plugin of Cytoscape software v.3.7.2. Biochemical pathways were extracted from the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database. Biological terms were identified based on the kappa score value. Significant DEGs were assessed through directed protein-protein interaction (PPI) network analysis to identify critical genes. Queried genes were connected via activation, inhibition and expression links using CluePedia application of Cytoscape software. The common DEGs identified through pathway analysis and directed PPI network analysis were selected as the critical targeted genes affected by AB1.

2.4. Statistical analysis

Comparability of samples was verified via median-centered samples in box plot analysis. Significant DEGs were identified based on adjusted p-values less than 0.05. The statistical options included enrichment/depletion, two-sided hypergeometric test, Bonferroni step down and p-value correction. Network specificities greater than the medium threshold were addressed when investigating associated biochemical pathways.

 

  1. Results and Discussion

3.1. Pre-assessment analysis

Gene expression profiles of the treated cells exposed to AB1 and the control cells were statistically matched (Figure 1). The compared profiles were median-centered and comparable. As shown in Figure 2, volcano plot visualization of the analysis showed several significant up and down-regulated DEGs. Totally, 934 significant DEGs, distinguishing treated cells from the controls, were identified. Data were cleaned and 623 DEGs, including 294 upregulated and 329 downregulated DEGs, were selected for further investigations.

3.2. Gene ontology enrichment

Results of the gene ontology assessment are shown in Figures 3 and 4. As shown in Figure 3, 15 groups of biochemical pathways were associated to 43 upregulated DEGs. Network of the associated DEGs and biochemical pathways included eight subnetworks. The five down-regulated biochemical pathways were present together with ten associated genes (Figure 4). The sum of the 43 upregulated genes and the ten downregulated DEGs connected to the biochemical pathways were addressed as critical targeted genes by AB1 toxin. Significantly, analysis of Figures 3 and 4 indicated that the upregulation of genes was the dominant response to aflatoxin B1 exposure, highlighting a significant shift in gene express-ion profiles towards increased transcription in various biochemical pathways. As demonstrated in Figure 3, most biological terms were involved in cellular metabolism. Since downregulation was not a prominent process, no significant subnetwork was shown in Figure 4. Results of the directed PPI network are present in Figure 5. All 623 significant DEGs were included in the constructively directed PPI network. Totally, 569 DEGs were recognized using Cytoscape software. The PPI network, including 492 isolated genes, 16 paired DEGs, six triple subnetworks (18 nodes), one tetrad component, two subnetworks of seven DEGs (14 nodes) and a major connected component consisting of 25 nodes was created. Subnetworks, including three or fewer nodes, were not included in further analysis. The COL4A6, IRF1, SLC2A2, CITED2, SULT2A1, RRAS and PCYT1B were identified as common DEGs between the pathway analysis and the directed PPI network analysis (see Table 1). It is noteworthy that the log (fold change) values for all the identified common genes were positive, indicating that these genes were upregulated in response to aflatoxin B1 exposure (Table 1).

As demonstrated, seven genes including COL4A6, IRF1, SLC2A2, CITED2, SULT2A1, RRAS and PCYT1B, were highlighted in response to the presence of AB1 toxin in human intestinal Caco-2 cells. The following descriptions briefly review their physiological and pathological roles. Type IV collagen lattice network was accumulated from six chains coded by the COL4A1 to Col4A6 genes. These genes were major components of the basement membranes [20]. Investigation indicated that the COL4A6 gene was highly downregulated in prostate cancer and this down-regulation was accompanied by the cancer progression and metastasis [21]. Furthermore, it has been reported that Col4A6 is significantly downregulated in stomach adenocarcinoma tissues, compared to normal adjacent tissues [22]. As shown in Figure 5, COL4A6 was a part of the largest connected component of the directed PPI network. As shown in Figure 3, COL4A6 was directly associated to the biochemical pathway of amoebiasis, which is a leading cause of death from parasitic illness globally [23].

Interferon regulatory factor-1 (IRF1) is a transcription factor involved in the regulation of several genes associated to various pathological and physiological processes such as tumor immune surveillance, viral infection, development of the immune system and pro-inflammatory injuries. The critical role of IRF1 in connecting the adaptive and native immune systems has been reported in the literature [24]. Growth inhibition of various human cell types due to IRF1 overexpression is attributed to its regulatory effects on targeted genes that downregulate cell growth and proliferation. Investigations indicate that the upregulation of IRF1 significantly suppresses (15-fold) levels of the survival protein. This is similar to cancer therapy in breast cancer. Similar to P53, IRF1 acts as a tumor suppressor and regulates DNA damage-induced apoptosis [25].

Solute carrier family 2 (facilitated glucose transporter), member 2 (SLC2A2) is another upregulated gene in response to the presence of AB1 toxin in the treated cells. The SLC2A2, which is known as glucose transporter type 2 (GLUT2), is complicated with glucose transport into cells. This function of SLC2A2 is linked to its role in the overexpression associated with tumor invasiveness and development [26]. Chai et al. investigation demonstrated that the overexpression of SLC2 family members was associated to poor survival outcomes in papillary thyroid carcinoma [27]. The transcription co-factor Cbp/p300-interacting transactivator with Glu/Asp-rich carboxy-term-inal domain 2 (CITED2) is another highlighted gene in the current study. Technically, CITED2 plays significant roles in various cellular processes, including apoptosis, prolifer-ation, migration, differentiation and autophagy as address-ed in the literature [28]. As demonstrated in Figure 3, CITED2 was connected to the mitophagy biochemical pathway.

Sulfotransferase family cytosolic 2A dehydroepiandro-sterone (DHEA)-preferring member 1 (SULT2A1) is associated to the bile secretion pathway (Figure 3). Prominent role of the sulfotransferase family in regulating hormones and excreting xenobiotics is verified by the researchers [29]. It has been reported that SULT2A1 plays a role in initiation and progression of hepatocellular carcinoma [30]. Related RAS viral (r-ras) oncogene homolog (RRAS) such as CITED2 is linked to the mitophagy biochemical pathway. The R-RAS subfamily is closely associated to RAS proteins. Studies have shown that RRAS proteins are involved in regulating various cellular functions, including cell migration, adhesion and morphological changes [31]. Downregulation of RRAS has been documented in progression and pathogenesis of gliomas [32]. The last highlighted gene was phosphate cytidylyltransferase 1 choline beta (PCYT1B), which is associated with the glycerophospholipid biochemical pathway. This gene includes a mutual inhibition relation-ship with MAPK15 (Figure 5). The PCYT1B is involved in the Kennedy pathway [33]. Stored lipids in cells form lipid droplets, including a neutral lipid core and a peri-pheral phospholipid monolayer, are primarily composed of phosphatidylcholine. Balances between the neutral lipid core and the outer layer result from the activity of the Kennedy pathway [34]. The AB1 is known as a potent contaminant and a harmful type of aflatoxin. In fact, AB1 is associated with immune system suppression, character-istics of hepatocarcinogenesis, and mutagenic and tera-togenic effects. Additionally, AB1 exposure significantly affects metabolic processes, highlighting comprehensive biochemical disruptions caused by this mycotoxin (Figure 3).

To minimize aflatoxin contamination and increase food safety, modification in farming practices and use of resistant crop varieties are recommended [35]. Due to the widespread contamination of food and agricultural products by mycotoxins, many people face health risks associated with the consumption of aflatoxin-contaminated foods [36]. Level of the expressed biomarkers such as the recommended genes (COL4A6, IRF1, SLC2A2, CITED2, SULT2A1, RRAS and PCYT1B) is an appropriate criterion with aflatoxin measurement for introducing a novel standard method to detect practical harmful levels of aflatoxin contamination. The present findings suggest that the expression levels of these targeted genes can serve as reliable biomarkers for assessing aflatoxin contamination, potentially complementing existing detection methods. Regarding the current risks posed by aflatoxin contamination in food and agricultural products, integra-tion of the biomarker-based criteria into standard detection protocols can enhance food safety measures and hence public health consequences. Future studies should focus on validating these biomarkers in clinical settings and invest-igating further interplays between the aflatoxin exposure and metabolic pathways to develop effective moderation strategies.

  1. Conclusion

In conclusion, exposure to AB1 significantly alters numerous cellular functions, including cell proliferation, migration and apoptosis as well as various metabolic processes in human intestinal Caco-2 cells. The COL4A6, IRF1, SLC2A2, CITED2, SULT2A1, RRAS and PCYT1B were identified as the critical targeted genes for AB1. Due to the significant effects of aflatoxin contamination on food and agricultural products, it has been concluded that the levels of these critical targeted genes in response to aflatoxin can be addressed as an appropriate criterion for modifying standard methods of aflatoxin contamination assessments. Several trends are anticipated in the field of aflatoxin detection and management. First, integration of multiple-omics approaches such as genomics, proteomics and metabolomics can provide a further comprehensive understanding of the aflatoxin biological effects and enhance biomarker discovery. Second, use of machine learning algorithms to analyze complex datasets may facilitate identification of novel biomarkers and improve accuracy of detection methods. Third, educating farmers and food manufacturers on aflatoxin prevention strategies and diminishing techniques is critical in decreasing risks of aflatoxin exposure in the food supply chain. These not only include promises for improving food safety but also for enhancing overall understanding of mycotoxin-linked health effects. However, it is difficult to introduce the real safe levels of contamination.

  1. Ethical Code:

This study was approved via

IR.SBMU.RETECH.REC.1403.423 ethical code.

  1. Acknowledgements

This study was supported by Shahid Beheshti University of Medical Sciences.

  1. Author contributions:

Vahid Mansouri contributed to the conception and design of the study and literature review. All authors participated equally in study administration and writing of the primary draft of the manuscripts, providing critical revision and editing. All authors approved the final version of the manuscript.

References

  1. Amaike S, Keller NP. Aspergillus flavus. Annu Rev Phytopathol. 2011; 49(1): 107-133.

https://doi.org/10.1146/annurev-phyto-072910-095221

  1. Garcia-Lopez MT, Meca E, Jaime R, Puckett RD, Michailides TJ, Moral J. Sporulation and Dispersal of the Biological Control Agent Aspergillus flavus AF36 Under Field Conditions. Phytopathology. 2024; 114(5): 1118-1125.

https://doi.org/10.1094/PHYTO-06-23-0200-KC

  1. Allwood JG, Wakeling LT, Bean DC. Fermentation and the microbial community of Japanese koji and miso: A review. J Food Sci. 2021; 86(6): 2194-2207.

https://doi.org/10.1111/1750-3841.15773

  1. Alshannaq AF, Gibbons JG, Lee M-K, Han K-H, Hong S-B, Yu J-H. Controlling aflatoxin contamination and propagation of Aspergillus flavus by a soy-fermenting Aspergillus oryzae strain. Sci Rep. 2018; 8(1): 16871.

https://doi.org/10.1038/s41598-018-35246-1

  1. Frisvad JC, Hubka V, Ezekiel CN, Hong SB, Nováková A, Chen AJ, Arzanlou M, Larsen TO, Sklenář F, Mahakarnchanakul W, Samson RA, Houbraken J. Taxonomy of Aspergillus section Flavi and their production of aflatoxins, ochratoxins and other mycotoxins. Stud Mycol. 2019; 93(1): 1-63.

https://doi.org/10.1016/j.simyco.2018.06.001

  1. Varga J, Frisvad J, Samson R. A reappraisal of fungi producing aflatoxins. World Mycotoxin J. 2009; 2(3): 263-277.

https://doi.org/10.3920/WMJ2008.1094

  1. Grenier B, Applegate TJ. Modulation of intestinal functions following mycotoxin ingestion: Meta-analysis of published experiments in animals. Toxins. 2013; 5(2): 396-430.

https://doi.org/10.3390/toxins5020396

  1. Elzaki MEA, Xue R-r, Hu L, Wang J-d, Zeng R-s, Song Y-y. Bioactivation of aflatoxin B1 by a cytochrome P450, CYP6AE19 induced by plant signaling methyl jasmonate in Helicoverpa armigra (Hübner). Pestic Biochem Physiol. 2019; 157: 211-218.

https://doi.org/10.1016/j.pestbp.2019.03.020

  1. Sergent T, Ribonnet L, Kolosova A, Garsou S, Schaut A, De Saeger S, Van Peteghem C, Larondelle Y, Pussemier L, Schneider YJ. Molecular and cellular effects of food contaminants and secondary plant components and their plausible interactions at the intestinal level. Food Chem Toxicol. 2008; 46(3): 813-841.

https://doi.org/10.1016/j.fct.2007.12.006

  1. Gao Y, Meng L, Liu H, Wang J, Zheng N. The compromised intestinal barrier induced by mycotoxins. Toxins. 2020; 12(10): 619.

https://doi.org/10.3390/toxins12100619

  1. Brück S, Strohmeier J, Busch D, Drozdzik M, Oswald S. Caco‐2 cells–expression, regulation and function of drug transporters compared with human jejunal tissue. Biopharm Drug Dispos. 2017; 38(2): 115-126.

https://doi.org/10.1002/bdd.2025

  1. Ji J, Wang Q, Wu H, Xia S, Guo H, Blaženović I, Zhang Y, Sun X. Insights into cellular metabolic pathways of the combined toxicity responses of Caco-2 cells exposed to deoxynivalenol, zearalenone and Aflatoxin B1. Food Chem Toxicol. 2019; 126: 106-112.

https://doi.org/10.1016/j.fct.2018.12.052

  1. Zhang J, Zheng N, Liu J, Li F, Li S, Wang J. Aflatoxin B1 and aflatoxin M1 induced cytotoxicity and DNA damage in differentiated and undifferentiated Caco-2 cells. Food Chem Toxicol. 2015; 83: 54-60.

https://doi.org/10.1016/j.fct.2015.05.020

  1. Dohnal V, Wu Q, Kuča K. Metabolism of aflatoxins: Key enzymes and interindividual as well as interspecies differences. Arch Toxicol. 2014; 88: 1635-1644.

https://doi.org/10.1007/s00204-014-1312-9

  1. Scholl PF, Turner PC, Sutcliffe AE, Sylla A, Diallo MS, Friesen MD, Groopman JD, Wild CP. Quantitative comparison of aflatoxin B1 serum albumin adducts in humans by isotope dilution mass spectrometry and ELISA. Cancer Epidemiol Biomarkers Prev. 2006; 15(4): 823-826.

https://doi.org/10.1158/1055-9965.EPI-05-0890

  1. Engin AB, Engin A. DNA damage checkpoint response to aflatoxin B1. Environ Toxicol Pharmacol. 2019; 65: 90-96.

https://doi.org/10.1016/j.etap.2018.12.006

  1. Rivlin N, Brosh R, Oren M, Rotter V. Mutations in the p53 tumor suppressor gene: Important milestones at the various steps of tumorigenesis. Genes & cancer. 2011; 2(4): 466-474.

https://doi.org/10.1177/1947601911408889

  1. Cai P, Zheng H, She J, Feng N, Zou H, Gu J, Yuan Y, Liu X, Liu Z, Bian J. Molecular mechanism of aflatoxin-induced hepatocellular carcinoma derived from a bioinformatics analysis. Toxins. 2020; 12(3): 203.

https://doi.org/10.3390/toxins12030203

  1. Gauthier T, Duarte-Hospital C, Vignard J, Boutet-Robinet E, Sulyok M, Snini SP, Alassane-Kpembi I, Lippi Y, Puel S, Oswald IP, Puel O. Versicolorin A, a precursor in aflatoxins biosynthesis, is a food contaminant toxic for human intestinal cells. Environ Int. 2020; 137: 105568.

https://doi.org/10.1016/j.envint.2020.105568

  1. Segal Y, Zhuang L, Rondeau E, Sraer J-D, Zhou J. Regulation of the Paired Type IV Collagen GenesCOL4A5 and COL4A6: Role Of The Proximal Promoter Region. J Biol Chem. 2001; 276(15): 11791-11797.

https://doi.org/10.1074/jbc.M007477200

  1. Ma J-B, Bai J-Y, Zhang H-B, Gu L, He D, Guo P. Downregulation of collagen COL4A6 is associated with prostate cancer progression and metastasis. Genet Test Mol Biomark. 2020; 24(7): 399-408.

https://doi.org/10.1089/gtmb.2020.0009

  1. Xu Y, Jin H, Chen Y, Yang Z, Xu D, Zhang X, Yang J, Wang Y. Comprehensive analysis of the expression, prognostic, and immune infiltration for COL4s in stomach adenocarcinoma. BMC Med Genomics. 2024; 17(1): 168.

https://doi.org/10.1186/s12920-024-01934-3

  1. Stanley SL. Amoebiasis. The Lancet. 2003; 361 (9362): 1025-1034.

https://doi.org/10.1016/S0140-6736(03)12830-9

  1. Dou L, Liang H-F, Geller DA, Chen Y-F, Chen X-P. The regulation role of interferon regulatory factor-1 gene and clinical relevance. Hum Immunol. 2014; 75(11): 1110-1114.

https://doi.org/10.1016/j.humimm.2014.09.015

  1. Alsamman K, El-Masry OS. Interferon regulatory factor 1 inactivation in human cancer. Biosci Rep. 2018; 38(3): BSR20171672.

https://doi.org/10.1042/BSR20171672

  1. Zhang W, Zhou D, Song S, Hong X, Xu Y, Wu Y, Li S, Zeng S, Huang Y, Chen X, Liang Y, Guo S, Pan H, Li H. Prediction and verification of the prognostic biomarker SLC2A2 and its association with immune infiltration in gastric cancer. Oncol Lett. 2023; 27(2): 70.

https://doi.org/10.3892/ol.2023.14203

  1. Chai YJ, Yi JW, Oh SW, Kim YA, Yi KH, Kim JH, Lee KE. Upregulation of SLC2 (GLUT) family genes is related to poor survival outcomes in papillary thyroid carcinoma: Analysis of data from the cancer genome atlas. Surgery. 2017; 161(1): 188-194.

https://doi.org/10.1016/j.surg.2016.04.050

  1. An B, Ji X, Gong Y. Role of CITED2 in stem cells and cancer. Oncol Lett. 2020; 20(4): 107.

https://doi.org/10.3892/ol.2020.11968

  1. Yalcin EB, More V, Neira KL, Lu ZJ, Cherrington NJ, Slitt AL, King RS. Downregulation of sulfotransferase expression and activity in diseased human livers. Drug Metab Dispos. 2013; 41(9): 1642-1650.

https://doi.org/10.1124/dmd.113.050930

  1. Peng H, Feng K, Jia W, Li Y, Lv Q, Zhang Y. An integrated investigation of sulfotransferases (SULTs) in hepatocellular carcinoma and identification of the role of SULT2A1 on stemness. Apoptosis. 2024; 29(5): 898-919.

https://doi.org/10.1007/s10495-024-01938-5

  1. Weber SM, Carroll SL. The role of R-Ras proteins in normal and pathologic migration and morphologic change. Am J Pathol. 2021; 191(9): 1499-1510.

https://doi.org/10.1016/j.ajpath.2021.05.008

  1. Ohta T, Kim YH, Oh JE, Satomi K, Nonoguchi N, Keyvani K, Pierscianek D, Sure U, Mittelbronn M, Paulus W, Vital A. Alterations of the RRAS and ERCC1 genes at 19q13 in gemistocytic astrocytomas. J Neuropathol Exp Neurol. 2014; 73(10): 908-915.

https://doi.org/10.1146/annurev-phyto-072910-095221

  1. Wang K, Xu H, Zou R, Zeng G, Yuan Y, Zhu X, Zhao X, Li J, Zhang L. PCYT1A deficiency disturbs fatty acid metabolism and induces ferroptosis in the mouse retina. BMC Biol. 2024; 22(1): 134.

https://doi.org/10.1186/s12915-024-01932-y

  1. Moessinger C, Klizaite K, Steinhagen A, Philippou-Massier J, Shevchenko A, Hoch M, Ejsing CS, Thiele C. Two different pathways of phosphatidylcholine synthesis, the Kennedy Pathway and the Lands Cycle, differentially regulate cellular triacylglycerol storage. BMC Cell Biol. 2014; 15: 1-17.

https://doi.org/10.1186/s12860-014-0043-3

  1. Kumar A, Pathak H, Bhadauria S, Sudan J. Aflatoxin contamination in food crops: Causes, detection, and management: A review. Food Prod Process. Nutr. 2021; 3: 1-9.

https://doi.org/10.1186/s43014-021-00064-y

  1. Jallow A, Xie H, Tang X, Qi Z, Li P. Worldwide aflatoxin contamination of agricultural products and foods: From occurrence to control. Compr Rev Food Sci Food Saf. 2021; 20(3): 2332-2381.

https://doi.org/10.1111/1541-4337.12734

Recent advances in engineered edible probiotic vaccines: Promising agents in the effectiveness of probiotics

Nader Hashemi, Forough Shams, Elham Sharif, Maedeh Bakhshi Valilou

Applied Food Biotechnology, Vol. 11 No. 2 (2024), 26 Aban 2024, Page e5
https://doi.org/10.22037/afb.v11i2.46302

Background and Objective: Associations between probiotics as dietary supplements for health enhancement and illness management and human health include a long history. Currently, probiotics represent a potential category of microorganisms used in the development of oral vaccines for the treatment of allergies, infectious diseases, and cancers. The vaccine promises as safe therapeutic options, their capacity to elicit mucosal and systemic immune responses, and their cost-effectiveness resulting from the absence of complex purification processes have been addressed.

Results and Conclusion: Despite the advantages of probiotics as oral vaccines, their uses still include problems such as inadequate targeted colonization, diminished immune response in populations with low hygiene standards, reliance on individual microbiota, poor stability, limited efficacy, and absence of targeted immunogenicity. To address these problems, probiotics can be engineered using gene editing technologies, particularly CRISPR/Cas system. Concerns are reported regarding the safety of genetic alterations and deficiencies in efficient delivery mechanisms linked to the use of modified probiotics as oral vaccines. Further studies are needed to assess problems associated with accurate genetic alteration and efficient delivery methods to achieve the ultimate goal of further effective and safer vaccinations.

Conflict of interest: The authors declare no conflict of interest.

  1. Introduction

 

The term “probiotics” was generally specified to describe live microorganisms and substances that assist and promote intestinal microbial balances. However, this term was changed by the joint FAO/WHO group as “living microorganisms that when used in adequate contexts, provide a health benefit”. The relationship between the microbiome and human health provides new opportunities for developing novel biotherapeutics. Probiotic research has revealed a myriad of potential health benefits, particularly in the modulation of the immune system and production of bioactive compounds for a broad spectrum of uses such as skin health, oral health and treatment of specific health conditions, including metabolic disorders, allergies, inflammatory bowel disease (IBD) and cancers. Nobel Prize was awarded to Dr. Metchnikoff at the beginning of the last century for the positive effects of probiotics and live microorganisms such as Lactobacillus spp. in fermented milks on human health and life span [1].

Vaccination is addressed as one of the most significant achievements in global public health. The ongoing evolution in vaccine development has introduced seven types of vaccines, including inactivated, live-attenuated, biosynthetic, toxoid, recombinant, DNA, and edible vaccines. Each type includes its own set of potential challenges, particularly regarding production complexities and high costs associated with downstream processing and purification [2]. Nowadays, probiotics are a promising generation of microorganisms used to produce edible vaccines. Recent advancements have highlighted the promise of edible vaccines as safe agents in possible pathogenicity, good mucosal and systematic immunity stimulations, and reasonable costs due to the lack of complex purification processes [3, 4]. Although probiotic vaccines are not still approved, ongoing research into the complex interplay in key agents of probiotics' immune system, microbiota, and antigens is investigated [5]. Precise molecular mechanisms of probiotic action are still under investigation. However, probiotics can affect compositions and types of bacteria in the gut due to direct affection of bacterial colonization. In recent years, studies have been carried out on the positive and negative effects of intestinal microbiota changes on development of autoimmune, autoinflammatory and infectious diseases. Furthermore, probiotics can direct the immune system to specific immune responses and specific functions to decrease inflammation in the target strains, showing their roles as vaccines based on their characteristics [6].

Recent advances in novel technology such as genetic sequencing, biomolecular biology, microbiology, bioinfor-matics, and medical immunology have discovered complex relationships between the microbiota of various organs and conditions of health or disease. These approaches have helped researchers look for ways to intervene in the colonization pattern of microbiota and make effective edible probiotic vaccines to improve various pathological conditions. One of the most important types of probiotic vaccines is recombinant vaccines. Genome engineering and gene editing technologies such as zinc finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN), and CRISPR/Cas-linked nucleases as a bioactive field have made valuable progress in various aspects [7]. Therefore, edible probiotic vaccines with improved beneficial attributes have been engineered more frequently in recent years. Hence, the aim of this study was to introduce mucosal and systemic immune modulating potentials of probiotics as oral vaccines and use advanced genome-editing technologies to promote these micro-organisms as antigen carriers and producers. Furthermore, problems in converting these microorganisms into therapeutic substitutes for current vaccinations are discussed. This study focused on the immune-modulating role of probiotics and their interactions to increase systemic protective immunity. Furthermore, assessing the effectiveness of the engineered probiotics based on novel genome editing tools and their challenges in generation of edible probiotic vaccines were addressed.

  1. Edible vaccines

Vaccination is the process of preparing the body to combat pathogens. This therapeutic approach is different from the conventional techniques of treatment, often administered post emergence of a certain disease. Vaccines not only prime the immune system to combat specific pathogens, but also provide long-lasting protection against them. Due to this function, vaccines have effectively contributed to a significant decrease in devastating infec-tious diseases over time [8]. Vaccines often consist of epitope/antigen structures that have restricted activity, particularly designed to activate immune system of the host without causing damages. Vaccines are often administered by injection [intramuscularly, subcutan-eously, or intradermally]. However, there are limitations associated with injectable vaccines. Injectable vaccination needs disposable equipment [e.g. syringes and gloves], strict rules for transportation [e.g. cold chain] and high costs of products linked to injectable vaccines [9]. Furthermore, administration of injectable vaccinations may lead to complexities such as allergic immunological reactions and localized edema, which can eventually prevent patients from accepting the vaccine. Therefore, the use of alternative formulations has always been an important concern of scientists. With the progress in vaccine technology, the edible vaccine model emerged as an effective solution. In this type of vaccine, antigen production is carried out in various organisms such as plants, algae, silkworms, yeasts, and microorganisms known as probiotics [10]. Edible vaccines exert their immunity by immunizing the intestinal mucosa. Furthermore, edible vaccinations are appropriate for every age range and user-friendly, as they do not necessitate professionals to provide the doses [11].

  1. Gut Microbiota: Functions & Benefits

Probiotics are prokaryotic microorganisms generally recognized as safe (GRAS). Probiotics possess a distinctive capacity to activate the immune system [12]. Due to their ability to express antigens and deliver them to specific immunological targets, these microorganisms include the potential as highly effective agents for vaccination. Bacillus subtilis, Saccharomyces cerevisiae , and lactic acid bacteria [LAB] are examples of probiotics commonly used in this sector [10]. In addition, intro-duction of interested genes has enabled the expression of foreign proteins in probiotic systems. Furthermore, presence of antigens within cells, outside or on the surface of cells, may carefully be addressed when developing probiotics as targeted agents for immunization purposes [13]. Up to date, numerous antigens have been produced in diverse probiotic strains. Therefore, scientists have carried out extensive research to develop probiotics as targeted mucosal vaccines, demonstrating their substantial poten-tials for immunization. Moreover, several probiotics have clinically been assessed [14]. This development increases optimism for the shift from conventional vaccine prototypes to edible ones.

3.1 Advantages of probiotics as edible vaccines

Probiotic microorganisms possess qualities that make them promising candidates for edible vaccination treat-ments. Probiotics are prevalent non-colonizing microorganisms detected in the gastrointestinal tract (GIT). Thus, they include low adverse effects. Probiotic-based therapeutic vaccines offer further cost-effective alternat-ives to other types of immunization. The optimal dose of vaccination may readily be assessed based on the quantity of probiotics. Additionally, probiotics can include a large effect by outperforming pathogens, generating materials that are toxic to pathogens, or inhibiting their ability to attach to intestinal epithelia [15]. These microorganisms serve a dual function by producing appropriate antigens and acting as carriers for oral administration, allowing desired antigens to present to immune cells in the gut. These microorganisms are present in the bodies of individuals worldwide. Therefore, produced vaccine can be manufactured and administrated globally [10]. Due to their edible nature, this type of vaccines includes adaptations from pediatric to geriatric patients. Due to the lack of side effects associated with probiotic-based vaccines, people of any age can take probiotics at any time of the day [16]. Therefore, because of to their unique character-istics, probiotics are more appropriate candidates for edible vaccine production than others. For example, Bacillus spp. are distinguished by their ability to form endospores, which show exceptional resilience under hostile condi-tions. Hence, these attributes make them appropriate carriers for edible vaccines in dormant or vegetative forms [17]. Genetic manipulation of probiotics has been verified as reliable and effective over an extended period, demonstrating its efficacy in preventing bacterial and parasitic infections, such as malaria. Using of these engineered probiotics, which contain antigens specific to viruses, includes promises. These have been provided to fight against viral infections such as HIV, herpes virus, and influenza virus[18]. Moreover, these microorganisms may genetically be modified to enhance production of immune components, including cytokines and interleukins, which can help prevent cancers [19].

3.2 Edible probiotics and the immune response

In 1907, Elie Metchnikoff first discovered that probiotics could activate the immune system. There are several ways; by which, this interaction can be accomplished, including probiotics targeted by pattern recognition receptors (PRRs) on intestinal epithelial cells through their microorganism-associated molecular patterns. This interaction facilitated their phagocytosis, further stimulating mucosal, and systemic immunity by antigen-presenting cells (APC) activity [20]. Probiotics exert their immunogenicity by affecting intestinal immunity. Intestinal immunity is achieved by maintaining homeostasis and controlled by gut-associated lymphoid tissues, antimicrobial peptides, Peyer's patches, secretory immunoglobulins of IgA (sIgA), T cells, and various inflammatory mediators such as cytokines, chemokines, and beneficial microorganisms. The intestinal epithelial layer contains specialized cells, including entero-endocrine cells, goblet cells, paneth cells, enterocytes, tufts and M cells, which are critical components of mucosal immunity. The M cells are antigen-delivery cells that transfer luminal particles and antigens to APC. These cells are responsible for receiving antigens and subsequent transmission of the antigens to the underlying immune cells via the mecha-nisms of transcytosis, phagocytosis, and shedding of macrovesicles [21].

Goblet cells are linked to the process of presenting antigens to dendritic cells [22]. Dendritic cells, functioning as APCs, include an essential role in regulating cellular and humoral immune responses. By receiving antigenic subunits, these cells present them through the major histocompatibility complexes (MHC) I and II, which further activate the T cell set and create a cellular immune response through the differentiation of cell subsets such as CD4+ cells, macrophages, and Foxp3+ T-cells. They enter mesenteric lymph nodes and induce the gut-homing process [23]. Additionally, APCs stimulate activation of B cells and subsequently promote the synthesis of IgA secretory antibodies. These antibodies play a critical role in generating humoral immune responses. When probiotics are used as carriers or expressers of a particular antigen, they can stimulate specific cellular and humoral immunities associated with the antigen [24].

3.3 Engineered probiotics in edible vaccines

Using probiotics in edible vaccinations can effectively address specifically targeted immune responses that are not in conventional delivery techniques such as attenuated pathogens [25]. The edible vaccination uses probiotics as antigen carriers and adjuvants. It is important to address engineering of the immune responses to a single heterologous antigen rather than the whole microorganism [26]. The LAB strains are ideal for expressing one or more foreign proteins, particularly on their surfaces. This is because LAB are non-pathogenic and non-invasive and can stimulate a specific immune response. Probiotics may present antigens to specific immune cells as either proteins or DNA. Hence, the probiotic bacteria transfers antigen-linked DNA into the APCs, enabling these cells to produce the active antigen. To improve efficiency, desired prob-iotics may genetically be modified to possess the essential mechanisms for invasion [27]. Creating and programming induction of probiotics used as edible vaccines are possible.

In response to quorum sensing signals sent by tumors, bacteria and viruses, these cells secrete specific molecules that counteract these agents. In addition, researchers are investigating and developing probiotics that specifically target the immune responses of oral mucosal pathways, including buccal, gingival, and sublingual mucosa, to enhance mucosal and systemic immune responses. Sublingual administration bypasses mucosal barriers of the GIT and may be considered as an alternative method to improve the efficacy of sensitive immunizations [28].

3.4 Highlights regarding formulation of probiotics as edible vaccines

Purified antigens are often sensitive to various mucosal barriers, resulting in the attenuation of their immunogenicity. Using probiotic bacteria as antigen producers and carriers can solve this problem in several ways, including safeguarding the target antigen in high acidic and basic environments and enhancing delivery of the target antigen to specific immune cells (M-cells in gut-associated lymphoid tissues) [29]. Edible probiotic vaccines should be assessed from several aspects, including A) stability of the formulation, since vaccine formulation is prepared orally, the most important expected goal is to stabilize formulations containing probiotics and antigens against the harsh conditions of the digestive system such as acidic and basic pH values, bile salts and enzymatic breakdowns. Thus, it is necessary to develop an edible formulation that can resist these circumstances [30]. Furthermore, it is critical to maintain the viability of the live microorganism, especially when the production of new antigens is associated with the proliferation of these germs in the host and preserving the immunogenicity of the antigen is provided through the production of an edible vaccine using probiotics. This includes precautions during processes such as lyophil-ization [31]. If necessary, cellular modifications should be used to enhance the microbial resistance in the relevant area. Hence, development of an optimal vaccine using probiotics needs the establishment of immunological tolerance against these microorganisms. Use of probiotics as enhancers and transport systems for delivering and administering purified antigens has grown due to technical advancements such as MimoPath systems. An example of this scenario includes the Gram-positive Lactococcus lactis, which tolerates treatments under highly acidic conditions. Then, its biological components are eliminated, leading to the emergence of bacteria-like particles (BLPs). These BLPs are then combined with a pure antigen in the following stage, resulting in production of an Ag-BLP vaccine [29, 32]. B) appropriate design of the target antigen to bind to the specific immune cells and the optimal edible vaccination should have mucosal adhesion capabilities. The antigen in the vaccine must attach to a certain cell matrix such as M cells to initiate an immune response. Hence, it is critical to create edible vaccines that show mucosal adhesion qualities and effectively transport target antigens to prompt a potent immune response; and C) immune response stability, due to the limited colonization capacity of probiotics in the GIT, antigens in the edible probiotic vaccines include a short duration to induce the immune cells in the target. One solution is to co-administer an adjuvant with an edible probiotic vaccination. This can enhance the immune system activity and improve its effectiveness at the suggested site [26].

  1. Genetic engineering in probiotics

Recent advancements in synthetic biology, genetic engineering, and genome sequencing technologies have strengthened the current understanding of microbial functions and promoted the development of so-called designer probiotics [33]. Investigating genome-engineering techniques has reached an innovative milestone and broadening its potential use within the medical, agri-cultural, and food sectors. This technology can be used for humans, provided precautions are highlighted to eradicate antibiotic resistance and ensure that modifications involve the organism's own DNA or DNA from GRAS organisms [34]. Significant uses of probiotics necessitate compre-hensive genome analysis of these microorganisms. Advanced omics methodologies and system biology approaches enable researchers to optimize metabolic processes within the gut microbiome, revealing previously uncharacterized biosynthetic pathways. Such insights promote novel metabolic engineering strategies, facilitating genetic manipulation of the probiotics to address their roles in intestinal microbiota and enhance their beneficial characteristics [35]. Gene editing techn-iques such as site-specific recombination and the advent of innovative gene editing tools such as TALEN, ZFNs, and CRISPR/Cas9 help precise targeting and modification of specific genes (Figure 1). Merging CRISPR technology, synthetic biology and high-throughput genomic analysis represents a significant advancement in development of personalized probiotic treatments. Developing further robust and effective CRISPR editing systems for probiotics presents two key objectives: improving probiotic traits and engineering probiotics for therapeutic uses. The first goal focuses on enhancing the inherent qualities of probiotics. This includes targeted localization of probiotics within specific niches of the gut microbiome, stress tolerance, decreased allergenicity, and virulence factors in probiotics to ensure their safety for widespread uses. The second goal emphasizes the potential of engineered probiotics for specific therapeutic functions such as autoimmune diseases, cancers, metabolic disorders, and infectious diseases. By focusing on the improvement of probiotic traits and developing targeted therapies, scientists can create safer and more effective probiotics that not only enhance individual well-being but also contribute to further understanding of microbiome interactions within the host (Figure 2) [36].

4.1 Engineered probiotic strains: therapeutic, prophylactic, and diagnostic vaccines

The field of probiotic engineering is significantly transformed, using bacterial strains that are specifically designed to colonize the GIT and produce specific molecules to serve as therapeutic, prophylactic, and diagnostic vaccines [37]. Key mechanisms of action include immunomodul-ation, where probiotics produce cytokines, antigens and allergens, and pathogen exclusion through the synthesis of antimicrobial peptides, biosensing capabilities useful for disease diagnosis, and metabolic modifications within the host [38]. The rational engineering of probiotics involves a systematic process that encompasses three essential steps of the selection of novel health-promoting strains, exploration of their molecular interactions with the host and its microbiota and the genetic modification to enhance or design functional characteristics that support their probiotic characteristics [39]. Several species have been investigated for their potential as vaccine vectors, effectively delivering a wide range of bacterial and viral antigens [40]. Various delivery systems, including cell wall, extracellular and cytoplasmic media, have been designed to target heterologous proteins to specific cellular locations [41]. Production of genetically modified probiotics over the past decade has expanded the scope of mucosal delivery systems to include not only prophylactic vaccines but also therapeutic agents such as enzymes, cytokines, and allergens [42]. Probiotic-based bacterial ghost vaccination represents a promising advancement in immunotherapy, particularly for DNA and protein antigen vaccines [43]. Traditional naked DNA vaccines often suffer from weak immunogenicity, which necessitates incorporation of adjuvants. However, limitations of the current adjuvants and restricted options available for recombinant vaccine manufacturers highlight necessity of alternative strategies to improve vaccine efficacy [44]. Bacterial ghosts, which are non-viable bacterial cells that include their structural integrity, offer a unique platform for vaccine delivery. They can efficiently be internalized by various cell types, including Caco-2 cells, human dendritic cells, dendritic cells, and RAW 264.7 macrophages [45].

In a diverse group of microorganisms, Lactobacillus spp. are highlighted, particularly for their members, which comprise a significant portion of the human gut microbiota. Key probiotic strains, including L. acidophilus L. plantarum, L. johnsonii, L. gasseri, L. casei, L. reuteri, L. salivarius, and L. brevis, are important for their contributions to gastrointestinal health [35]. Engineering specific Lacticaseibacillus spp. to express anti-Listeria monocytogenes antigens [46], L. lactis secrete IL-10, L. lactis CHW9 has been used to produce the peanut Ara 2 allergen, and L. lactis NZ9800 has been generated to deliver the major birch allergen of Bet-v1 [47]. Furthermore, innovative methodologies have emerged in the realm of plant-probiotic interplay, wherein modified plant viruses such as tobacco mosaic virus and geminiviruses function as advanced plant expression systems and serve as vectors for delivering genetic components into plant cells [48]. In various platforms for creating edible vaccines, genetically modified microalgae, particularly Chlamydomonas reinhardtii, have emerged as significant candidates due to their unique characteristics and advantages, compared to traditional plant systems [49]. Developmental process of an edible vaccine using microalgae involves the insertion of a pathogen's gene of interest (GOI) into the microalgal genome through various genetic engineering techniques. Industrial benefits of microalgae such as scalability and ease of cultivation make them as further versatile options for vaccine production [50]. Gregory et al. have demonstrated that C. reinhardtii can successfully express unmodified versions of these proteins, yielding antibodies that inhibit the sexual development of malaria parasites [51]. Several engineered probiotics have reached preclinical and clinical trial phases, highlighting their potentials as edible vaccines. Noteworthy examples include recombinant Bifidobacteria longum, the oral bacTRL-Spike vaccine designed to immunize against coronavirus disease 2019 [52], recombinant L. lactis [LL-Thyy12] to treat patients with Crohn’s disease [53], L. lactis AG019 developed for type-1 diabetes treatment [54], recombinant L. casei expressing the HPV16 E7 protein [55], and recombinant L. lactis-derived anti-malarial vaccine [56]. Clinical trials assessing the efficacy of recombinant LAB have yielded promising results, highlighting their viability as live biotherapeutics. With the current research and development, the integration of LAB in DNA vaccine delivery systems includes the potency to revolutionize preventive and therapeutic medicines [57].

4.2 Genetic design tools used to engineer probiotics

Genome editing techniques frequently used in the engineering of probiotics include gene knockouts, gene insertions, and targeted mutations. Efficacy and precision of various genome-editing methods play critical roles in shaping functionality and use of engineered probiotics. Probiotic engineering can be approached through two principal strategies top-down and bottom-up designs. Top-down approaches involve genome decreases, where existing probiotic strains are simplified to create a minimal genome chassis containing essential genes for survival and metabolic functionality. In contrast, bottom-up approaches include synthesis of a minimal genome with synthetic oligonucleotides, needing careful assembly of genetic elements, metabolic pathways, and cellular membranes. Being innovative, these methods are complex and resource intensive [58].

Synthetic biology tools enhance efficiency of genomic modifications and development of counter-selectable markers has been carried out. Traditional strategies using double-stranded (dsDNA) or single-stranded (ssDNA) recombineering enable knock-in of GOI; however, limitations are reported regarding size and copy number of the inserted DNA fragments. Role of synthetic biology tools such as site-specific genome editing systems, homologous recombination, and recombineering, is critical in advancing probiotic strains. Recombineering represents a phage-derived approach that facilitates gene modification through in vivo homologous recombination [47]. This Red/RecET system has effectively been used in multiple Lactobacillus strains, allowing for fine-tuning of genomes through subtle modifications such as point mutations. In addition to these precise methodologies, strategies using non-homology based approaches such as transposon-mediated random DNA insertion broaden the scope of genetic engineering in non-model organisms. To avoid these restrictions and facilitate the integration of larger DNA constructs or clusters, site-specific recombination systems have emerged as valuable alternatives [59]. Induction of double-strand breaks (DSBs) through targeted nucleases such as ZFNs, TALEN, and RNA-guided nucleases can prompt repair mechanisms but includes risks as probiotics may show pronounced susceptibility to DSBs, leading to cell death [60].

Various methods are described to deliver genetic elements into probiotics. Direct gene delivery or vector-independent nature methods play essential roles in plant biotechnology such as biolistic, enabling the introduction of desired DNA, or RNA directly into the nuclear and chloroplast of plant cells. Chloroplast transformation, as a preferred method for producing edible vaccines, has attracted intentions owing to its ability to generate high levels of protein expression while minimizing risks of gene dispersal and horizontal gene transfer to other organisms [61]. Biolistic methods have successfully been used to develop vaccines against numerous pathogens, including cholera, Lyme disease, rotavirus, and canine parvovirus [62]. In contrast, indirect gene delivery methods use vector-mediated approaches to achieve gene transfer. A prominent example includes Agrobacterium-mediated gene transfer of foreign genes into the plant nucleus. This method has been set to create vaccines against various diseases, including diarrhea, tuberculosis, dengue fever, avian flu, and Ebola [63]. Despite the promising potential of editing techniques, use of genome editing for probiotic LAB is limited, compared to model organisms such as Escherichia coli and S. cerevisiae. This limitation largely originates from stringent regulatory frameworks and cautious market response to genetically modified organisms (GMOs) [64]. Introduction of counter-selectable markers, including upp, oroP, pheS, and mazF, has revolutionized genome editing methodologies [65].

4.3 Design of gene constructs for probiotic vaccines

Construction of recombinant proteins in probiotics needs meticulous design of various components, multiple cloning sites (MCS), promoters, and terminators. Heterologous expression systems used in engineered probiotics can be categorized into two major types of constitutive and inducible. Expression systems for heterologous proteins in engineered probiotics can vary significantly, using intracellular (e.g. Pcyt), secretion (e.g. pSEC), or cell wall anchoring (e.g. pCWA) mechanisms [66]. Common constitutive promoters derived from the L. lactis genomic library include P21, P23, and P59, which are characterized as strong promoters as well as P32 and P44 [67]. To lessen the overproduction of protein risk, there is a growing need for novel controlled-expression systems that act in response to external environmental factors such as temperature, bile salt concentration, pH, and antimicrobial peptides [68]. One of the most prevalent inducible systems used in probiotic engineering is the nisin-controlled gene expression (NICE) system, including key components of NisK, NisR, and NisA promoter (PnisA) [69]. In addition to the NICE system, other expression systems have been developed such as the pSIP system that controls quorum sensing mechanisms, using a series of pSIP-based vectors, and promoters of class II bacteriocins such as sakacin A and sakacin P [70]. Additionally, innovative strategies such as zinc-inducible P znZitR system, heat shock protein promoter-driven SICE, and auto-inducible P170 promoter from L. lactis have emerged for protein expression under specific conditions [66, 71, 72]. The available research demonstrates successful uses of suicide plasmids such as the pSA3-based vector pTRK327 and elements such as IS1223 for the targeted manipulation of genomes in various probiotic LAB. These proteins often use the Sec pathway for efficient secretion, particularly when proteins possess an N-terminal signal peptide. Furthermore, protein folding and secretion facilitators such as bacterial ffh genes are critical in ensuring appropriate protein maturation [73]. Engineering of inducible pathways in probiotics can enable them to sense and respond to specific signals from tumors or infected cells, prompting release of therapeutic factors.

4.4 The CRISPR/Cas technology for engineering probiotic vaccines

Genetic engineering, particularly in the realm of probiotics with the potential for edible vaccination [74]. Using CRISPR, scientists can modify probiotic strains to express desired antigens, optimize metabolic pathways, and improve stability of these microorganisms under various environmental conditions. This capacity for targeted genomic intervention not only accelerates strain development but also increases potential for creating vaccines that are effective and adaptable to evolving pathogens. Moreover, use of CRISPR technology can lead to the investigation of novel probiotic candidates that were previously unreachable through conventional methods. This innovation is particularly critical in an era characterized by the rapid emergence of infectious diseases, where traditional vaccine development strategies may fail for speed and specificity. Integration of genome editing tools enhances ability to carry out extensive host-pathogen interaction studies. By creating genetically modified probiotics, researchers can better understand mechanisms underlying immune responses, creating vaccines that not only target specific pathogens but also promote overall gut health. The potential to personalize probiotics based on individual genomic profiles represents another advantage that genome editing technologies can help researchers achieve [75]. Established efficiency and specificity of CRISPR-mediated editing tools facilitate precise gene insertion, deletion, and mutation across species such as E. coli, B. subtilis, and Lactobacillus spp., underscoring their versatility [76]. The CRISPR/Cas systems are present in approximately 50% of the bacterial populations, including a significant prevalence in Lactobacilli spp. and Bifidobacteria spp., investigation of these systems for probiotic engineering is well-supported by natural diversity [77]. The use of exogenous CRISPR/Cas systems through plasmid-based approaches has revolutionized the capability for genetic modification in probiotic strains, particularly in those lacking inherent CRISPR/Cas systems or showing limited efficacy [78].

Recent advancements suggest that the potential of CRISPR-based editing tools in LAB is far from that fully realized, particularly with the introduction of multiple-locus editing and Cas9 non-homologous end joining (NHEJ) repair mechanisms that have successfully been implemented in other bacteria such as E. coli and Mycobacterium tuberculosiss [79]. One promising achievement of research involves use of CRISPR-associated transposases (CAST) to integrate gene construct directly into chromosomal target sites without the need of positive selection, achieving integration frequencies of up to 80%. Inspired by these developments, harnessing endogenous CRISPR/Cas systems in LAB strains provides a robust framework for genetic manipulation [80]. For example, the subtype I-E CRISPR/Cas system present in L. crispatus has facilitated flexible efficient genetic engineering, while specific subtypes in other LAB species such as Pediococcus acidilactici and L. gasseri have been used to enhance lactic acid production and carry out promoter replacements [81]. Recent advancements show successful implementation of CRISPR/Cas assisted genome editing in various Lactobacillus. spp., including L. reuteri ATCC PTA 6475 and L. plantarum. Significantly, pLCNICK plasmid, developed by Song et al., uses the CRISPR/Cas9D10A system for rapid genome editing in L. casei, demonstrating broad host compatibility for other Lactobacillus strains [82]. Engineered L. plantarum WCFS1 for N-acetylglucosamine production and enhanced galactose fermentation capabilities of Brucella ATCC MYA-796, illustrate the potential for engineered probiotics to provide novel health benefits [83]. Integration of RecE/T improves homologous recombination; thereby, enhancing effectiveness of CRISPR/Cas9 technologies in strain engineering [60]. Additionally, engineered CRISPR/nCas9 systems have shown successful adaptation in various Lactobacillus spp., indicating the broad applicability of these technologies [84]. In other probiotic strains such as Streptococcus spp. and Clostridium spp., CRISPR systems have been used to modify native traits and enhance production capabilities by executing targeted genomic edits [85]. Use of CRISPR technology is expanding beyond Lactobacillus, with a growing number of studies focusing on various microorganisms, including Corynebacterium glutamicum, Candida parapsilosis, and E. coli [76]. Versatility of Cas9 nickase (nCas9) and dead Cas9 (dCas9) tools enables precise genome editing without inducing double-strand breaks, while Cas13 nucleases provide a unique mechanism for RNA cleavage, further expanding the toolkit available for microbial genetic engineering. One of the pioneering companies, Zbiotics, has emerged as the first to commercialize genetically engineered probiotic products. In addition to LAB, CRISPR/Cas systems have verified advantageous for yeasts such as S. cerevisiae and S. boulardii [86].

4.5 Defense mechanisms of bacteria against foreign DNA: implications for genome editing

Bacteria have developed sophisticated defense mechanisms to protect their genomic integrity from the invasion of foreign DNA. Two primary systems that exemplify this defensive strategy include restriction-modification systems and CRISPR/Cas systems. Over 90% of bacteria possess defense mechanisms such as restriction-modification systems, underscoring their importance in safeguarding microbial genomes against exogenous genetic materials. The CRISPR/Cas systems, detected in approximately 85.2% of archaea and 42.3% of bacteria, present another layer of complexity in the landscape of microbial defense against invasive DNAs [87]. Within the Lactobacillus genus that includes a significant representation of probiotics, occurrence of CRISPR/Cas systems is significantly high. The significance of these defense mechanisms becomes particularly evident in the context of genome editing techniques, which necessitate the successful integration of foreign DNA into bacterial genomes. To evade bacterial defenses, researchers have suggested three distinct strategies. First, using an intermediate host that boasts compatible methylation patterns, facilitating transformation processes. Second, using recombinant intermediate hosts that express the requisite methyltransferases anticipated in the target microorganism [57]. Third, another innovative approach involves in vitro incubation of foreign DNA with commercial methyltransferases to match the host's DNA methylation patterns, a technique that has significantly improved transformation efficiency in species such as L. plantarum. By effectively overcoming these bacterial defense mechanisms, researchers can enhance efficiency and reliability of the genetic modifications in bacteria .

methylation patterns, a technique that has significantly improved transformation efficiency in species such as L. plantarum. By effectively overcoming these bacterial defense mechanisms, researchers can enhance efficiency and reliability of the genetic modifications in bacteria [88]

  1. Improvement of Probiotic Efficacy Through Bioengineering

5.1. Enhancement of colonization exclusion

Increasing occurrence of pathogenic bacteria and their subsequent infections necessitates innovative strategies for prevention and management. One promising approach is the enhanced colonization of probiotics in the intestinal mucosal surface, which can create a competitive environment that limits colonization of harmful microorganisms. Genetic modification of adhesions, flagellins and fimbriae, help probiotics to serve as effective barriers to pathogen colonization in the GIT [89]. For example, the internalin A gene from L. monocytogenes expressed in the probiotic L. lactis, a recombinant L. paracasei strain that expressed the Listeria adhesion protein (LAP) suppresses the adhesion of L. monocytogenes [89]. Expressing the surface-associated flagellin of B. cereus in L. lactis, to competitive inhibition of pathogenic bacteria such as E. coli and Salmonella enterica from binding to intestinal epithelia [90].

 

5.2. A focus on stress tolerance

Numerous probiotic strains show limited tolerance to environmental stresses such as temperature, salinity and oxygen level. These limitations lessen stability and effectiveness of the probiotics as edible vaccines. Through genetic modification, researchers aim to enhance stress adaptation capabilities of the probiotics; thus, ensuring their viability and therapeutic efficacy [38]. A major key to this approach includes manipulation of heat-shock proteins, particularly GroES and GroEL, which are critical for the probiotic viability within various temperatures. A study by Desmond et al. demonstrated that overexpressing these chaperones in L. paracasei NFBC338 significantly improved its thermotolerance and solvent resistance, suggesting that strategic gene modifications can enhance robustness of the probiotic strains. Moreover, investigation of heterologous gene expression further contributes to probiotic resilience. For example, expression of the BetL gene in L. salivarius UCC118 through a nisin-controlled system demonstrated enhanced resistance to various environmental stresses [91]. Similarly, research involving Bifidobacterium breve UCC2003, which harbored the BetL gene, revealed an enhanced tolerance to gastric acidity and high osmolarity, indicating that genetic adaptations could significantly improve stress tolerance [92]. Additionally, cloning of the trehalose synthesis gene (ostAB) from E. coli into L. lactis has demonstrated improved survival rates for probiotics during freeze-drying processes and in high bile concentrations. These findings verify feasibility of using genes from pathogenic organisms to enhance stress resilience of the beneficial probiotics.

5.3. Receptor mimicry probiotic system

Understanding of the precise dynamics of toxin-receptor interactions on the surface of human intestinal cells provides a foundation for developing innovative strategies aimed to interrupting pathogen adhesion and disrupting pathogen; thereby, decreasing incidence and severity of infections [93]. One promising therapeutic approach involves expression of toxin receptors on the surface of probiotic strains to mimic the host's receptor environment. Such receptor mimics can produce lipopolysaccharides recognized by toxins such as cholera toxin or enterotoxigenic E. coli (ETEC) heat-labile toxin. Expression of Neisseria gonorrhoeae galactosyl-transferase genes in non-pathogenic E. coli strains resulted in a 100% efficacy rate in treating lethal infections caused by Shigatoxigenic E. coli (STEC) in mice [94]. Similarly, expression of a glycosyltransferase gene specific to meningococcal toxins in probiotic E. coli created a competitive environment that inhibited pathogen binding and hence subsequent infections [93]. Mimicking of host receptors on probiotic surfaces can serve as an effective strategy for preventing specific toxin-mediated pathologies [95]. Moreover, concept of disrupting pathogen virulence through innovative means such as quorum sensing manipulation has attracted intentions. As seen for E. coli Nissle 1917 strain producing cholera autoinducer 1 (CAI-1), interference with virulence gene expression upon reco-gnition of specific signaling molecules decreased colon-ization of Vibrio cholerae in experimental models [96].

5.4. The promising landscape of next-generation probiotics

The objective is to develop next-generation probiotics (NGPs) that offer enhanced therapeutic potentials, compared to those traditional probiotics. The integration of computational biology with genetic engineering is particularly advantageous, as it supports the design of next-generation synthetic LAB capable of overcoming challenges associated with industrial scale-up and complex biologics production. A relevant attempt in this field includes the Lactochassis project, which aims to create synthetic LAB, specifically for biomedical uses. This initiative underscores a paradigm shift in probiotic development, transitioning conventional probiotics from mere vectors for therapeutic delivery to engineered microbial ‘physicians’ that actively respond to health challenges [58]. Certain strains show significant promises due to their effects on health and disease. Good examples are Eubacterium hallii, Faecalibacterium prausnitzii, Roseburia spp., Akkermansia muciniphila, and Bacteroides fragilis. Research suggests that these microbes play vital roles in key physiological processes, including modulation of gastrointestinal immunity, enhancement of immunotherapy efficacy in cancer patients, maintenance of intestinal barrier integrity, and generation of beneficial metabolites, particularly short-chain fatty acids (SCFAs). Despite their promising potentials, widespread use of these NGPs produces significant problems. Key challenges include development of appropriate methods for culturing and storing these oxygen-sensitive organisms, which complicates their integration into consumer products [97]. As microbiome research evolves, efforts are made to genetically engineer or modify these NGPs, enabling them to target specific diseases or health conditions more effectively. Uses of this technology range from edible health to the management of allergies, metabolic disorders, cancers, and IBD. Engineered probiotics for edible vaccines in the treatment of diseases are represented in Table 1.

  1. Challenges Associated with Engin-eered Probiotics as Edible Vaccines

Advances in probiotic-based edible vaccine technol-ogy have reached the clinical stage [14]. However, there are still several problems in this way. Edible probiotic vaccines may decrease the immune response in populations with low hygiene and increases in the occurrence of illnesses associated with harmful microorganisms. These germs dominate the symbiotic community with other microbiota because of the weakened immune system of the human body and specific manifestation of the disease. Furthermore, variety in microbiota in individuals residing in a certain area includes a significant effect on the interactions with probiotics. Thus, assessing appropriate reactions to the edible probiotic vaccines becomes further challenging. Another critical concern includes health of the digestive system. Intestinal dysfunction can weaken the immune response. Moreover, idiopathic variables such as environmental enteropathy, a condition that damages intestinal health with no recognized origin, are reported. These factors are accountable for the diminished responses to vaccines [122].

In elderly people, cellular and humoral immune responses are weakened and these factors were accountable for decreasing vaccine protective efficacy. The next challenge includes non-colonizing nature of the probiotics, which needs daily treatment with high doses (approximately 100 times the injection dose) to have specific health benefits for the host, due to the variable pH values in various parts of the gut. Therefore, improvement of parameters linked to microorganisms and formulation should be addressed to overcome this challenge [30, 123]. In protein-based vaccinations, selecting an appropriate strain for protein expression is difficult because of the genetic variability linked to probiotic strains. Various species show distinct patterns of antigen expression, making it challenging to select the optimal pattern. One further concern includes the absence of post-translational processes in prokaryotes. For example, when the target antigen is overexpressed in the probiotic bacterial species E.coli, it leads to the formation of inclusion bodies (IBs). To achieve optimal effectiveness, these IBs need further processing to unfold and refold desired proteins to back to their original conditions. This is a separate process and needs extra financial resources. In addition, developing a reliable protocol for modifying proteins after their expression during production of vaccines is inherently difficult. This is because various post-translational mechanisms may be necessary depending on the desired structures and functional behaviors of the protein antigens [10]

Due to their inherent nature, protein-based edible probiotic vaccines must overcome physical-chemical barriers such as pH and bile salts, as well as biological barriers such as intestinal-epithelial barriers. To ensure effectiveness and safety of drug delivery, it is essential to carry out comprehensive studies on formulations to overcome difficulties associated with oral administration of medicines [124]. Plasmids are the predominant approach used in probiotic manipulation. Relatively, type of the probiotic genetic manipulation is important. Lifespan of the plasmids in probiotics is limited, leading to a gradual decrease in production of the desired protein. Hence, development of further appropriate alternatives is necessity to design probiotics and efficient desired antigen production. For plasmids, horizontal transfer of genetically modified genes and antibiotic resistance of the probiotics present significant hazards. Hence, it is critical to use conservation strategies with the progress of these GMOs. Probiotics engineering using novel genome editing tools provides a promising solution to overcome limitations and expanding their clinical uses. The CRISPR approach has been highlighted as an innovative technological tool that helps in development of novel and highly targeted edible vaccines. The quick progress and critical steps in CRISPR/Cas9 use include a wide range, converting it into a significant area of biotechnology. In addition to the significant knowledge achieved, several fundamental issues are still unresolved. The use of CRISPR technology includes substantial barriers that involve concerted efforts for resolution. It is important to address these challenges before the technology integrates into clinical settings. A forward-looking perspective urges a concerted focus on extensive research studies to thoroughly assess efficacy and safety of CRISPR-based treatment protocols, specifically for edible vaccines. Moreover, combining CRISPR with complementary technologies such as nanotechnology may further enhance effectiveness of the probiotics, enriching knowledge of edible vaccines, and opening new horizons for novel innovative solutions in immunization [125].

  1. Conclusion

Findings of this study demonstrate the potential of engineered probiotics as a revolutionary solution in immunization. Using advanced genetic tools such as CRISPR/Cas genome editing and synthetic biology, these modified microorganisms can act as carriers and producers of specific antigens, providing a novel approach to vaccine delivery. Probiotic-based oral vaccines include significant advantages such as targeted antigen delivery and enhancement of mucosal and systemic immune responses. Furthermore, these vaccines are affordable for the general public. The potential of engineering probiotics for preventive and therapeutic uses to change the prescription pattern from traditional injectable vaccines to oral alternatives makes their uses further customer-friendly and fit with modern health care needs. Despite these promising advances, several challenges are still addressed, including variations in immune responses in various individuals dependence on the health of the GIT, instability of the probiotics in various gastrointestinal environments, and possibility of the potential horizontal transfer of genetically modified genes. Addressing these challenges needs comprehensive studies to optimize probiotic formulations, including use of nanotechnology methods and ensuring their safety through advanced genetic techniques. This facilitates further sustainable, user-friendly immunization strategies globally that are fit with emerging public health demands.

  1. Acknowledgements

The authors acknowledge Shahid Beheshti University of Medical Sciences, School of Advanced Technologies in Medicine.

  1. Author Contributions

N.H., F.SH., E.SH. and M.BV. wrote the manuscript and F.SH. and E.SH. revised the manuscript.

  1. Conflict of Interest

The authors declare no conflict of interest.

References

  1. 1.Idrees M, Imran M, Atiq N, Zahra R, Abid R, Alreshidi M, Roberts T, Abdelgadir A, Tipu MK, Farid A: Probiotics, their action modality and the use of multi-omics in metamorphosis of commensal microbiota into target-based probiotics. Front Nutr. 2022, 9:959941.

https://doi.org/10.3389/fnut.2022.959941

2    Saleh A, Qamar S, Tekin A, Singh R, Kashyap R: Vaccine development throughout history. Cureus 2021, 13[7]..

https://doi.org/10.7759/cureus.16635

3    Esmael H, Hirpa E: Review on edible vaccine. Acad J Nutr. 2015, 4[1]:40-49

.https:// doi.org/10.5829/idosi.ajn.2015.4.1.956

  1. Foroutan NS, Tabandeh F, Khodabandeh M, Mojgani N, Maghsoudi A, Moradi M: Isolation and Identification of an Indigenous Probiotic Lactobacillus Strain: Its Encapsulation with Natural Branched Polysaccharids to Improve Bacterial Viability. Appl Food Biotechnol. 2017, 4[3]:133-142..

https://doi.org/10.22037/afb.v4i3.16471

  1. Fong FLY, Shah NP, Kirjavainen P, El-Nezami H: Mechanism of action of probiotic bacteria on intestinal and systemic immunities and antigen-presenting cells. Int Rev Immunol. 2016, 35[3]:179-188.

https://doi.org/10.3109/08830185.2015.1096937

  1. La Fata G, Weber P, Mohajeri MH: Probiotics and the gut immune system: indirect regulation. Probiotics Antimicrob Proteins. 2018, 10:11-21.

https://doi.org/10.1007/s12602-017-9322-6

  1. Dixit S, Kumar A, Srinivasan K, Vincent PDR, Ramu Krishnan N: Advancing genome editing with artificial intelligence: opportunities, challenges and future directions. Front Bioeng Biotechnol. 2024, 11:1335901.

https://doi.org/10.3389/fbioe.2023.1335901

8    Sautto GA, Kirchenbaum GA, Diotti RA, Criscuolo E, Ferrara F: Next generation vaccines for infectious diseases. J Immunol Res. 2019;2019:5890962.

https://doi.org/10.1155/2019/5890962

  1. Criscuolo E, Caputo V, Diotti R, Sautto G, Kirchenbaum G, Clementi N: Alternative methods of vaccine delivery: an overview of edible and intradermal vaccines. J Immunol Res. 2019;2019:8303648.

https://doi.org/10.1155/2019/8303648

  1. Rosales-Mendoza S, Angulo C, Meza B: Food-grade organisms as vaccine biofactories and oral delivery vehicles. Trends Biotechnol. 2016, 34[2]:124-136.

https://doi.org/10.1016/j.tibtech.2015.11.007

  1. O'Hagan DT: Microparticles and polymers for the mucosal delivery of vaccines. Adv Drug Deliv Rev. 1998, 34[2-3]:305-320.

https://doi.org/10.1016/s0169-409x[98]00045-3

  1. Oh J-H, van Pijkeren J-P: CRISPR–Cas9-assisted recombineering in Lactobacillus reuteri. Nucleic Acids Res. 2014, 42[17]:e131-e131.

https://doi.org/10.1093/nar/gku623

  1. Jiang B, Li Z, Ou B, Duan Q, Zhu G: Targeting ideal oral vaccine vectors based on probiotics: a systematical view. Appl Microbiol Biotechnol. 2019, 103:3941-3953.

https://doi.org/10.1007/s00253-019-09770-7

  1. LeCureux JS, Dean GA: Lactobacillus mucosal vaccine vectors: immune responses against bacterial and viral antigens. Msphere 2018, 3[3]:10.1128/msphere. 00061-00018.

https://doi.org/10.1128/mSphere.00061-18

  1. MacDonald TT, Bell I: Probiotics and the immune response to vaccines. Proc Nutr Soc. 2010, 69[3]:442-446.

https://doi.org/10.1017/S0029665110001758

  1. Alimolaei M, Golchin M, Ezatkhah M: Orally administered recombinant Lactobacillus casei vector vaccine expressing β-toxoid of Clostridium perfringens that induced protective immunity responses. Res Vet Sci. 2017, 115:332-339.

https://doi.org/10.1016/j.rvsc.2017.06.018

17  Amuguni H, Tzipori S: Bacillus subtilis: a temperature resistant and needle free delivery system of immunogens. Hum Vaccin Immunother. 2012, 8[7]:979-986

https://doi.org/10.4161/hv.20694

  1. Al Kassaa I, AL KASSAA I: Antiviral probiotics: a new concept in medical sciences. New insights on antiviral probiotics: From research to applications. Springer, Cham. 2017:1-46.

https://doi.org/10.1007/978-3-319-49688-7_1

  1. Zhang W, Azevedo MS, Wen K, Gonzalez A, Saif LJ, Li G, Yousef AE, Yuan L: Probiotic Lactobacillus acidophilus enhances the immunogenicity of an oral rotavirus vaccine in gnotobiotic pigs. Vaccine. 2008, 26[29-30]:3655-3661.

https://doi.org/10.1016/j.vaccine.2008.04.070

  1. van Baarlen P, Wells JM, Kleerebezem M: Regulation of intestinal homeostasis and immunity with probiotic lactobacilli. Trends Immunol. 2013, 34[5]:208-215.

https://doi.org/10.1016/j.it.2013.01.005

21  Ramirez JEV, Sharpe LA, Peppas NA: Current state and challenges in developing oral vaccines. Adv Drug Deliv Rev. 2017, 114:116-131.

https://doi.org/10.1016/j.addr.2017.04.008

  1. Allaire JM, Crowley SM, Law HT, Chang S-Y, Ko H-J, Vallance BA: The intestinal epithelium: central coordinator of mucosal immunity. Trends Immunol. 2018, 39[9]:677-696.

https://doi.org/10.1016/j.it.2018.04.002

  1. Tokuhara D, Kurashima Y, Kamioka M, Nakayama T, Ernst P, Kiyono H: A comprehensive understanding of the gut mucosal immune system in allergic inflammation. Allergol Int. 2019, 68[1]:17-25.

https://doi.org/10.1016/j.alit.2018.09.004

  1. Owen JL, Sahay B, Mohamadzadeh M: New generation of oral mucosal vaccines targeting dendritic cells. Curr Opin Chem Biol. 2013, 17[6]:918-924.

https://doi.org/10.1016/j.cbpa.2013.06.013

  1. Barhate G, Gautam M, Gairola S, Jadhav S, Pokharkar V: Enhanced mucosal immune responses against tetanus toxoid using novel delivery system comprised of chitosan functionalized gold nanoparticles and botanical adjuvant: Characterization, immunogenicity and stability assessment. J Pharm Sci 2014, 103[11]:3448-3456.

https://doi.org/10.1002/jps.24161

  1. Woodrow KA, Bennett KM, Lo DD: Mucosal vaccine design and delivery. Annu Rev Biomed Eng. 2012, 14[1]:17-46.

https://doi.org/10.1146/annurev-bioeng-071811-150054

  1. Hu Q, Wu M, Fang C, Cheng C, Zhao M, Fang W, Chu PK, Ping Y, Tang G: Engineering nanoparticle-coated bacteria as oral DNA vaccines for cancer immunotherapy. Nano Lett. 2015, 15[4]:2732-2739.

https://doi.org/10.1021/acs.nanolett.5b00570

  1. Van Overtvelt L, Moussu H, Horiot S, Samson S, Lombardi V, Mascarell L, Van de Moer A, Bourdet-Sicard R, Moingeon P: Lactic acid bacteria as adjuvants for sublingual allergy vaccines. Vaccine. 2010, 28[17]:2986-2992.

https://doi.org/10.1016/j.vaccine.2010.02.009

  1. Singh M: Novel immune potentiators and delivery technologies for next generation vaccines: Springer Science & Business Media; 2013.

https://doi.org/10.1007/978-1-4614-5380-2

  1. Govender M, Choonara YE, Kumar P, du Toit LC, van Vuuren S, Pillay V: A review of the advancements in probiotic delivery: Conventional vs. non-conventional formulations for intestinal flora supplementation. Aaps PharmSciTech 2014, 15:29-43.

https://doi.org/10.1208/s12249-013-0027-1

  1. Huyghebaert N, Vermeire A, Neirynck S, Steidler L, Remaut E, Remon JP: Development of an enteric-coated formulation containing freeze-dried, viable recombinant Lactococcus lactis for the ileal mucosal delivery of human interleukin-10. Eur J Pharm Biopharm. 2005, 60[3]:349-359.

https://doi.org/10.1016/j.ejpb.2005.02.012

  1. van Roosmalen ML, Kanninga R, El Khattabi M, Neef J, Audouy S, Bosma T, Kuipers A, Post E, Steen A, Kok J: Mucosal vaccine delivery of antigens tightly bound to an adjuvant particle made from food-grade bacteria. Methods 2006, 38[2]:144-149.

https://doi.org/10.1016/j.ymeth.2005.09.015

  1. Hag S, Poondla N: Genetically engineered probiotics. Probiotic Research in Therapeutics: Volume 1: Applications in Cancers and Immunological Diseases 2021:295-328.

https://doi.org/10.1007/978-981-15-8214-1_16

  1. Bravo D, Landete JM: Genetic engineering as a powerful tool to improve probiotic strains. Biotechnol Genet Eng Rev. 2017, 33[2]:173-189.

https://doi.org/10.1080/02648725.2017.1408257

  1. Yadav R, Kumar V, Baweja M, Shukla P: Gene editing and genetic engineering approaches for advanced probiotics: a review. Crit Rev Food Sci Nutr. 2018, 58[10]:1735-1746.

https://doi.org/10.1080/10408398.2016.1274877

  1. Gupta RM, Musunuru K: Expanding the genetic editing tool kit: ZFNs, TALENs and CRISPR-Cas9. J Clin Invest. 2014, 124[10]:4154-4161.

https://doi.org/10.1172/JCI72992

  1. Braff D, Shis D, Collins JJ: Synthetic biology platform technologies for antimicrobial applications. Adv Drug Deliv Rev. 2016, 105:35-43.

https://doi.org/10.1016/j.addr.2016.04.006

  1. Mathipa MG, Thantsha MS: Probiotic engineering: towards development of robust probiotic strains with enhanced functional properties and for targeted control of enteric pathogens. Gut Pathog. 2017, 9:1-17.

https://doi.org/10.1186/s13099-017-0178-9

  1. Zuo F, Chen S, Marcotte H: Engineer probiotic bifidobacteria for food and biomedical applications-Current status and future prospective. Biotechnol Adv. 2020, 45:107654.

https://doi.org/10.1016/j.biotechadv.2020.107654

  1. Wells J, Robinson K, Chamberlain L, Schofield K, Le Page R: Lactic acid bacteria as vaccine delivery vehicles. Antonie Van Leeuwenhoek. 1996, 70:317-330.

https://doi.org/10.1007/BF00395939

  1. Le Loir Y, Azevedo V, Oliveira SC, Freitas DA, Miyoshi A, Bermúdez-Humarán LG, Nouaille S, Ribeiro LA, Leclercq S, Gabriel JE: Protein secretion in Lactococcus lactis: an efficient way to increase the overall heterologous protein production. Microb Cell Fact. 2005, 4:1-13.

https://doi.org/10.1186/1475-2859-4-2

  1. Wells J: Mucosal vaccination and therapy with genetically modified lactic acid bacteria. Annu Rev Food Sci Technol. 2011, 2[1]:423-445.

https://doi.org/10.1146/annurev-food-022510-133640

  1. Chen H, Ji H, Kong X, Lei P, Yang Q, Wu W, Jin L, Sun D: Bacterial ghosts-based vaccine and drug delivery systems. Pharmaceutics. 2021, 13[11]:1892.

https://doi.org/10.3390/pharmaceutics13111892

  1. Xu Y, Yuen P-W, Lam JK-W: Intranasal DNA vaccine for protection against respiratory infectious diseases: the delivery perspectives. Pharmaceutics. 2014, 6[3]:378-415.

https://doi.org/10.3390/pharmaceutics6030378

  1. Stein E, Inic-Kanada A, Belij S, Montanaro J, Bintner N, Schlacher S, Mayr UB, Lubitz W, Stojanovic M, Najdenski H: In vitro and in vivo uptake study of Escherichia coli Nissle 1917 bacterial ghosts: cell-based delivery system to target ocular surface diseases. Invest Ophthalmol Vis Sci. 2013, 54[9]:6326-6333.

https://doi.org/10.1167/iovs.13-12044

  1. Drolia R, Amalaradjou MAR, Ryan V, Tenguria S, Liu D, Bai X, Xu L, Singh AK, Cox AD, Bernal-Crespo V: Receptor-targeted engineered probiotics mitigate lethal Listeria infection. Nat Commun. 2020, 11[1]:6344.

https://doi.org/10.1038/s41467-020-20200-5

  1. Wu J, Xin Y, Kong J, Guo T: Genetic tools for the development of recombinant lactic acid bacteria. Microb Cell Fact. 2021, 20[1]:118.

https://doi.org/10.1186/s12934-021-01607-1

  1. Fujiki M, Kaczmarczyk JF, Yusibov V, Rabindran S: Development of a new cucumber mosaic virus-based plant expression vector with truncated 3a movement protein. Virology. 2008, 381[1]:136-142.

https://doi.org/10.1016/j.virol.2008.08.022

  1. Jiji MG, Ninan MA, Thomas V, Thomas BT: Edible microalgae: potential candidate for developing edible vaccines. Vegetos. 2023:1-6.

https://doi.org/10.1007/s42535-023-00636-y

  1. Gangl D, Zedler JA, Rajakumar PD, Martinez EMR, Riseley A, Włodarczyk A, Purton S, Sakuragi Y, Howe CJ, Jensen PE: Biotechnological exploitation of microalgae. J Exp Bot. 2015, 66[22]:6975-6990.

https://doi.org/10.1093/jxb/erv426

  1. Gregory JA, Li F, Tomosada LM, Cox CJ, Topol AB, Vinetz JM, Mayfield S: Algae-produced Pfs25 elicits antibodies that inhibit malaria transmission. PloS one 2012, 7[5]:e37179.

https://doi.org/10.1371/journal.pone.0037179

  1. Silveira MM, Moreira GMSG, Mendonça M: DNA vaccines against COVID-19: Perspectives and challenges. Life Sci. 2021, 267:118919.

https://doi.org/10.1016/j.lfs.2020.118919

  1. Braat H, Rottiers P, Hommes DW, Huyghebaert N, Remaut E, Remon JP, Van Deventer SJ, Neirynck S, Peppelenbosch MP, Steidler L: A phase I trial with transgenic bacteria expressing interleukin-10 in Crohn’s disease. Clin Gastroenterol Hepatol. 2006, 4[6]:754-759.

https://doi.org/10.1016/j.cgh.2006.03.028

  1. Mathieu C, Wiedeman A, Cerosaletti K, Long SA, Serti E, Cooney L, Vermeiren J, Caluwaerts S, Van Huynegem K, Steidler L: A first-in-human, open-label Phase 1b and a randomised, double-blind Phase 2a clinical trial in recent-onset type 1 diabetes with AG019 as monotherapy and in combination with teplizumab. Diabetologia. 2024, 67[1]:27-41.

https://doi.org/10.1007/s00125-023-06014-2

  1. Komatsu A, Igimi S, Kawana K: Optimization of human papillomavirus [HPV] type 16 E7-expressing lactobacillus-based vaccine for induction of mucosal E7-specific IFNγ-producing cells. Vaccine. 2018, 36[24]:3423-3426.

https://doi.org/10.1016/j.vaccine.2018.05.009

  1. Sirima SB, Mordmüller B, Milligan P, Ngoa UA, Kironde F, Atuguba F, Tiono AB, Issifou S, Kaddumukasa M, Bangre O: A phase 2b randomized, controlled trial of the efficacy of the GMZ2 malaria vaccine in African children. Vaccine. 2016, 34[38]:4536-4542.

https://doi.org/10.1016/j.vaccine.2016.07.041

  1. Mugwanda K, Hamese S, Van Zyl WF, Prinsloo E, Du Plessis M, Dicks LM, Thimiri Govinda Raj DB: Recent advances in genetic tools for engineering probiotic lactic acid bacteria. Biosci Rep. 2023, 43[1]:BSR20211299.

https://doi.org/10.1042/BSR20211299

  1. Romero-Luna HE, Hernández-Mendoza A, González-Córdova AF, Peredo-Lovillo A: Bioactive peptides produced by engineered probiotics and other food-grade bacteria: A review. Food Chem X. 2022, 13:100196.

https://doi.org/10.1016/j.fochx.2021.100196

  1. Yang P, Wang J, Qi Q: Prophage recombinases-mediated genome engineering in Lactobacillus plantarum. Microb Cell Fact. 2015, 14:1-11.

https://doi.org/10.1186/s12934-015-0344-z

  1. 60. Huang H, Song X, Yang S: Development of a RecE/T-assisted CRISPR–Cas9 toolbox for Lactobacillus. Biotechnol J. 2019, 14[7]:1800690.

https://doi.org/10.1002/biot.201800690

  1. 61. Kim T-G, Yang M-S: Current trends in edible vaccine development using transgenic plants. Biotechnol Bioproc. 2010, 15:61-65.

https://doi.org/10.1007/s12257-009-3084-2

  1. 62. Kurup VM, Thomas J: Edible vaccines: promises and challenges. Mol Biotechnol. 2020, 62[2]:79-90.

https://doi.org/10.1007/s12033-019-00222-1

  1. 63. William S: A review of the progression of transgenic plants used to produce plant bodies for human usage. J Young Invest. 2002;4:56–61.
  2. 64. Börner RA, Kandasamy V, Axelsen AM, Nielsen AT, Bosma EF: Genome editing of lactic acid bacteria: opportunities for food, feed, pharma and FEMS Microbiol Lett. 2019, 366[1]:fny291.

https://doi.org/10.1093/femsle/fny291

  1. Van Zyl WF, Dicks LM, Deane SM: Development of a novel selection/counter-selection system for chromosomal gene integrations and deletions in lactic acid bacteria. BMC Mol Biol. 2019, 20:1-16.

https://doi.org/10.1186/s12867-019-0127-x

  1. 66. Levit R, Cortes-Perez NG, de Moreno de Leblanc A, Loiseau J, Aucouturier A, Langella P, LeBlanc JG, Bermúdez-Humarán LG: Use of genetically modified lactic acid bacteria and bifidobacteria as live delivery vectors for human and animal health. Gut Microbes 2022, 14[1]:2110821.

https://doi.org/10.1080/19490976.2022.2110821

  1. Morello E, Bermudez-Humaran L, Llull D, Sole V, Miraglio N, Langella P, Poquet I: Lactococcus lactis, an efficient cell factory for recombinant protein production and secretion. J Mol Microbiol Biotechnol. 2008, 14[1-3]:48-58.

https://doi.org/10.1159/000106082

  1. Benbouziane B, Ribelles P, Aubry C, Martin R, Kharrat P, Riazi A, Langella P, Bermúdez-Humarán LG: Development of a Stress-Inducible Controlled Expression [SICE] system in Lactococcus lactis for the production and delivery of therapeutic molecules at mucosal surfaces. J Biotechnol. 2013, 168[2]:120-129.

https://doi.org/10.1016/j.jbiotec.2013.04.019

  1. Mierau I, Kleerebezem M: 10 years of the nisin-controlled gene expression system [NICE] in Lactococcus lactis. Appl Microbiol Biotechnol. 2005, 68[6]:705-717.

https://doi.org/10.1007/s00253-005-0107-6

  1. 70. Nguyen T-T, Nguyen T-H, Maischberger T, Schmelzer P, Mathiesen G, Eijsink VG, Haltrich D, Peterbauer CK: Quantitative transcript analysis of the inducible expression system pSIP: comparison of the overexpression of Lactobacillus spp. β-galactosidases in Lactobacillus plantarum. Microb Cell Fact. 2011, 10:1-10.

https://doi.org/10.1186/1475-2859-10-46

  1. 71. Cho SW, Yim J, Seo SW: Engineering tools for the development of recombinant lactic acid bacteria. Biotechnol J. 2020, 15[6]:1900344.

https://doi.org/10.1002/biot.201900344

  1. 72. Tavares LM, De Jesus LC, Da Silva TF, Barroso FA, Batista VL, Coelho-Rocha ND, Azevedo V, Drumond MM, Mancha-Agresti P: Novel strategies for efficient production and delivery of live biotherapeutics and biotechnological uses of Lactococcus lactis: the lactic acid bacterium model. Front Bioeng Biotechnol. 2020, 8:517166.

https://doi.org/10.3389/fbioe.2020.517166

  1. 73. Yin J-Y, Guo C-Q, Wang Z, Yu M-L, Gao S, Bukhari SM, Tang L-J, Xu Y-G, Li Y-J: Directed chromosomal integration and expression of porcine rotavirus outer capsid protein VP 4 in Lactobacillus casei ATCC393. Appl Microbiol Biotechnol. 2016, 100:9593-9604.

https://doi.org/10.1007/s00253-016-7779-y

  1. 74. Goh YJ, Barrangou R: Harnessing CRISPR-Cas systems for precision engineering of designer probiotic lactobacilli. Curr Opin Biotechnol. 2019, 56:163-171.

https://doi.org/10.1016/j.copbio.2018.11.009

  1. Liu L, Helal SE, Peng N: CRISPR-Cas-based engineering of probiotics. Biodes Res. 2023, 5:0017.

https://doi.org/10.34133/bdr.0017

  1. Mougiakos I, Bosma EF, de Vos WM, van Kranenburg R, van der Oost J: Next generation prokaryotic engineering: the CRISPR-Cas toolkit. Trends Biotechnol. 2016, 34[7]:575-587.

https://doi.org/10.1016/j.tibtech.2016.02.004

  1. Westra ER, Van Houte S, Gandon S, Whitaker R: The ecology and evolution of microbial CRISPR-Cas adaptive immune systems. Philos Trans R Soc Lond B Biol Sci. 2019;374[1772]:20190101.

https://doi.org/10.1098/rstb.2019.0101

  1. Pan M, Nethery MA, Hidalgo-Cantabrana C, Barrangou R: Comprehensive mining and characterization of CRISPR-Cas systems in Bifidobacterium. Microorganisms. 2020, 8[5]:720.

https://doi.org/10.3390/microorganisms8050720

  1. 79. Jiang Y, Chen B, Duan C, Sun B, Yang J, Yang S: Multigene editing in the Escherichia coli genome via the CRISPR-Cas9 system. Appl Environ Microbiol. 2015, 81[7]:2506-2514.

https://doi.org/10.1128/AEM.04023-14

  1. 80. Klompe SE, Vo PL, Halpin-Healy TS, Sternberg SH: Transposon-encoded CRISPR–Cas systems direct RNA-guided DNA integration. Nature. 2019, 571[7764]:219-225.

https://doi.org/10.1038/s41586-019-1323-z

  1. 81. Liu L, Yang D, Zhang Z, Liu T, Hu G, He M, Zhao S, Peng N: High-efficiency genome editing based on endogenous CRISPR-Cas system enhances cell growth and lactic acid production in Pediococcus acidilactici. Appl Environ Microbiol. 2021, 87[20]:e00948-00921.

https://doi.org/10.1128/AEM.00948-21

  1. 82. Song X, Huang H, Xiong Z, Ai L, Yang S: CRISPR-Cas9D10A nickase-assisted genome editing in Lactobacillus casei. Appl Environ Microbiol. 2017, 83[22]:e01259-01217.

https://doi.org/10.1128/AEM.01259-17

  1. 83. Leenay RT, Vento JM, Shah M, Martino ME, Leulier F, Beisel CL: Genome editing with CRISPR‐Cas9 in Lactobacillus plantarum revealed that editing outcomes can vary across strains and between methods. Biotechnol J. 2019, 14[3]:1700583.

https://doi.org/10.1002/biot.201700583

  1. 84. Goh YJ, Barrangou R: Portable CRISPR-Cas9N system for flexible genome engineering in Lactobacillus acidophilus, Lactobacillus gasseri and Lactobacillus paracasei. Appl Environ Microbiol. 2021, 87[6]:e02669-02620.

https://doi.org/10.1128/AEM.02669-20

  1. 85. Zhang J, Zong W, Hong W, Zhang Z-T, Wang Y: Exploiting endogenous CRISPR-Cas system for multiplex genome editing in Clostridium tyrobutyricum and engineer the strain for high-level butanol production. Metab Eng. 2018, 47:49-59.

https://doi.org/10.1016/j.ymben.2018.03.007

  1. 86. Zhou D, Jiang Z, Pang Q, Zhu Y, Wang Q, Qi Q: CRISPR/Cas9-assisted seamless genome editing in Lactobacillus plantarum and its application in N-acetylglucosamine production. Appl Environ Microbiol. 2019, 85[21]:e01367-01319.

https://doi.org/10.1128/AEM.01367-19

  1. 87. Plagens A, Richter H, Charpentier E, Randau L: DNA and RNA interference mechanisms by CRISPR-Cas surveillance complexes. FEMS Microbiol Rev. 2015, 39[3]:442-463.

https://doi.org/10.1093/femsre/fuv019

  1. 88. Bober JR, Beisel CL, Nair NU: Synthetic biology approaches to engineer probiotics and members of the human microbiota for biomedical applications. Annu Rev Biomed Eng. 2018, 20[1]:277-300.

https://doi.org/10.1146/annurev-bioeng-062117-121019

  1. 89. Ramarao N, Lereclus D: Adhesion and cytotoxicity of Bacillus cereus and Bacillus thuringiensis to epithelial cells are FlhA and PlcR dependent, respectively. Microbes Infect. 2006, 8[6]:1483-1491.

https://doi.org/10.1016/j.micinf.2006.01.005

  1. Ascón MA, Ochoa-Repáraz J, Walters N, Pascual DW: Partially assembled K99 fimbriae are required for protection. Infect Immun. 2005, 73[11]:7274-7280.

https://doi.org/10.1128/IAI.73.11.7274-7280.2005

  1. Sheehan VM, Sleator RD, Fitzgerald GF, Hill C: Heterologous expression of BetL, a betaine uptake system, enhances the stress tolerance of Lactobacillus salivarius UCC118. Appl Environ Microbiol. 2006, 72[3]:2170-2177.

https://doi.org/10.1128/AEM.72.3.2170-2177.2006

92  Sheehan VM, Sleator RD, Hill C, Fitzgerald GF: Improving gastric transit, gastrointestinal persistence and therapeutic efficacy of the probiotic strain Bifidobacterium breve UCC2003. Microbiology. 2007, 153[10]:3563-3571.

https://doi.org/10.1099/mic.0.2007/006510-0

  1. Paton AW, Morona R, Paton JC: Designer probiotics for prevention of enteric infections. Nat Rev Microbiol. 2006, 4[3]:193-200.

https://doi.org/10.1038/nrmicro1349

  1. 94. Paton AW, Morona R, Paton JC: A new biological agent for treatment of Shiga toxigenic Escherichia coli infections and dysentery in humans. Nat Med. 2000, 6[3]:265-270.

https://doi.org/10.1038/73111

  1. 95. Focareta A, Paton JC, Morona R, Cook J, Paton AW: A recombinant probiotic for treatment and prevention of cholera. Gastroenterology. 2006, 130[6]:1688-1695..

https://doi.org/10.1053/j.gastro.2006.02.005

  1. 96. Duan F, March JC: Engineered bacterial communication prevents Vibrio cholerae virulence in an infant mouse model. Proceedings of the National Academy of Sciences Proc Natl Acad Sci U S A. 2010, 107[25]:11260-11264.

https://doi.org/10.1073/pnas.1001294107

  1. 97. Lopez-Siles M, Khan TM, Duncan SH, Harmsen HJ, Garcia-Gil LJ, Flint HJ: Cultured representatives of two major phylogroups of human colonic Faecalibacterium prausnitzii can utilize pectin, uronic acids and host-derived substrates for growth. Appl Environ Microbiol. 2012, 78[2]:420-428.

https://doi.org/10.1128/AEM.06858-11

  1. 98. Shirai M, Pendleton CD, Ahlers J, Takeshita T, Newman M, Berzofsky JA: Helper-cytotoxic T lymphocyte [CTL] determinant linkage required for priming of anti-HIV CD8+ CTL in vivo with peptide vaccine constructs. J Immunol. 1994;152[2]:549-556.
  2. 99. Oggioni MR, Medaglini D, Romano L, Peruzzi F, Maggi T, Lozzi L, Bracci L, Zazzi M, Manca F, Valensin PE: Antigenicity and immunogenicity of the V3 domain of HIV type 1 glycoprotein 120 expressed on the surface of Streptococcus gordonii. AIDS Res Hum Retroviruses. 1999, 15[5]:451-459.

https://doi.org/10.1089/088922299311204

  1. 100. Zegers N, Kluter E, van Der Stap H, Van Dura E, Van Dalen P, Shaw M and, Baillie L: Expression of the protective antigen of Bacillus anthracis by Lactobacillus casei: towards the development of an oral vaccine against anthrax. J Appl Microbiol. 1999, 87[2]:309-314.

https://doi.org/10.1046/j.1365-2672.1999.00900.x

  1. 101. Villena J, Medina M, Racedo S, Alvarez S: Resistance of Young Mice to Pneumococcal Infection can be Improved by Oral Vaccination with Recombinant Lactococcus lactis. J Microbiol Immunol Infect. 2010, 43[1]:1-10.

https://doi.org/10.1016/S1684-1182[10]60001-1

  1. 102. Buddenborg C, Daudel D, Liebrecht S, Greune L, Humberg V, Schmidt MA: Development of a tripartite vector system for live oral immunization using a gram-negative probiotic carrier. Int J Med Microbiol. 2008, 298[1-2]:105-114.

https://doi.org/10.1016/j.ijmm.2007.08.008

  1. 103. Lee SF: Oral colonization and immune responses to Streptococcus gordonii: potential use as a vector to induce antibodies against respiratory pathogens. Curr Opin Infect Dis. 2003, 16[3]:231-235.

https://doi.org/10.1097/00001432-200306000-00008

  1. 104. Wang M, Fu T, Hao J, Li L, Tian M, Jin N, Ren L, Li C: A recombinant Lactobacillus plantarum strain expressing the spike protein of SARS-CoV-2. Int J Biol Macromol. 2020 160:736-740.

https://doi.org/10.1016/j.ijbiomac.2020.05.239

  1. 105. Fredriksen L, Mathiesen G, Sioud M, Eijsink VG: Cell wall anchoring of the 37-kilodalton oncofetal antigen by Lactobacillus plantarum for mucosal cancer vaccine delivery. Appl Environ Microbiol. 2010, 76[21]:7359-7362.

https://doi.org/10.1128/AEM.01031-10

  1. 106. Agarwal P, Khatri P, Billack B, Low W-K, Shao J: Oral delivery of glucagon like peptide-1 by a recombinant Lactococcus lactis. Pharm Res. 2014, 31:3404-3414.

https://doi.org/10.1007/s11095-014-1430-3

  1. 107. Durrer KE, Allen MS, Hunt von Herbing I: Genetically engineered probiotic for the treatment of phenylketonuria [PKU]; assessment of a novel treatment in vitro and in the PAHenu2 mouse model of PKU. PloS one 2017, 12[5]:e0176286.

https://doi.org/10.1371/journal.pone.0176286

  1. 108. Wong CC, Zhang L, Wu WK, Shen J, Chan RL, Lu L, Hu W, Li MX, Li LF, Ren SX: Cathelicidin-encoding Lactococcus lactis promotes mucosal repair in murine experimental colitis. J Gastroenterol Hepatol. 2017, 32[3]:609-619.

https://doi.org/10.1111/jgh.13499

  1. 109. Jing H, Yong L, Haiyan L, Yanjun M, Yun X, Yu Z, Taiming L, Rongyue C, Liang J, Jie W: Oral administration of Lactococcus lactis delivered heat shock protein 65 attenuates atherosclerosis in low-density lipoprotein receptor-deficient mice. Vaccine 2011, 29[24]:4102-4109.

https://doi.org/10.1016/j.vaccine.2011.03.105

  1. 110. Wang X, Chen W, Tian Y, Mao Q, Lv X, Shang M, Li X, Yu X, Huang Y: Surface display of Clonorchis sinensis enolase on Bacillus subtilis spores potentializes an oral vaccine candidate. Vaccine 2014, 32[12]:1338-1345.

https://doi.org/10.1016/j.vaccine.2014.01.039

  1. 111. Chen T, Tian P, Huang Z, Zhao X, Wang H, Xia C, Wang L, Wei H: Engineered commensal bacteria prevent systemic inflammation-induced memory impairment and amyloidogenesis via producing GLP-1. Appl Microbiol Biotechnol. 2018, 102:7565-7575.

https://doi.org/10.1007/s00253-018-9155-6

  1. 112. Hwang IY, Koh E, Wong A, March JC, Bentley WE, Lee YS, Chang MW: Engineered probiotic Escherichia coli can eliminate and prevent Pseudomonas aeruginosa gut infection in animal models. Nat Commun. 2017, 8[1]:15028.

https://doi.org/10.1038/ncomms15028

  1. 113. Scott BM, Gutierrez-Vazquez C, Sanmarco LM, da Silva Pereira JA, Li Z, Plasencia A, Hewson P, Cox LM, O’Brien M, Chen SK: Self-tunable engineered yeast probiotics for the treatment of inflammatory bowel disease. Nat Med. 2021, 27[7]:1212-1222.

https://doi.org/10.1038/s41591-021-01390-x

  1. 114. Zhang H, Chen B, Wang Z, Peng K, Liu Y, Wang Z: Resensitizing tigecycline-and colistin-resistant Escherichia coli using an engineered conjugative CRISPR/Cas9 system. Microbiol Spectr. 2024, 12[4]:e03884-03823.

https://doi.org/10.1128/spectrum.03884-23

  1. 115. Neil K, Allard N, Roy P, Grenier F, Menendez A, Burrus V, Rodrigue S: High-efficiency delivery of CRISPR-Cas9 by engineered probiotics enables precise microbiome editing. Mol Syst Biol. 2021, 17[10]:e10335.

https://doi.org/10.15252/msb.202110335

  1. 116. Ou B, Jiang B, Jin D, Yang Y, Zhang M, Zhang D, Zhao H, Xu M, Song H, Wu W: Engineered recombinant Escherichia coli probiotic strains integrated with F4 and F18 fimbriae cluster genes in the chromosome and their assessment of immunogenic efficacy in vivo. ACS Synth Biol .2020, 9[2]:412-426.

https://doi.org/10.1021/acssynbio.9b00430

  1. 117. Wang L, Cheng X, Bai L, Gao M, Kang G, Cao X, Huang H: Positive interventional effect of engineered butyrate-producing bacteria on metabolic disorders and intestinal flora disruption in obese mice. Microbiol Spectr. 2022, 10[2]:e01147-01121.

https://doi.org/10.1128/spectrum.01147-21

  1. 118. Ninyio N, Schmitt K, Sergon G, Nilsson C andersson S, Scherbak N: Stable expression of HIV-1 MPER extended epitope on the surface of the recombinant probiotic bacteria Escherichia Coli Nissle 1917 using CRISPR/Cas9. Microb Cell Fact. 2024, 23[1]:39.

https://doi.org/10.1186/s12934-023-02290-0

  1. 119. Bagherpour G, Ghasemi H, Zand B, Zarei N, Roohvand F, Ardakani EM, Azizi M, Khalaj V: Oral administration of recombinant Saccharomyces boulardii expressing ovalbumin-CPE fusion protein induces antibody response in mice. Front Microbiol. 2018, 9:723.

https://doi.org/10.3389/fmicb.2018.00723

  1. 120. Zhang Y, Bailey TS, Hittmeyer P, Dubois LJ, Theys J, Lambin P: Multiplex genetic manipulations in Clostridium butyricum and Clostridium sporogenes to secrete recombinant antigen proteins for oral-spore vaccination. Microb Cell Fact. 2024, 23[1]:119.

https://doi.org/10.1186/s12934-024-02389-y

  1. 121. Li F, Zhao H, Sui L, Yin F, Liu X, Guo G, Li J, Jiang Y, Cui W, Shan Z: Assessing immunogenicity of CRISPR-NCas9 engineered strain against porcine epidemic diarrhea virus. Appl Microbiol Biotechnol. 2024, 108[1]:248.

https://doi.org/10.1007/s00253-023-12989-0

  1. 122. Valdez Y, Brown EM, Finlay BB: Influence of the microbiota on vaccine effectiveness. Trends Immunol. 2014, 35[11]:526-537.

https://doi.org/10.1016/j.it.2014.07.003

  1. 123. Rhee JH, Lee SE, Kim SY: Mucosal vaccine adjuvants update. Clin Exp Vaccine Res. 2012, 1[1]:50-63.

https://doi.org/10.7774/cevr.2012.1.1.50

  1. 124. Ensign LM, Cone R, Hanes J: Oral drug delivery with polymeric nanoparticles: the gastrointestinal mucus barriers. Adv Drug Deliv Rev. 2012, 64[6]:557-570.

https://doi.org/10.1016/j.addr.2011.12.009

  1. 125. Zahedipour F, Zahedipour F, Zamani P, Jaafari MR, Sahebkar A: Harnessing CRISPR technology for viral therapeutics and vaccines: from preclinical studies to clinical applications. Virus Res. 2024, 341:199314.

https://doi.org/10.1016/j.virusres.2024.199314

 

 

Probiotics as an Adjunct Therapy for Decreasing Gastrointestinal Complications from Chemoradiotherapy

Mohammad Reza Talaei Mashhadi, Mohammad Mousavi, Ghazaleh Khalili-Tanha, Elham Nazari

Applied Food Biotechnology, Vol. 11 No. 2 (2024), 26 Aban 2024, Page e6
https://doi.org/10.22037/afb.v11i2.46338

Background and Objective: Cancer treatments, including chemotherapy and radiotherapy, can cause diarrhea, which negatively affects the quality of life and willingness of patients to continue their treatment plans. In recent years, evidence have shown that the gut microbiome contributes to the development of these adverse effects as well as decreasing their severity. Probiotics have been shown to decrease treatment-related gastrointestinal symptoms. Probiotics are defined as live microorganisms that provide health benefits to the host.

Results and Conclusion: This report assessed the effectiveness of various probiotic strains, including Lactobacillus and Bifidobacterium, in decreasing diarrhea associated with chemotherapy and radiotherapy. Clinical trials have shown that certain probiotic formulations can decrease diarrhea occurrence and severity, decrease antidiarrheal medication needs and improve overall treatment tolerance. Despite these promising findings, further research is needed to optimize probiotic use in cancer therapy and fully understand how probiotics modulate gut health during chemotherapy. Therefore, probiotics can be used as an adjunctive treatment strategy in chemoradiotherapy cases to improve patient quality of life and clinical outcomes.

 

 

 

An Overview of the Relationships between the Food Industry and Nanotechnology

Mehdi Koushki, Nasrin Amiri-Dashatan, Hossein Pourghadamyari, Hadi Khodabandehloo, Fatemeh Bagheri, Masoumeh Farahani, Lobat Tayebi

Applied Food Biotechnology, Vol. 11 No. 2 (2024), 26 Aban 2024, Page e7
https://doi.org/10.22037/afb.v11i2.46737

Background and Objective: Due to the growth of the global population, food demands are increasing. Hence, the need to develop more efficient methods for producing better quality, safer, and more sustainable food seems essential. In the past decades, the use of nanoscale materials has increased greatly due to the unique chemical, physical, and biological characteristics of nanomaterials compared to bulk materials. This research presents nanotechnology role in improving sensorial properties (taste, appearance, and texture) and safety aspects as well as processing and packaging of foods. The use of nano-omics-based technologies and artificial intelligence-nanotechnology-based technologies in the food industry is also discussed.

Results and Conclusion: Linking food science and nanotechnology as a multidisciplinary scientific field can help safeguard food security, production, processing, storage, and quality control. Although nanotechnology has addressed the challenges in different sectors of food biotechnology, there are still various problems and opportunities to improve the current conditions as well as the complications and health risks caused by nanotechnology.

Conflict of interest: The authors declare no conflict of interest.

  1. Introduction

 

The food industry needs innovative and advanced technologies to improve safety and quality programs while developing fresh, genuine, and tasty food products. Nanotechnology is a powerful technology that provides the potential for fundamental advances in essential fields such as medicine, agriculture, and food systems. Nanotechnology is used to address various food issues by developing nano-filters, nano-biocatalysts, nanotechnology-derived food ingredients, nanocomposite films, edible coatings, nano-sensors, nano-tracers, and nano-delivery systems to improve food processing, manufacturing, packaging, pathogen detection, monitoring, safety, security, and quality [1–3]. Protecting humans against pathogens in water, foods, and the environment and helping to maintain food security are issues that can be achieved through innovations in nanotechnology. Studies show that nanotechnology outperforms traditional food processing methods by extending the shelf life of food products, decreasing the risk of contamination and enhancing the overall quality of foods. The use of nanotechnology within food industries has resulted in the generation of larger volumes of sustainable, safer, and healthier food products for human consumption, addressing the escalating demands of a burgeoning global population. Nanotechnology is expected to revolutionize the entire food industry, changing the methods of production, processing, packaging, transportation, and consumption. Foods undergo numerous transformations after harvesting and processing that may affect their biological and biochemical characteristics. Innovations in nanotechnology have the potential to help in this area [4, 5]. This review intends to provide an update on the possible uses and impacts of nanotechnology in food systems. It emphasizes the characteristics of food nanotechnology, as well as its current and emerging uses within the field of food science. In the following, the present review discusses the potential applications of nanotechnology in improving strategies related to the processing, packaging, as well as sensorial and nutritional quality of food. The use of nano-omics-based technologies and artificial intelligence (AI)-nanotechnology in the food industry has also been discussed.

  1. Results and Discussion

2.1. Nanotechnology in food processing and production

2.1.1. Nanofiltration technology

Nanofiltration is an emerging membrane technology with unique advantages in the separation of molecules based on size and charge. The pore size of nanofilteration membranes is typically between 0.1 and 1 nanometer, making them ideal for the separation of small solutes and larger ions [6]. Unlike reverse osmosis, nanofilteration allows salts and small molecules to pass through, providing a selective filtration mechanism. The food industry has recognized the potential of nanofiltration as it addresses a number of processing challenges while improving product quality, reducing waste and improving sustainability.

Nanofiltration as a pressure-driven process is similar to reverse osmosis but at lower pressures (5–40 bar), allowing selective permeability based on molecular weight cut-off and solute charge. Nanofilteration membranes, which are made from polymeric or ceramic materials, are defined by their pore size, charge characteristics and composition [7].

Key filtration mechanisms include size exclusion; by which, molecules larger than the pore size of the membrane are retained while smaller molecules pass through; Donnan exclusion, which enables separation of salts and charged molecules by repelling or attracting charged solutes; and solution diffusion; in which, solutes dissolve in the membrane and diffuse through under pressure. Due to its versatility, nanofiltration is used in various processes in the food industry, e.g., for demineralization, concentration and fractionation of food ingredients [8].

However, the most important approaches in nanofiltration include [7] a) Donnan exclusion, which distinguishes charged molecules based on their ionic characteristics, b) solution diffusion, which depends on the solubility of the solutes and their diffusion through the membrane under pressure, c) hydrophilic and hydrophobic interactions; by which, customized membrane materials improve separation performance and d) hybrid systems including integration of nanofiltration with additional technologies (e.g. ultrafiltration) to improve selectivity and throughput.

2.1.2. Applications of nanofiltration in the food industry

The food industry has utilized nanofiltration for various intentions, focusing on improving product quality, increasing production efficiency, and promoting sustainability. The major applications of nanofiltration in various food sectors are as follows.

2.1.2.1. Dairy industry

One of the most important uses of nanofiltration in the food industry is the processing of dairy products; where it is used for various purposes [8]. Nanofiltration plays a key role in whey protein concentration, a valuable byproduct of cheese production by removing water and solutes with low molecular weights (LMW) such as lactose and salts while leaving larger protein molecules behind [9]. This process leads to the production of whey protein concentrate, which is widely used in sports nutrition and food fortification. Nanofiltration is also used to reduce lactose through partial demineralization [9]. This selectively removes the lactose while retaining the protein content, making it an effective technique for the production of dairy products that are lactose-free or low-lactose, designed for lactose-intolerant consumers [10]. In addition, nanofiltration membranes are used in the processing of skimmed milk to remove excess water and minerals. The result includes a concentrated product with a higher protein content, which is particularly beneficial in the production of high-protein milk drinks and yogurts [11].

2.1.2.2. Beverage Industry industry

Nanofiltration is used extensively in the beverage industry, particularly in the processing of fruit juices and wine. In the juice industry, nanofiltration is used to clarify fruit juices by removing suspended solids, pectins, and colloidal particles while preserving essential flavors, vitamins, and minerals; thereby, improving the shelf life, quality, and sensory characteristics of the juices [12]. Additionally, nanofiltration plays a role in sugar decrease by selectively removing monosaccharides without diluting natural flavors while preserving major polyphenols and flavors [13].

 

2.1.2.3. Water treatment

Water is a critical content in almost all food-processing operations and effective treatment of the process water is essential for food manufacturing. Nanofiltration is often used for water softening and desalination, as nanofiltration membranes efficiently delete divalent ions, including calcium (Ca2+) and magnesium (Mg2+) that contribute to water hardness [14]. This improves water quality for food processing and increases the effectiveness of CIP (cleaning-in-place) systems [15]. In addition, nanofiltration plays an essential role in wastewater treatment by separating and concentrating valuable components such as proteins and sugars from wastewater streams [15]. These components can be recycled back into the production process, decreasing water consumption, minimizing waste, and decreasing the overall environmental effects of food production.

2.1.2.4. Edible oil processing

Nanofiltration has shown considerable potential in the edible oil industry, particularly in refining and purification processes. One of its main applications is degumming, where nanofiltration is used to remove phospholipids and other mucilage from crude vegetable oils [16]. The result includes cleaner more stable oils with a longer shelf life and improved product quality. Nanofiltration is used to recover solvents in oil extraction processes where organic solvents are used [17]. By recovering these solvents for reuse, nanofiltration not only decreases operating costs but also contributes to further sustainable and eco-friendly methods of oil production.

2.1.2.5. Sugar and sweetener production

The sugar industry has adopted nanofiltration for the refining of raw sugar and the production of sweeteners such as high fructose corn syrups [18]. Nanofiltration is used to remove colorants, organic acids, and other impurities from sugar solutions to improve the purity and whiteness of the final products. In high-fructose corn syrup production, nanofiltration plays a key role in removing impurities and concentrating the fructose content, resulting in a higher-quality sweetener [18]. In addition, nanofiltration is used in the processing of molasses, helping to fractionate by separating and concentrating valuable compounds such as betaine and other nutrients, adding value to what would otherwise be a waste stream [19].

Although conventional filtration also has the aforementioned applications, it should be noted that one of the advantages of nanofiltration is its ability to selectively remove impurities while allowing some dissolved substances to pass through. First, this helps preserve important nutrients and flavors. Second, it saves energy compared to reverse osmosis as it works at a lower pressure. Third, it includes positive effects on the environment as fewer harmful chemicals are used to soften and purify water. Fourth, it preserves heat-sensitive ingredients as it operates at lower temperatures and protects sensitive food ingredients from heat damage [17]. Overall, nanofiltration is transforming the food industry by providing a versatile and efficient method of separating, concentrating and purifying foods. Its uses range from dairy processing and juice clarification to water treatment and edible oil refining. With the increasing demand for high-quality, sustainable, and cost-effective food processing technologies, nanofiltration will play an increasingly important role in the future horizon of food production. However, overcoming the challenges associated with fouling, costs, and durability of the membranes is critical to realizing their full potential.

2.1.3. Nanobiocatalysts in food systems

Enzymes, also known as biocatalysts, are tools that play important roles in food processing to improve production processes. The novel method of combining biotechnology and nanotechnology to achieve desired results in bioprocessing applications is known as nanobiocatalysis. These results consist of improved enzyme properties, including activity, stability, and capacity. Nanobiocatalysts have many applications in the food industry, including production and packaging, contaminant identification, purification, and clarification [20, 21]. Advances in nanobiotechnology have greatly contributed to the improvement of enzyme technology. The use of nanotechnology in enzyme-based food processing methods can increase cost-effectiveness and improve sustainable and multifunctional materials, as well as multicompartmentalized structures. These benefits are achieved by improving the catalytic efficiency of enzymes and providing optimal enzyme uses. Enzyme-nanostructure biocatalysts have several advantages such as enhanced activity, stability, and recyclability. Studies indicate that the use of nanomaterials as biocatalyst carriers will be of great importance in the food industry in the future [20].

2.2. Texture and quality of food

Texture serves as a key attribute for consumers in the determination of food qualities. The sense of food texture is developed when consumers feel the food in their mouths. Hence, food texture is a key factor in determining whether consumers enjoy a particular food item. The texture of a food results from its structure or microstructure [22]. The textural characteristics of food can be classified into three groups based on the sensation mechanisms, as described previously, including visual, acoustic, and tactile characteristics. The most common examples of visual texture characteristics include smoothness, glossiness, thinness, and viscosity. The visual texture is determined by the food's appearance and light reflection [23]. The most popular texture features related to hearing are crispness and crunchiness, which are associated with the noise created when solid food is broken down. Such texture features are closely related to the other sensory stimuli identified by mechanoreceptors. Internal scull vibration has also been shown to be important to the sensation of these texture features [24]. Among all texture properties, tactile texture is likely the most prevalent and frequently serves as the primary emphasis in the study of texture. The perception of tactile texture characteristics occurs through the direct interaction of food with the human skin, including the hands and oral surfaces [25]. Advertising, pricing, and consumption environment can affect how the consumers perceive the eating quality of foods, but sensory factors often are centered in the field of food quality. The significance of appearance, aroma, and taste has garnered considerably more focus compared to that of texture. This can stem from limited comprehensions of the physiological mechanisms underlying texture perception. This concept has emerged as a refresh word in the sensory literature. An extra concern may include that poor texture, in contrast to appearance, flavor, and odor, does not serve as a reliable indicator of spoilage. Thus, a high-quality texture is frequently linked to superior food preparation [26].

One further explanation for the comparatively limited focus on texture can include inherent complexity involved in the perception of texture. Tactile feedback from the fingers plays a significant role as it serves as a quality indicator when selecting fruits and vegetables. Use of utensils such as knives, forks and spoons to serve foods provides an additional type of tactile information. These tools serve as sources of textural details before the food is consumed [27]. In the mouth, the act of biting involves a sophisticated sequence of shearing and crushing movements, which serve to decrease the whole food into smaller fragments. Tongue and palate assume a progressively significant function during this process, skillfully manipulating and compressing the food until the textural characteristics of the bolus render it appropriate for swallowing [28]. Therefore, assessment of the texture can only be effectively carried out with the involvement of human subjects. Of the most important tools for the assessment of texture is quantitative descriptive analysis, which assesses all sensory characteristics of foods that are typically selected by the panelists during round-table discussions. A high-fructose corn syrup panel is normally trained to establish terminology specific to a product type and assigns scores reflecting the intensity of its attributes. While various forms of high-fructose corn syrup have become popular in the industry, use of trained panels can often be excessively time-consuming and costly [29]. Due to the challenges associated with sensory methods, significant efforts have been directed to use instrumental assessments to assess the physical characteristics of foods. Hence, varieties of empirical instruments have been created for particular uses. Typically, these devices are quite straightforward, frequently portable and provide a restricted scope of semi-quantitative information.

2.2.1. Nanotechnology-derived food ingredients

Nanotechnology involves the manipulation of nanomaterials for various uses, significantly affecting the food and agriculture industries. This helps in enhancing crop yields, improves the quality and safety of food, and promotes human health via innovative and novel methods [30]. Nanoparticles, including silver (Ag), gold (Au), zinc oxide (ZnO), titanium dioxide (TiO2), and carbon, find uses in air filtration systems, food storage solutions, deodorizing products, medical bandages, toothpaste formulations, paints, and various household items [31, 32]. Moreover, nanoparticles are repeatedly used as food additives to safeguard against contaminations; thereby, extending the shelf life of the food products. It can serve to identify food pathogens, functioning as indices of food quality and safety. The nanomaterial is divided as organic and nonorganic-based, as well as a mixture of the two [33]. The use of inorganic nanomaterials as antimicrobial agents in food packaging is extensive. Supplying of inorganic nanoparticles can be achieved via various methods, including sol-gel techniques, mechano-chemical processing, and physical vapor synthesis, which are determined by the specific type of inorganic nanoparticles that are produced [34]. Nanostructured materials consist of microstructures formed from nanoscale components or structures, with dimensions typically ranging 1–10 nm. These materials occupy a size range that bridges the nanoscale and microscale, allowing for the development of various forms [35].

Nanostructured materials primarily comprise nanorods, nanoparticles, nanowires, and similar entities, which typically assemble into layered films, wires, and atomic configurations. Nanostructured materials are classified into three distinct classes of (i) zero-dimensional for nanoparticles, (ii) one-dimensional for nanorods or nanotubes, and (iii) two-dimensional for thin films, with three-dimensional for nanocomposites and dendrimers. These classifications are determined by the dimensions of the structural elements [35]. Food-grade ingredients are used in creating nanostructures through a straightforward layer-by-layer method and a cost-effective method. A comprehensive understanding of the biological and physical processes occurring within food systems is necessary to understand distinct physical, chemical, and biological characteristics of these nanostructures and nanoparticles [35]. Various techniques, including milling, homogenization, ultrasound emulsification, and microfluidization, are used for the generation of nanoparticles [36]. Milling is a traditional method primarily used for grinding materials to achieve flour at the nanoscale through mechanical energy. Production of fine wheat flour and green tea powder, which enhance water binding capacity and antioxidative characteristics, include important uses.

Homogenization, a long-established technique, is used to decrease fat globules in milks; thereby, stabilizing emulsions. In contrast, high-pressure homogenization is further effective in creating highly fine emulsions using significant stresses [37]. Microfluidization not only facilitates size decrease and formation of emulsions but also improves texture and mouthfeel. This technique is used in the production of various products (e.g. yoghurts, syrups, creams, malted beverages, flavored oils, icing and fillings, and salad dressings) [38]. Numerous food ingredients are included in their natural state at the nanoscale such as milk proteins and casein, and distinct from synthetic nanomaterials. A majority of proteins are globular in shape, typically ranging 10–100 nm, while polysaccharides and lipids are generally smaller than 1 nm. The most important nanostructures in food systems include nanostructured proteins, nanoemulsions, nanocomposites, and liposomes, as they enhance bioavailability, safeguard bioactive components, and facilitate controlled releases [39].

2.2.2. Nanocomposite films and edible coatings

Nanocomposite food packaging, characterized by its affordability and high functionality, represents a superior alternative to traditional packaging substances such as papers, glasses and metals. Polymer matrices are enhanced with nanofillers such as nano-oxides, nanoclays, cellulose microfibrils, and nanotubes based on carbon to create nanocomposites. In these materials, at least one dimension of the phase is smaller than 100 nm [38]. Natural materials (e.g. cellulose, chitosan and carrageenan) and synthetic materials (e.g. nylon, polystyrene, polyamides, and polyolefins) are used in packaging processes [41, 42]. One of the important reasons in international trade includes protection of fresh horticultural products. Thus, prolonged shipping times and extended distribution durations may increase the risk of significant losses. In this regard, bionanocomposite films and coatings play significant roles in minimizing weight loss by serving as barriers against moisture evaporation. In addition, edible coatings can make the texture better, enhance the visual appeal of the products, and extend food shelf life by forming semi-permeable barriers that regulate gas exchanges. Edible coatings and packaging can be developed from various biological sources to enhance preservation of fruits, vegetables, and cheeses. This film effectiveness is determined by the specific materials and their natural characteristics, which can affect their barrier, microbial, mechanical, and optical characteristics [43]. Packaging needs change depending on the food type, as maturation rates, mechanical stability, and water content depend on their compositions [44]. Additionally, numerous disadvantages are linked to the use of biopolymers in food packaging. To moderate these issues, composite or multicomponent films are developed, which consist of a combination of various materials that possess beneficial characteristics. For example, nanosilver-coated fruits and vegetables maintain their activities throughout transportation and storage due to modifications in their respiration processes [45]. Studies are currently carried out on heavy-metal nanoparticles as components of edible coatings to enhance nutrient uptake and bioavailability. The development of self-cleaning nanomaterials is popular as a promising area of research, owing to the innovative self-cleaning characteristics provided by nanocomposites under specific storage conditions. By using thinner hybrid edible films with a thickness of less than 100 µm, moisture, CO2, water, and oxygen barrier characteristics can be enhanced, which in turn can improve the shelf life and sensory attributes of the food products [46]. Figure 1 presents a diagram that summarizes edible coating in the food industry. Nanocomposite films are emerging as an additional packaging solution, consisting of a reinforcement of the biological polymer matrix with nanocrystals that improve their strength, thermal stability, and barrier characteristics. This process remains in a nascent stage and is essential for the design and development of progressed processing technologies.

2.3. Food safety assessment and storage

Human health and life are directly affected by food safety, making it a major public health concern. Health risks linked to food safety are exacerbated by unhygienic conditions, incorrect storage, mishandling of foods, and contaminated food products, potentially causing several serious food-driven diseases [47]. For example, ranges of potentially harmful substances are reported that may occur in various food categories at every stage of food processing. These dangerous pollutants include heavy metals, toxic substances, pharmaceuticals, pesticides, prohibited additives, fungi, and bacteria [48]. Traditional strategies such as high-performance liquid chromatography (HPLC), mass spectrometry (MS), liquid chromatography-MS (LC-MS), gas chromatography-MS (GC-MS), and polymerase chain reaction (PCR) are well-established methods for assessing food quality, safety and detecting a wide range of chemical and biological components [49]. Despite their high sensitivity, accuracy, and stability, these methods typically involve slow and laborious processes that potentially delay the timely detection of hazards, thereby threatening consumer health. Nanotechnology, particularly nanobiosensors, has emerged as a promising solution for researchers and scientists to overcome these challenges.

2.3.1. Nanosensors for food safety

Nanosensors are tools used to assess and ensure the quality of food samples, water, and other environmental factors. The use of nanomaterials and biological recognition elements by these sensors allows researchers to analyze certain biological and chemical components in foods. A sensor is a complicated system capable of reacting to a target in a qualitative or quantitative manner [50]. Nanosensors have a similar structure to conventional sensors, but their manufacture includes nanoscales. In addition to advantages such as high speed and immediate monitoring, high sensitivity, and detection of contaminants and pathogens in foods, these can be used in portable handheld devices [51]. The distribution of biosensors based on nanosensor devices and their uses in food analysis are shown in Figure 2. Nanobiosensors operate by using various methods that involve the use of nanomaterials and biological components to identify and assess specific analytes. Nanosensors can be categorized into three primary classes based on the physical phenomena used to interpret outputs of the interactions between the targets and analytes, including mechanical, electrochemical, and optical nanosensors.

2.3.1.1. Optical nanosensors

Optical sensors include extensive uses of quantum dots, gold, silver, up-conversion nanoparticles, and metal oxide-based, and organic fluorescent molecular-based nanomaterials to enhance performance. This section highlights colorimetric biosensors, fluorescent, and surface plasmon resonance (SPR) biosensors for the analysis of foods, depending on the type of signal outputs.

- Colorimetric biosensors for the food safety analysis

Colorimetric sensors have attracted increased attentions due to their simplicity as sensing methods, compared to various optical techniques. Using colorimetric method, color shifts can quickly be observed with the bare eyes, eliminating needs of complicated and expensive tools [52]. Major used nanomaterials are gold and silver, especially gold nanoparticles in various forms such as spheres, hollow spheres, nanorods, and nanotubes. Gold nanoparticles are extensively used in creating colorimetric assay sensors for detecting food contaminants within various nanomaterials, owing to their distinctive optical characteristics, straightforward synthesis process, excellent stability, and effortless modification [53]. Colorimetric sensors using gold nanoparticles are typically fabricated based on changes in the distance of the gold nanoparticles. Gold nanoparticles dispersed typically display a red hue, with their peak absorption wavelength approximately at 520 nm. Nevertheless, gold nanoparticles undergo polymerization, the solution color shifts to a navy blue or purple hue, indicating a surface plasmon band that transits from a visible spectrum into a near-infrared one [54].

A colorimetric approach with gold nanoparticles is used for the detection of food contaminants, including pesticide residues, heavy metals, veterinary pharmaceuticals, infectious agents, and toxins. For example, a colorimetric aptasensor for aflatoxins and ochratoxin A (OTA) detections was established, relying on the hybridization process between the capture DNA modified with amino groups and an OTA-specific amine-modified aptamer [55]. In another study, Luan and colleagues established a label-free detection method for OTA, using aptamers in addition to gold nanoparticles and the polymer poly diallyldimethylammonium chloride [56]. Colorimetric nanobiosensors based on aptamers are used for the recognition of infectious microorganisms present in food items. Yuan et al. presented an aptasensor for Staphylococcus aureus using gold nanoparticles [57]. Surface of the microplate for this aptasensor was altered by incorporating a biotin aptamer specific to S. aureus via the avidin-biotin interaction [57]. Colorimetric nanobiosensors based on antibodies are sensing methods for the identification of pathogenic agents in food products.

- Fluorescence biosensors for the food safety analysis

Compared to the colorimetric method, fluorescence phenomena include several advantages over subjective colorimetric methods in food detection. These offer a lower background, increased sensitivity, enhanced objectivity, lower detection limits and improved repeatability. Xu et al., [58] discovered carbonate quantum dots, the fluorescence emission characteristics of these dots have made them valuable as sensing probes for analyzing foods. Other nanomaterials without fluorescent characteristics can produce fluorescence effects for food analysis when combined with fluorescent biomolecules. Several conventional fluorescent probes include a chromophore that generates strong fluorescence when it is in a separated state; however, aggregation-caused quenchingcan prevent these molecules from emitting energy at a lot concentrations or when concentrated. With detection of the aggregation-induced emission effect (AIE) phenomenon, limitations of the aggregation-caused quenching have largely been overcome, and AIE can be used for high fluorescence concentrations. Detection mechanisms for AIE luminescence (e.g. AIEgens) typically encompass the following elements of interactions involving electrostatic and hydrogen bonds, alterations in the solubility of AIEgens, disruptions in the quenching of AIE luminescence, fabrications of AIE at the nanoscale, and target-induced disaggregation of AIEgens. Fluorescent nanobiosensors based on antibody and aptamer are used for the discovery of infections and chemical contaminants in food processing. To detect Salmonella typhimurium, Hu et al. prepared a multisignal immunoassay platform for S. typhimurium recognition [59]. The method uses colorimetric fluorescent magnetic nanospheres as labeling agents and two formats of quantitation. To assessment efficiency of the prepared assay, it was used for the recognition of S. typhimurium in samples such as milk, and tap water. Results showed that the fabricated platform was appropriate for the recognition of S. typhimurium [59].

- Surface plasmon resonance for the food safety analysis

The other optical nanosensors is SPR sensor, which is a specific optical technique that recognizes alterations in the refractive index because of the connections between ligands and molecules present on the sensor surface within a sample. The SPR technique includes advantages, compared to other conventional optical techniques. In general, SPR is composed of a light source, an optical arrangement, a sensing component, and a detection mechanism. Currently, there are four distinct types of SPR biosensor platforms, including 1) FOSPR, fiber optic surface plasmon resonance, 2) LSPR, localized surface plasmon resonance, 3) SPRI, surface plasmon resonance imaging, and 4) TSPR, transmission surface plasmon resonance. Nanoparticle-based SPR technology is extensively used for identifying contaminants in foods. Park et al. introduced an aptasensor based on LSPR for the identification of OTA [60]. This method allowed for the quantitative detection of OTA at concentrations below 1 nM. Findings of this study demonstrated that the aptamer-functionalized gold nanorods (GNR) possessed significant efficacy, as it could be regenerated for reuses of more than seven times through heating in methanol at 70 °C to eliminate OTA. The suggested biosensor system demonstrated significant selectivity for OTA, compared to other mycotoxins [60].

2.3.1.2. Electrochemical nanobiosensors

One of the most commonly used techniques for ensuring food safety is the electrochemical detection approach. Because of their low cost, user-friendliness, natural sensitivity, rapid sensing capability, and ability to work with transportable systems, electrochemical biosensors are now the most swiftly expanding types of sensors. Traditional mercury-based electrodes have increasingly been substituted with modified electrodes using various appropriate nanomaterials, including biocompatible and highly conductive carbon nanomaterials, stable nanozymes with adjustable catalytic characteristics, and metal-organic frame-works known for their greater numbers of active sites and high porosity [61]. Electrochemical biosensors are categorized into three types based on the distinct conduction mechanisms of impedance, which are derived from the interactions between the target analytes and the detection elements located on the biosensor surface, including potentiometric, electrochemical, and impedance sensors [62].

- Antibody-based electrochemical nanobiosensors for the food safety analysis

Antibody-based biosensors are designed based on the principle of antibody-antigen interactions. Antibody-driven electrochemical systems facilitate fast and continuous monitoring of binding interactions without the necessity of extra reagents or washing/separation steps [63]. Electrochemical immunosensors are characterized by their simple construction, economical tools, low power demands, ability for mass production, quick response, high sensitivity, and adaptability for miniaturization. By miniaturizing the components and situating the signal detection elements close to the biocomponent, immunoelectrochemical sensors can convert centralized quantitative immunoassays into convenient portable biosensors [63]. In 2015, Xiang and colleagues created an electrochemical sensor for Salmonella identification [64].

- Aptamer-based electrochemical nanobiosensors for the food safety analysis

Nowadays, ensuring food safety is a primary concern for authorities and professionals involved in the supply chain for food products. Limitations of the current methods have resulted in the development of novel and innovative technologies, including biosensors. Biosensor design mostly depends on the development of novel receptors with higher affinity for the targets as well as high stability. Aptamers show these traits, making them promising substitutes for the natural receptors. Aptamers refer to short sequences of nucleotides generated by the systemic evolution of ligands through exponential enrichment, capable of high-affinity binding to specific ligands [65]. In 2018, Hasan et al. suggested an electrochemical biosensor using an amino-modified aptamer for the detection of whole-cell Salmonella spp., which was based on an indium tin oxide electrode coated with multi-walled carbon nanotubes. This designed aptasensor was well used for the identification of Salmonella spp. in food products [66]. Recently, Tang et al. reviewed gold-based aptasensors for detecting kanamycin in foods [67]. Kanamycin is isolated from the Streptomyces kanamyceticus and predominantly used in a sulfated form as an antibiotic in medicine. Moreover, kanamycin can be condensed into the human body and transmitted in the food chain. Thus, presence of kanamycin in foods derived from animals is a potential hazard to human health.

2.3.2. Nanomaterials for nanosensors

Nanomaterials can be synthesized through various bottom-up techniques, including cutting, ball milling, extruding, chipping, and pounding, as well as top-down methods, leading to diverse structural types [68]. Relatively, several carbon-based nanomaterials are described for an improved understanding of the differing nature of nanomaterials. Carbon-derived nanomaterials include carbon nanotubes, nanowires, nanoparticles and fullerenes. Carbon nanotube is the most-used carbon-based nanomaterial. A major use of the carbon nanotube as a sensor is in the field-effect transistor. A broad variety of field-effect transistor are manufactured by chemical doping of carbon nanotubes [69]. Carbon nanotube field-effect transistors are used to detect various types of gases such as CO2, NH3, O2, NO2, and N2. Carbon nanotube-FETs are used for detections in biological sciences [69]. Another carbon-based nanomaterial is nanowire. Nanowires are commonly used in manufacturing nanosensors, rather than carbon nanotubes. Nanowires are produced through a variety of processes such as chemical vapor deposition, laser ablation, alternating current electrodeposition, and thermal evaporation [70]. Moreover, nanowires are used to produce gas sensors that can qualitatively detect NH3.

Nanoparticles are commonly used nanomaterials not only in sensor fabricating but also in numerous other designing uses. Nanoparticles themselves are not simple molecules and therefore consist of three layers. These layers include (a) the surface layer, which can be used to functionalize the nanoparticle; (b) the shell layer and (c) the core, which is essentially the central portion of the nanoparticles [71]. Nanoparticles can be classified into various classes of (a) carbon-based nanoparticle, (b) metal nanoparticle, (c) ceramic nanoparticle, (d) semiconductor nanoparticle, and (e) polymer nanoparticle [72].

2.3.3. Antibacterial characteristics of nanoparticles for food safety

Foodborne pathogens, which include bacteria, parasites, and viruses, include the potential to contaminate food products, resulting in foodborne illnesses such as foodborne infections and intoxications. Foodborne poisoning occurs when a toxin-producing pathogen contaminates food and the resulting toxin is consumed by the people [73]. The general rate of foodborne problem causes special attentions to infectious products; therefore, numerous attempts are assigned to the creation of novel and functional agents to deal with food-pathogenic microorganisms. Packaging of food products is globally used to keep the standard of food quality and broaden the shelf life. Properly designed packaging can defend food against microbial threats and various types of environmental contaminations, whereas inadequate packaging increases food waste and occurrence of foodborne illnesses. Nanoscience develops nanomaterials with unique physicochemical and antimicrobial characteristics in the field of the food industry [32,74]. Antimicrobial nanomaterials generally involve addition of inorganic nanoparticles with antimicrobial characteristics. Common inorganic nanoparticles used in food packaging are silver nanoparticles, renowned for its outstanding toxicity against various microorganisms, as well as low volatility and stability at high temperatures [75]. TiO2 nanoparticles are other types of antimicrobial nanoparticles that have been used in food packaging. Titanium dioxide nanoparticles exhibit photocatalytic characteristics that lead to the peroxidation of polyunsaturated phospholipids within the microbial cell membranes. This characteristic has been used to inactivate various foodborne pathogens [76]. In addition to silver and TiO2 nanoparticles, numerous other nanomaterials have been used by the food industry. Classification of nanomaterials with antimicrobial characteristics and their uses are summarized in Table 1.

2.4. Developing functional foods for health and disease conditions using nanotechnology

A functional food is defined as a food that offers additional health benefits in addition to the food's usual nutritional value. The use of nanotechnology can lead to significant advancements in human health by enhancing the delivery of nutraceuticals and bioactive compounds within functional foods [77]. The use of nanotechnology extends beyond the creation of functional foods; it is used in food processing, packaging, and swift identification of foodborne pathogens in various food products. The term "nanofood" highlights products that have been grown, manufactured, processed, and/or packaged through the use of nanotechnological methods or instruments or those that have been improved with the incorporation of nanomaterials [78]. Functional foods include a diverse range of components, nutrients, and non-nutrients that enhance human health and decrease the risks of diseases. Bioactive compounds demonstrate protective characteristics against hypertension, cardiovascular diseases (CVD), and various cancers [79]. Despite this, there are some challenges that the functional food industry faces in optimizing the activity of bioactive compounds, including the stability of bioactive compounds, limited solubility in water, and low bioavailability of bioactive compounds [80]. The process of encapsulation involves enclosing active agents within a carrier substance to enhance the efficient delivery of bioactive molecules and living cells to foods. This technique is used extensively for improving the delivery of bioactive molecules and living cells to foods [81]. In this process, functional ingredients are packed in a protective coating to prevent degradation of the functional ingredients during processing, storage, and use of the ingredients [77, 82]. It must not affect the appearance, taste, flavor, texture, and shelf life of the products and materials used in the encapsulation systems, and it must be safe [79]. Various technologies are used to create nanocapsules that contain a multitude of bioactive and active ingredients; after they deliver their active ingredients, these nanocapsules are absorbed like common foods [83]. Biopolymeric nanoparticles, nanoliposomes, nanoemulsions, and nano-precipitation are the most effective nanoencapsulation methods for creating nanoencapsulated bioactive ingredients, which enhance the stability of both chemical and physical properties, along with the bioavailability [84]. Nanotechnology has ultimately contributed to the enhancement of the safety and nutritional quality of food products by making it possible to encapsulate highly stable and highly effective antioxidants, antimicrobials, and anticancer agents [85, 86].

 

2.5. Nutrition-based nanotechnology in food nutrient delivery

The alteration of the dimensions of manufactured materials to the nanoscale can significantly enhance the stability and solubility of the essential nutrients while facilitating their efficient and regulated transport through nanovehicles [87]. Significant increases have been reported in the use of nutraceuticals in recent years. Nutraceuticals are used for the management and prevention of various illnesses, including various cancers, skin diseases, gastrointestinal diseases, ophthalmic disorders, diabetes mellitus, obesity, and diseases linked to the central nervous system (CNS). Nutritious food supplies the essential nutrients necessary for the human body through dietary intake. However, many bioactive compounds in these nutrients show high lipophilicity and low solubility in water, which results in inadequate dissolution and oral bioavailability [88]. Thus, nutraceuticals such as curcumin, carotenoids, anthocyanins, omega-3 fatty acids, vitamins C and B12, and quercetin face challenges including low solubility, chemical instability, unpleasant taste, and undesirable odor. Furthermore, factors such as gastrointestinal membrane barriers, various pH levels, and interactions with gastrointestinal enzymes contribute to the degradation of specific nutraceuticals [89]. Nanotechnology-driven nutrient delivery systems have the potential to enhance oral bioavailability by improving the stability of nutraceuticals in food and the gastrointestinal tract, increasing their solubility in intestinal fluids, and decreasing first-pass metabolism in the gut and liver [90]. In recent years, food scientists have focused on creating innovative food products that possess improved functional characteristics. The development of nanocolloids for food uses has emerged as an effective approach for encapsulating nutrients and bioactive compounds, improving their absorption and bioavailability in the gastrointestinal tract [91]. Commonly encountered nanocolloids consist of nanoemulsions, nanomicelles, nanocapsules, and analogous structures. The growing interest in nanocapsules can be attributed to their uses in sophisticated drug delivery systems within the human body, as well as the development of liposomal-based nanocapsules that are used in food research, healthcare, and agriculture [92]. Nanocapsules provide a safeguard against degradation throughout the processing phase, facilitating the possibility of controlled and/or sustained releases of active ingredients. The application of delivery techniques involving synthetic chemicals in the field of biomedicine or pharmaceuticals may not be appropriate for the food industry, due to the critical need for safe substances. Therefore, probiotic nanoencapsulation presents a practical and safe method for the delivery of bioactive compounds in food products [93]. In recent years, the nanoencapsulation of probiotics has been importantly highlighted, especially through the fabrication of nanofibers using electrospinning methods. The process of encapsulating probiotic strains within nanofiber mats composed of corn starch and sodium alginate has significantly improved their stability and viability, exceeding the performance of non-encapsulated cells [94]. The incorporation of nanoencapsulated probiotics in food products presents numerous benefits, including enhanced protection of the probiotics and effectiveness in their transport to the intestine [95]. However, it is important to address certain problems such as increased costs, production complexities, potential adverse effects at nanoscales, intricate regulatory needs, sustainability issues, and the possibility of instability during processing [96].  In the following, we more focus on the encapsulation of nutrients and nanoencapsulation methods.

2.5.1. Encapsulation of nutrients and nanoencapsulation methods

Nutraceutical foods have attracted significant attention in studies due to their effects on the prevention and management of various health conditions. However, bioactive compounds encounter various challenges such as inadequate stability, low water solubility, and limited bioavailability. Encapsulating bioactive compounds within an appropriate carrier is beneficial as it enhances their solubility in water and protects them from degradation due to ecological and biological effects [97]. Of these methods, microencapsulation includes the capacity to improve the stability and bioactivity of bioactive nutraceuticals. The particles that are encapsulated are generally classified into three categories; microcapsules (0.2–5000 µm), macrocapsules (larger than 5000 µm), and nanocapsules (smaller than 0.2 µm). Larger in size and possessing a decreased surface area, macro- and micro-particles may show inadequate absorption of nutrients and other encapsulated substances within the gastrointestinal tract. Moreover, larger particles demonstrate extended dissolution rimes, which may hinder the body ability to release and absorb nutrients before their transition through the digestive system. Additionally, these substances can aggregate, leading to an irregular distribution within the gastrointestinal tract and affecting nutrient absorption in specific regions [98]. The elevated levels of the elements of the wall within encapsulated particles may hinder the b biodegradability of these particles in the gastrointestinal tract. Such hindrance can influence the release of encapsulated nutrients, thereby reducing their availability for absorption. Additionally, certain wall materials in the encapsulation process may provide resistance against digestive enzymes of the gastrointestinal tract [99]. This resistance may lead to a delayed release of the encapsulated nutrients, which can affect their absorption.

The nanoencapsulation of bioactive substances typically offers significant advantages [100]. The bioavailability of food components is greatly improved when the surface region available for their uptake increases [101]. This is especially important for flavoring agents that have a low solubility in liquids and for flavors and aromas that can be perceived at minimal concentrations [102]. Nanocarriers play a critical role in enhancing nutrient absorption by protecting these compounds during the digestive process and enabling a regulated release along the digestive tract [103]. Furthermore, the nanoencapsulation of bioactive compounds decreases undesirable connections with other constituents and provides protection against spoilage both during and after consumption [102]. The solubility of ingredients that are poorly soluble in water, like omega-3 fish oil, can be enhanced through the formation of a micelle network or by minimizing interactions with other components within the matrix [103]. This approach not only avoids discoloration but also mitigates off-flavors by covering undesirable tastes and odors. Furthermore, it facilitates the controlled release of mineral components, ensures optimal preservation during production and storage, and also improves the physical properties of the products [104]. Nanoencapsulated bioactive compounds are capable of releasing their contents in a controlled and sustained state, which further enhances their solubility and bioavailability for biological procedures. The enhanced solubility in water and regulated release of food substances when encapsulated at the nanoscale can be ascribed to particular factors [105].

2.6. Personalized nutrition

Nanotechnology has made significant progress in the medical sciences and allows for biological processes that were not previously possible. A nanotechnology approach is used to determine the positioning of nutrients or bioactive food components within tissues, cells, and cellular components with high accuracy. This detects nutrient and biomolecular interactions within specific tissues, as well as improves the nutritional standard quality of food, presenting numerous opportunities for a deeper understanding of the structures and functions of foods. Nanotechnology has the potential to improve nutritional assessment and measure bioavailability. In nutrition research, it may enable the identification of targets for nutrient action and biosignatures of nutrient efficacy to improve "personalized nutrition" [106]. Personalized nutrition develops tailored dietary strategies that address the unique nutritional needs of each individual; thereby, managing and potentially preventing various diseases [107]. An individual's needs are described by biological data (e.g. genetics, epigenetics, proteomics, metabolomics, microbiome, and physical descriptions), lifestyle, and health statuses [108, 109]. Omics technologies are used to capture risk variants in specific genes that can be used to modify diseases before symptoms appear [110]. Nutrigenomics is the study of how bioactive dietary compounds affect genes involved in signaling and biological pathways of various diseases [111].

Although dietary supplements make a significant contribution to disease prevention and include biological effects such as antioxidants and anticancer effects, some dietary supplements are difficult to dissolve in water and are not readily absorbed via the oral cavity, so delivery systems are vital [112]. Food colloids science focuses on improving human health through dietary interventions, and it can be useful for developing delivery systems [113]. As a result of stable-state precision delivery mechanisms, food functional factors can be delivered accurately to different individuals, enriching them in specific cells or tissues. Furthermore, functional factors are protected during transport and released at the designated location to enhance their efficacy [106, 114]. For example, the colloidal delivery systems are designed to break down in the gastrointestinal system and release their contents there, and then the bioactives are absorbed via the epithelial cells and carried into the body by the lymphatic system [115]. Nano-delivery systems such as nanocapsules and nanosphere, can enhance the bioavailability of bioactive compounds. This bioavailability causes more nutritional absorption and subsequently results in the production of active substances. The use of nanomaterials optimizes health outcomes by delivering nutrients and bioactive compounds in the right amounts and at the right time based on the genetic profile of the individual [116].

The integration of the use of omics-based technologies and biostatistics to investigate food and nutrition interactions at the molecular level is called "foodomics" [117]. Advances in the combination of food technology, omics-based data, and nanotechnology could pave the way for the development of personalized nutrition products and reveal the important role of nano-omics-based technologies in food science.

2.7. Integration of artificial intelligence into nanotech-nology in food science

Nowadays, the execution of cutting-edge innovation of AI, particularly machine learning, with its advantages and disadvantages, in several scholarly and mechanical areas is well known. Food science and technology and their associated issues are no exceptions to this rule. Improving agriculture, facilitating crop classification, developing formulation and novel food and nutraceutical-associated products, more accurate sensory evaluation, industrial processing, improving food quality, ensuring the safety of food, controlling the supply chain, reusing waste, saving costs and time, and lastly estimating calorie and nutrient amounts are some achievements of the combination of food science and artificial intelligence. Consequently, the merging of artificial intelligence with nanotechnology in the food science sector has formed a fresh domain of study and creativity called artificial intelligence -nanotechnology in food science. Consequently, artificial intelligence can play a crucial role in discoveries and innovations, resulting in the identification of novel nanomaterials that exhibit enhanced electrical, optical, and mechanical properties, along with unique functionalities suitable for various applications such as energy storage, sensing, and biomedicine. In summary, the future exploration of the challenges and opportunities linked to the integration of artificial intelligence in food science nanotechnology is worthy of investigation. Data collection related to food security and quality can help to develop suitable models and highlight the effective role of artificial intelligence in food quality assessment.

  1. Conclusion

Nanotechnology involves the manipulation and use of materials at small scales, where characteristics of the materials differ significantly, compared to their bulk similar materials, allowing useful alterations of their physical, chemical, mechanical, electrical and biological attributes. Hence, the field of nanotechnology as a transformative and promising platform can lead to the advancement of innovative and high-quality products in food systems. The applicability of nano-based materials in the fields of food processing, packaging, safety and the improvement of taste, appearance, texture and nutritional quality of foods is well known. Compared to traditional monitoring methods, the use of smart nanosensors helps ease operation and save costs and time at various stages of food production. Omics technologies in food science can uncover possible molecular mechanisms of food and nutrition interactions and can create promising approaches in combination with nanotechnology to improve personalized nutrition and food security. However, there are various obstacles associated with the application of nanomaterials in food biotechnology, including health effects and environmental damage that require to be overcome.

  1. Acknowledgements

This study was supported by Shahid Beheshti University of Medical Sciences (grant no. 43012047).

  1. Conflict of Interest

The authors report no conflict of interest.

  1. Author contributions

All authors participated in project administration and writing of the first draft of the manuscripts, providing critical revision and editing. All authors approved the final version of the manuscript.

 

  1. Ethical Code

This project was approved via

IR.SBMU.RETECH.REC.1403.422 ethical code.

References

  1. Jagtiani E: Advancements in nanotechnology for food science and industry. Food Front. 2022; 56-82.

https://doi.org/10.1002/fft2.104

  1. Mohammad ZH, Ahmad F, Ibrahim SA, Zaidi S: Application of nanotechnology in different aspects of the food industry. Discov Food. 2022; 2(1).

https://doi.org/10.1007/s44187-022-00013-9

  1. Rashidi L, Khosravi-Darani K: The applications of nanotechnology in food industry. Crit Rev Food Sci Nutr. 2011; 51(8): 723-730.

https://doi.org/10.1080/10408391003785417

  1. Nile SH, Baskar V, Selvaraj D, Nile A, Xiao J, Kai G: Nanotechnologies in food science: applications, recent trends, and future perspectives. Nano-Micro Lett. 2020; 12:34.

https://doi.org/10.1007/s40820-020-0383-9

  1. Ameta SK, Rai AK, Hiran D, Ameta R, Ameta SC: Use of nanomaterials in food science. Biogenic nano-particles and their use in agro-ecosystems, 2020; 457-488.

https://doi.org/10.1007/978-981-15-2985-6_24

  1. Suhalim NS, Kasim N, Mahmoudi E, Shamsudin IJ, Mohammad AW, Mohamed Zuki F, Jamari NL: Rejection mechanism of ionic solute removal by nanofiltration membranes: an overview. Nanomater. 2022; 12(3): 437.

https://doi.org/10.3390/nano12030437

  1. Linder C, Kedem O: History of Nanofiltration Membranes from 1960 to 1990. Nanofiltration: Principles, Applications, and New Materials. 2021; 1-34.

https://doi.org/10.1002/9783527824984.ch1

  1. Yadav D, Karki S, Ingole PG: Nanofiltration membrane processing in the food industry. Food Eng Rev. 2022; 14(4): 579-595.

https://doi.org/10.1007/s12393-022-09320-4

  1. Bobrova A, Ostretsova N: Prospects for use of nanofiltration buttermilk and whey concentrates in the technology of fermented milk products with an increased mass fraction of protein. in IOP Conference Series: Earth and Environmental Science, 2021; 624(1): 012137.

https://doi.org/10.1088/1755-1315/624/1/012137

  1. Li A, Zheng J, Han X, Yang S, Cheng S, Zhao J, Zhou W, Lu Y: Advances in low-lactose/lactose-free dairy products and their production. Foods. 2023; 12(13): 2553.

https://doi.org/10.3390/foods12132553

  1. Mishra A, Sunkesula V, Hammam AR, Metzger LE: Effect of Cavitation and High-Temperature Nanofiltration of Ultrafiltered Skim Milk on the Functionality of Milk Protein Concentrate Powder. Dairy. 2024. 5(4): 610-624.

https://doi.org/10.3390/dairy5040046

  1. Han WH, Li X, Yu GF, Wang BC, Huang LP, Wang J, Long YZ: Recent advances in the food application of electrospun nanofibers. J Ind Eng Chem: 2022 Jun 25;110: 15-26.

https://doi.org/10.1016/j.jiec.2022.02.044

  1. Mallakpour S, Azadi E: Nanofiltration membranes for food and pharmaceutical industries. Emergent Mater. 2022; 5(5): 1329-1343.

https://doi.org/10.1007/s42247-021-00290-7

  1. Labban O, Chong TH: Design and modeling of novel low-pressure nanofiltration hollow fiber modules for water softening and desalination pretreatment. Desalination. 2018; 439: 58-72.

https://doi.org/10.1016/j.desal.2018.04.002

  1. Kim W-J, Huellemeier H, Heldman DR: Recovery of cleaning agents from Clean-In-Place (CIP) wastewater using nanofiltration (NF) and direct contact membrane distillation (DCMD). Food Res Int. 2023; 167: 112724.

https://doi.org/10.1016/j.foodres.2023.112724

  1. Kaya D, Hung Y-T: Advances in treatment of vegetable oil refining wastes. Environmental and natural resources engineering. 2021; 325-375.

https://doi.org/10.1007/978-3-030-54626-7_8

  1. Nath K, Dave HK, Patel TM: Revisiting the recent applications of nanofiltration in food processing industries: Progress and prognosis. Trends Food Sci Tech. 2018; 73: 12-24.

https://doi.org/10.1016/j.tifs.2018.01.001

  1. Lipnizki F: Membrane processes for sugar and starch processing, in Engineering Aspects of Membrane Separation and Application in Food Processing. 2017; 241-268.

https://doi.org/10.4324/9781315374901

  1. Luo J, Guo S, Wu Y, Wan Y: Separation of sucrose and reducing sugar in cane molasses by nanofiltration. Food Bioprocess Technol. 2018; 11: 913-925.

https://doi.org/10.1007/s11947-018-2062-0

  1. Safdar A, Ismail F, Hussain A, Rafeeq H, Maqsood S, Iqbal HM, Zdarta J, Jesionowski T: Implementation of nanobiocatalysis in food industry, in Bionanocatalysis: from Design to Applications. 2023; 223-248.

https://doi.org/10.1016/B978-0-323-91760-5.00019-1

  1. Verma ML, Abraham RE, Puri M: Nanobiocatalyst designing strategies and their applications in food industry, in Biomass, Biofuels, Biochemicals. 2020; 171-189.

https://doi.org/10.1016/B978-0-12-819820-9.00010-7

  1. Jeltema M, Beckley J, Vahalik J: Food texture assessment and preference based on mouth behavior. Food Qual Prefer. 2016; 52: 160-171.

https://doi.org/10.1016/j.foodqual.2016.04.010

  1. Rahman MS, Al-Attabi ZH, Al-Habsi N, Al-Khusaibi M: Measurement of instrumental texture profile analysis (TPA) of foods. Techniques to Measure Food Safety and Quality: Microbial, Chemical, and Sensory. 2021:427-65.

https://doi.org/10.1007/978-3-030-68636-9_17

  1. Pascua Y, Koç H, Foegeding EA: Food structure: Roles of mechanical properties and oral processing in determining sensory texture of soft materials. Curr Opin Colloid Interface Sci. 2013; 18(4): 324-333.

https://doi.org/10.1016/j.cocis.2013.03.009

  1. Manfredi LR, Saal HP, Brown KJ, Zielinski MC, Dammann III JF, Polashock VS, Bensmaia SJ: Natural scenes in tactile texture. J Neurophysiol: 2014; 111(9): 1792-1802.

https://doi.org/10.1152/jn.00680.2013

  1. Lieber JD, Bensmaia SJ: The neural basis of tactile texture perception. Curr Opin Neurobiol. 2022; 76: 102621.

https://doi.org/10.1016/j.conb.2022.102621

  1. Patterson NM, Zhong Y, James BJ, Gant N, Hautus MJ. Effect of basic structural variation, aimed at increasing perceivable textures in model foods, on the perception of textural complexity. Food Qual Prefer. 2021; 104196.

https://doi.org/10.1016/j.foodqual.2021.104196

  1. Panda S, Chen J, Benjamin O: Development of model mouth for food oral processing studies: Present challenges and scopes. Innov Food Sci Emerg Technol. 2020; 66: 102524.

https://doi.org/10.1016/j.ifset.2020.102524

  1. De Pilli T, Alessandrino O, Baiano A: Quantitative Descriptive Analysis as a strategic tool in research activities relating to innovative meat tenderisation technologies. Heliyon. 2024; 10(11): e32618

https://doi.org/10.1016/j.heliyon.2024.e32618

  1. Chinnasami Sivaji MR, Saravanan V, Sharma R: The Applications of Nanotechnologies in Food Science using the ARAS Method. J Appl Chem Phys. 2024; 3:1.

https://doi.org/10.46632/jacp/3/1/2

  1. Rodriguez-Garraus A, Azqueta A, Vettorazzi A, Lopez de Cerain A: Genotoxicity of silver nanoparticles. Nanomaterials. 2020; 10(2): p.251.

https://doi.org/10.3390/nano10020251

  1. Jafroudi ZT, Ahari H, Haghdoost NS, Shabani S: Production of Nanocomposite Silver Packaging using Solution Blending Method for the Supplement of Antibacterial Coating. Appl Food Biotechnol. 2024; 11(1): 16-e16.

https://doi.org/10.22037/afb.v11i1.43876

  1. Yuan Y, Ma M, Xu Y, Wang D: Construction of biopolymer-based nanoencapsulation of functional food ingredients using the pH-driven method: A review. Crit Rev Food Sci Nutr. 2023; 63(22): 5724-5738.

https://doi.org/10.1080/10408398.2021.2023858

  1. Saxena A, Bhardwaj M, Allen T, Kumar S, Sahney R: Adsorption of heavy metals from wastewater using agricultural–industrial wastes as biosorbents. Water Sci. 2017; 31(2): 189-197.

https://doi.org/10.1016/j.wsj.2017.09.002

  1. Pathakoti K, Manubolu M, Hwang H-M: Nanostructures: Current uses and future applications in food science. J Food Drug Anal. 2017; 25(2): 245-253.

https://doi.org/10.1016/j.jfda.2017.02.004

  1. Degant O, Schwechten D: Wheat flour with increased water binding capacity and process and equipment for its manufacture. German Patent DE10107885A1, 2002.
  2. Hebishy E, Buffa M, Guamis B, Blasco-Moreno A, Trujillo A-J: Physical and oxidative stability of whey protein oil-in-water emulsions produced by conventional and ultra high-pressure homogenization: Effects of pressure and protein concentration on emulsion characteristics. Innov Food Sci Emerg Technol. 2015; 32: 79-90.

https://doi.org/10.1016/j.ifset.2015.09.013

  1. Ashaolu TJ: Nanoemulsions for health, food, and cosmetics: a review. Environ Chem Lett. 2021; 19(4): 3381-95.

https://doi.org/10.1007/s10311-021-01216-9

  1. Subramani T, Ganapathyswamy H: An overview of liposomal nano-encapsulation techniques and its applications in food and nutraceutical. J Food Sci Technol. 2020; 57(10): 3545-55.

https://doi.org/10.1007/s13197-020-04360-2

  1. Bastarrachea L, Dhawan S, Sablani SS: Engineering properties of polymeric-based antimicrobial films for food packaging: a review. Food Eng Rev. 2011; 3: 79-93.

https://doi.org/10.1007/s12393-011-9034-8

  1. Rhim J-W, Park H-M, Ha C-S: Bio-nanocomposites for food packaging applications. Prog Polym Sci. 2013; 38(10-11): 629-1652.

https://doi.org/10.1016/j.progpolymsci.2013.05.008

  1. Khosravi-Darani K, Bucci D: Application of poly (hydroxyalkanoate) in food packaging: Improvements by nanotechnology. Chem Biochem Eng Q. 2015; 29(2):275-285.

https://doi.org/10.15255/CABEQ.2014.2260

  1. Jafarzadeh S, Nafchi AM, Salehabadi A, Oladzad-Abbasabadi N, Jafari SM: Application of bio-nanocomposite films and edible coatings for extending the shelf life of fresh fruits and vegetables. Adv Colloid Interface Sci. 2021; 291: 102405.

https://doi.org/10.1016/j.cis.2021.102405

  1. Jafarzadeh S, Salehabadi A, Nafchi AM, Oladzadabbasabadi N, Jafari SM: Cheese packaging by edible coatings and biodegradable nanocomposites; improvement in shelf life, physicochemical and sensory properties. Trends Food Sci Tech. 2021; 116: 218-231.

https://doi.org/10.1016/j.tifs.2021.07.021

  1. Ahari H, Lahijani LK: Migration of silver and copper nanoparticles from food coating. Coatings. 2021; 11(4): 380.

https://doi.org/10.3390/coatings11040380

  1. Flores-López ML, Cerqueira MA, de Rodríguez DJ, Vicente AA: Perspectives on utilization of edible coatings and nano-laminate coatings for extension of postharvest storage of fruits and vegetables. Food Eng Rev. 2016; 8: 292-305.

https://doi.org/10.1007/s12393-015-9135-x

  1. Bhat R, Gómez-López VM: Practical food safety: Contemporary issues and future directions. 2014: John Wiley & Sons.
  2. Bahadır EB, Sezgintürk MK: Applications of commercial biosensors in clinical, food, environmental, and biothreat/biowarfare analyses. Anal Biochem. 2015; 478: 107-120.

https://doi.org/10.1016/j.ab.2015.03.011

  1. Nakagawa H, Hashimoto R, Matsuo Y, Sago Y, Yokoyama K, Takahashi H: Detection and determination of fumonisins B 1, B 2, and B 3 contaminating Japanese domestic wine by liquid chromatography coupled to tandem mass spectrometry (LC–MS/MS). Curr Microbiol. 2020; 77: 3057-3064.

https://doi.org/10.1007/s00284-020-02113-0

  1. Thangadurai D, Sangeetha J, Prasad R: Nanotechnology for food, agriculture, and environment. 2020; Springer.

https://doi.org/10.1007/978-3-030-31938-0

  1. Kakimova Z, Orynbekov D, Zharykbasova K, Kakimov A, Zharykbasov Y, Mirasheva G, Toleubekova S, Muratbayev A, Ntsomboh Ntsefong G: Advancements in nano bio sensors for food quality and safety assurance-a review. Potr. S. J. F. Sci. 2023; 17(1).

https://doi.org/10.5219/1903

  1. Yoo SM, Lee SY: Optical biosensors for the detection of pathogenic microorganisms. Trends Biotechnol. 2016; 34(1): 7-25.

https://doi.org/10.1016/j.tibtech.2015.09.012

  1. Chen H, Zhou K, Zhao G: Gold nanoparticles: From synthesis, properties to their potential application as colorimetric sensors in food safety screening. Trends Food Sci. Technol. 2018; 78: 83-94.

https://doi.org/10.1016/j.tifs.2018.05.027

  1. Liu G, Lu M, Huang X, Li T, Xu D: Application of gold-nanoparticle colorimetric sensing to rapid food safety screening. Sensors. 2018; 18(12): 4166.

https://doi.org/10.3390/s18124166

  1. Wang C, Qian J, Wang K, Yang X, Liu Q, Hao N, Wang C, Dong X, Huang X: Colorimetric aptasensing of ochratoxin A using Au@ Fe3O4 nanoparticles as signal indicator and magnetic separator. Biosens Bioelectron. 2016; 77:1183-1191.

https://doi.org/10.1016/j.bios.2015.11.004

  1. Luan Y, Chen J, Li C, Xie G, Fu H, Ma Z, Lu A: Highly sensitive colorimetric detection of ochratoxin A by a label-free aptamer and gold nanoparticles. Toxins. 2015; 7(12): 5377-5385.

https://doi.org/10.3390/toxins7124883

  1. Yuan J, Wu S, Duan N, Ma X, Xia Y, Chen J, Ding Z, Wang Z: A sensitive gold nanoparticle-based colorimetric aptasensor for Staphylococcus aureus. Talanta. 2014; 127: 163-168.

https://doi.org/10.1016/j.talanta.2014.04.013

  1. Kharlamova MV, Burdanova MG, Paukov MI, Kramberger C: Synthesis, sorting, and applications of single-chirality single-walled carbon nanotubes. Materials. 2022 Aug 26; 15(17): 5898.

https://doi.org/10.3390/ma15175898

  1. Hu J, Jiang YZ, Tang M, Wu LL, Xie HY, Zhang ZL, Pang DW: Colorimetric-fluorescent-magnetic nanosphere-based multimodal assay platform for Salmonella detection. Anal Chem. 2018; 91(1): 1178-1184.

https://doi.org/10.1021/acs.analchem.8b05154

  1. Park JH, Byun JY, Mun H, Shim WB, Shin YB, Li T, Kim MG: A regeneratable, label-free, localized surface plasmon resonance (LSPR) aptasensor for the detection of ochratoxin A. Biosens Bioelectron. 2014; 59: 321-327.

https://doi.org/10.1016/j.bios.2014.03.059

  1. He YQ, Gao Y, Gu HW, Meng XZ, Yi HC, Chen Y, Sun WY. Target-induced activation of DNAzyme for sensitive detection of bleomycin by using a simple MOF-modified electrode. Biosens Bioelectron. 2021; 178: 113034.

https://doi.org/10.1016/j.bios.2021.113034

  1. Qiao X, He J, Yang R, Li Y, Chen G, Xiao S, Huang B, Yuan Y, Sheng Q, Yue T: Recent advances in nanomaterial-based sensing for food safety analysis. Processes. 2022; 10(12): 2576.

https://doi.org/10.3390/pr10122576

  1. Dutta N, Kumar A, Kumari A, Maan S, Dutta G, Joshi VG: Antibody-based sensors for pathogen detection. InProtocols for the Diagnosis of Pig Viral Diseases New York, NY: Springer US. 2022; pp. 171-193.

https://doi.org/10.1007/978-1-0716-2043-4_12

  1. Xiang C, Li R, Adhikari B, She Z, Li Y, Kraatz H-B: Sensitive electrochemical detection of Salmonella with chitosan–gold nanoparticles composite film. Talanta. 2015; 140: 122-127.

https://doi.org/10.1016/j.talanta.2015.03.033

  1. Xiang C, Li R, Adhikari B, She Z, Li Y, Kraatz H-B: Nanomaterials-based aptasensors for rapid detection and early warning of key food contaminants: A review. Food Chem. 2024; 140990.

https://doi.org/10.1016/j.foodchem.2024.140990

  1. Hasan MR, Pulingam T, Appaturi JN, Zifruddin AN, Teh SJ, Lim TW, Ibrahim F, Leo BF, Thong KL: Carbon nanotube-based aptasensor for sensitive electrochemical detection of whole-cell Salmonella. Anal Biochem. 2018; 554: 34-43.

https://doi.org/10.1016/j.ab.2018.06.001

  1. Tang J, Tao X: Gold nanoparticle-based aptasensors for detecting kanamycin in foods: Recent advances and perspectives. Microchem J. 2023; 109313.

https://doi.org/10.1016/j.microc.2023.109313

  1. Magasinski A, Dixon P, Hertzberg B, Kvit A, Ayala J, Yushin GJ: High-performance lithium-ion anodes using a hierarchical bottom-up approach. Nat. Mater. 2010; 9:353–358.

https://doi.org/10.1038/nmat2725

  1. Zhang BT, Zheng X, Li HF, Lin JM: Application of carbon-based nanomaterials in sample preparation: A review. Anal Chim Acta. 2013; 784: 1–17.

https://doi.org/10.1016/j.aca.2013.03.054.

  1. Palani G, Arputhalatha A, Kannan K, Lakkaboyana SK, Hanafiah MM, Kumar V, Marella RK: Current trends in the application of nanomaterials for the removal of pollutants from industrial wastewater treatment—A review. Molecules. 2021; 26(9): 2799.

https://doi.org/10.3390/molecules26092799

  1. Shin WK, Cho J, Kannan AG, Lee YS, Kim DW: Cross-linked composite gel polymer electrolyte using mesoporous methacrylate-functionalized SiO2 nanoparticles for lithium-ion polymer batteries. Sci Rep. 2016; 6: 26332.

https://doi.org/10.1038/srep26332

  1. Javaid M, Haleem A, Singh RP, Rab S, Suman R: Exploring the potential of nanosensors: A brief overview. Sens. Int. 2021 Jan 1;2:100130.

https://doi.org/10.1016/j.sintl.2021.100130

  1. Bintsis T: Foodborne pathogens. AIMS Microbiol. 2017; 3(3): 529.

https://doi.org/10.3934/microbiol.2017.3.529

  1. Singh T, Shukla S, Kumar P, Wahla V, Bajpai VK, Rather IA: Application of nanotechnology in food science: perception and overview. Front Microbiol. 2017; 8: 1501.

https://doi.org/10.3389/fmicb.2017.01501

  1. Kavakebi E, Anvar AA, Ahari H, Motalebi AA: Green biosynthesized Satureja rechingeri Jamzad-Ag/poly vinyl alcohol film: quality improvement of Oncorhynchus mykiss fillet during refrigerated storage. Food Sci. Technol. 2021; 41(1): 267-278.  

https://doi.org/10.1590/fst.62720 

  1. Chorianopoulos N, Tsoukleris D, Panagou E, Falaras P, Nychas G-J: Use of titanium dioxide (TiO2) photocatalysts as alternative means for Listeria monocytogenes biofilm disinfection in food processing. Food Microbiol. 2011; 28(1): 164-170.

https://doi.org/10.1016/j.fm.2010.07.025

  1. Singh R, Mann B, Sharma R, Singh S: Application of Nanotechnology in Functional Foods, in Nanoscience for Sustainable Agriculture, R.N. Pudake, N. Chauhan, and C. Kole, Editors. Springer International Publishing: Cham. 2019; 547-579.

https://doi.org/10.1002/9781119905059

  1. Momin JK, Jayakumar C, Prajapati JB: Potential of nanotechnology in functional foods. Emir J Food Agric. 2013; 25(1): 10-19.

https://doi.org/10.9755/ejfa.v25i1.9368

  1. Singh H.: Nanotechnology Applications in Functional Foods; Opportunities and Challenges. Prev Nutr Food Sci. 2016; 21(1): 1-8.  

https://doi.org/10.3746/pnf.2016.21.1.1

  1. Sabliov C, Chen H, Yada R: Nanotechnology and functional foods: effective delivery of bioactive ingredients. John Wiley & Sons. 2015.

https://doi.org/10.1002/9781118462157

  1. Nedovic V, Kalusevic A, Manojlovic V, Levic S, Bugarski B: An overview of encapsulation technologies for food applications. Procedia Food Sci. 2011; 1: 1806-1815.

https://doi.org/10.1016/j.profoo.2011.09.265

  1. Akhavan S, Assadpour E, Katouzian I, Jafari SM: Lipid nano scale cargos for the protection and delivery of food bioactive ingredients and nutraceuticals. Trends Food Sci. Technol. 2018; 74: 132-146.

https://doi.org/10.1016/j.tifs.2018.02.001

  1. Sekhon BS: Food nanotechnology – an overview. Nanotechnol Sci Appl. 2010. 3(null):nanofilteration1-15.
  2. Bhattacharya T, Ahmed S: Nanotechnology in Functional Foods. 2022: Wiley.

https://doi.org/10.1002/9781119905059

  1. Kanugo A, Goyal R, Sharma S, Gautam RK: Recent Advancement of Nanotechnology in Functional Foods, in Nanotechnology in Functional Foods. 2022; 293-328.

https://doi.org/10.1002/9781119905059.ch12

  1. Morya S, Awuchi CG, Menaa F: Advanced functional approaches of nanotechnology in food and nutrition. In Environmental Management Technologies. 2022; pp. 257-272).

https://doi.org/10.1201/9781003239956  

  1. Kaya-Celiker H, Mallikarjunan K: Better nutrients and therapeutics delivery in food through nanotechnology. Food Eng Rev. 2012; 4: 114-123.

https://doi.org/10.1007/s12393-012-9050-3

  1. McClements DJ: Modeling the rheological properties of plant‐based foods: Soft matter physics principles. Sustain Food Proteins. 2023; 1(3):101-132.

https://doi.org/10.1002/sfp2.1015

  1. Puri V, Nagpal M, Singh I, Singh M, Dhingra GA, Huanbutta K, Dheer D, Sharma A, Sangnim T: A comprehensive review on nutraceuticals: therapy support and formulation challenges. Nutrients. 2022; 14(21): 4637.

https://doi.org/10.3390/nu14214637

  1. Duncan TV: Applications of nanotechnology in food packaging and food safety: barrier materials, antimicrobials and sensors. J. Colloid Interface Sci. 2011; 363(1): 1-24.

https://doi.org/10.1016/j.jcis.2011.07.017

  1. Altemimi AB, Farag HA, Salih TH, Awlqadr FH, Al-Manhel AJ, Vieira IR, Conte-Junior CA: Application of Nanoparticles in Human Nutrition: A Review. Nutrients, 2024; 16(5): 636.

https://doi.org/10.3390/nu16050636

  1. Vieira IRS, Tessaro L, Lima AKO, Velloso IPS, Conte-Junior CA: Recent progress in nanotechnology improving the therapeutic potential of polyphenols for cancer. Nutrients. 2023; 15(14): 3136.

https://doi.org/10.3390/nu15143136

  1. Centurion F, Basit AW, Liu J, Gaisford S, Rahim MA, Kalantar-Zadeh K: Nanoencapsulation for probiotic delivery. ACS Nano. 2021; 15(12): 18653-18660.

https://doi.org/10.1021/acsnano.1c09951

  1. Ghorbani S, Maryam A: Encapsulation of lactic acid bacteria and Bifidobacteria using starch‐sodium alginate nanofibers to enhance viability in food model. J. Food Process. Preserv. 2021; 45(12): 16048.

https://doi.org/10.1111/jfpp.16048

  1. Teng Z, Luo Y, Wang Q: Nanoparticles synthesized from soy protein: preparation, characterization, and application for nutraceutical encapsulation. J Agric Food Chem. 2012; 60(10): 2712-2720.

https://doi.org/10.1021/jf205238x 

  1. Arroyo-Maya IJ, McClements DJ: Biopolymer nanoparticles as potential delivery systems for anthocyanins: Fabrication and properties. Food Res Int. 2015; 69: 1-8.

https://doi.org/10.1016/j.foodres.2014.12.005

  1. Ishak K, Annuar MM, Ahmad N: Nano-delivery systems for nutraceutical application, in Nanotechnology applications in food. 2017; 179-202.

https://doi.org/10.1016/B978-0-12-811942-6.00009-1

  1. Assadpour E, Jafari SM: Importance of release and bioavailability studies for nanoencapsulated food ingredients, in Release and bioavailability of nanoencapsulated food ingredients. 2020; 1-24.

https://doi.org/10.1016/B978-0-12-815665-0.00001-1

  1. Perry SL, McClements DJ: Recent advances in encapsulation, protection, and oral delivery of bioactive proteins and peptides using colloidal systems. Molecules. 2020; 25(5): 1161.

https://doi.org/10.3390/molecules25051161

  1. Rostamabadi H, Falsafi SR, Boostani S, Katouzian I, Rezaei A, Assadpour E, Jafari SM: Design and formulation of nano/micro-encapsulated natural bioactive compounds for food applications, in Application of nano/microencapsulated ingredients in food products. 2021; 1-41.

https://doi.org/10.1016/B978-0-12-815726-8.00001-5

  1. Gorantla S, Wadhwa G, Jain S, Sankar S, Nuwal K, Mahmood A, Dubey SK, Taliyan R, Kesharwani P, Singhvi G: Recent advances in nanocarriers for nutrient delivery. Drug Deliv. and Transl. Res. 2021; 1-26.

https://doi.org/10.1007/s13346-021-01097-z

  1. Pateiro M, Gómez B, Munekata PE, Barba FJ, Putnik P, Kovačević DB, Lorenzo JM: Nanoencapsulation of promising bioactive compounds to improve their absorption, stability, functionality and the appearance of the final food products. Molecules. 2021; 26(6): 1547.

https://doi.org/10.3390/molecules26061547

  1. Venugopalan VK, Gopakumar LR, Kumaran AK, Chatterjee NS, Soman V, Peeralil S, Mathew S, McClements DJ, Nagarajarao RC: Encapsulation and protection of omega-3-rich fish oils using food-grade delivery systems. Foods. 2021; 10(7): 1566.

https://doi.org/10.3390/foods10071566

  1. Bratovcic A, Suljagic J: Micro-and nano-encapsulation in food industry. Croat. J. Food Sci. Technol. 2019; 11(1): 113-121.

https://doi.org/10.17508/CJFST.2019.11.1.17

  1. Boostani S, Jafari SM: A comprehensive review on the controlled release of encapsulated food ingredients; fundamental concepts to design and applications. Trends Food Sci Technol. 2021; 109: 303-321.

https://doi.org/10.1016/j.tifs.2021.01.040

  1. Sonea C, Lupusoru M, Opris I: Augmentation of Nutrition by Nanotechnology. Modern Approaches to Augmentation of Brain Function. 2021; 415-431.

https://doi.org/10.1007/978-3-030-54564-2_19

  1. Campuzano S, Barderas R, Moreno-Casbas MT, Almeida Á, Pingarrón JM: Pursuing precision in medicine and nutrition: the rise of electrochemical biosensing at the molecular level. Anal Bioanal Chem. 2024; 416(9): 2151-2172.

https://doi.org/10.1007/s00216-023-04805-5

  1. de Toro-Martín J, Arsenault BJ, Després JP, Vohl MC: Precision Nutrition: A Review of Personalized Nutritional Approaches for the Prevention and Management of Metabolic Syndrome. Nutrients. 2017; 9(8).

https://doi.org/10.3390/nu9080913

  1. Gan J, Siegel JB, German JB: Molecular annotation of food - towards personalized diet and precision health. Trends Food Sci Tech. 2019; 91: 675-680.

https://doi.org/10.1016/j.tifs.2019.07.016

  1. Laddu D, Hauser M: Addressing the Nutritional Phenotype Through Personalized Nutrition for Chronic Disease Prevention and Management. Prog Cardiovasc Dis. 2019; 62(1): 9-14.

https://doi.org/10.1016/j.pcad.2018.12.004

  1. Fenech M, El-Sohemy A, Cahill L, Ferguson LR, French TA, Tai ES, Milner J, Koh WP, Xie L, Zucker M, Buckley M: Nutrigenetics and Nutrigenomics: Viewpoints on the Current Status and Applications in Nutrition Research and Practice. J Nutrigenet Nutrigenomics. 2011; 4(2): 69-89.

https://doi.org/10.1159/000327772

  1. Jiang T, Liao W, Charcosset C: Recent advances in encapsulation of curcumin in nanoemulsions: A review of encapsulation technologies, bioaccessibility and applications. Food Res Int. 2020; 132:109035.

https://doi.org/10.1016/j.foodres.2020.109035

  1. McClements DJ: The future of food colloids: Next-generation nanoparticle delivery systems. Curr Opin Colloid Interface Sci. 2017; 28: 7-14.

https://doi.org/10.1016/j.cocis.2016.12.002

  1. Yu X, Abd El-Aty AM, Su W, Tan M: Advancements in precision nutrition: Steady-state targeted delivery of food functional factors for nutrition intervention of chronic diseases. Food Safety and Health. 2023; 1(1): 22-40.  

https://doi.org/10.1002/fsh3.12006

  1. McClements DJ: Nano-enabled personalized nutrition: Developing multicomponent-bioactive colloidal delivery systems. Adv. Colloid Interface Sci. 2020; 282: 102211.

https://doi.org/10.1016/j.cis.2020.102211

  1. Dalal MS: Chapter-4 Nanotechnology for Nutrigenomics.
  2. Sharma H, Kumar A, Singh R, Malik A, Juyal R: Enhancing food security and sustainability: harnessing nano-omics-based technologies for quality monitoring. Sustainable Agriculture: Nanotechnology, Biotechnology, Management and Food Security. 2024; 191.

https://doi.org/10.1515/9783111369754-009

 

 

 

 

Assessing Efficacy of the Microbial Interventions in Heavy Metal Decontamination of the Environment and Food Production Systems

Fatemeh Fateminasab, Mehdi Koushki, Faranak Jafarian, Vahid Mansouri, Alireza Ahmadzadeh, Nasrin Amiri-Dashatan, Masoumeh Farahani

Applied Food Biotechnology, Vol. 11 No. 2 (2024), 26 Aban 2024, Page e8
https://doi.org/10.22037/afb.v11i2.46832

Background and Objective: Heavy metals such as arsenic (As), cadmium (Cd), chromium (Cr), mercury (Hg), Copper (Cu), Nickel (Ni), and lead (Pb) pose serious risks to ecosystems and human health, particularly affecting vulnerable groups such as children. These toxic heavy metals can accumulate in food matrices, leading to long-term health effects. People can be exposed to these pollutants through contaminated food, polluted water, inhalation, and direct skin contact. This review focused on the environmental pollution caused by industrialization, specifically heavy metal contamination. Moreover, it assessed bioremediation as a potential solution, which involved using microorganisms and plants to break down and detoxify these hazardous pollutants.

Results and Conclusion: This study investigated various bioremediation techniques, including the use of probiotics for decontamination. These methods aimed to restore contaminated sites and develop sustainable approaches to remove heavy metals from food products. The study highlighted the effectiveness of microbial interventions in addressing the harmful effects of heavy metals and toxins in the environment and food systems. These bioremediation strategies could lessen the risks associated with heavy metal contamination.

Conflict of interest: The authors declare no conflict of interest.

1. Introduction

The environment encompasses all natural components, including soil, water, and essential air for sustaining life on the planet. However, rapid industrialization over the past century has led to increased pollution levels and significant environmental challenges. Pollutants occur in various forms such as organic materials, plastics, and inorganic substances, including heavy metals (HM) and radioactive wastes [1]. People can be exposed to HMs in four major ways eating contaminated foods, breathing polluted air, drinking dirty water, and touching contaminated surfaces in farms, factories, homes, and industrial areas [2]. Metals can be stored in food matrices and at low concentrations can lead to long-term health problems, especially in children. The spread HMs are arsenic (As), cadmium (Cd), chromium (Cr-VI), mercury (Hg), copper, (Cu), nickel (Ni), and lead (Pb), where they are broadly known as “most problematic HMs” and “toxic heavy metals” (THM) [3]. The HMs such as Pb and Hg can accumulate in the food chain, posing threats to wildlife, plants, and humans. Long-term consequences of pollution extend beyond environmental damages; they destroy habitats, decrease biodiversity, and disrupt ecosystems. Furthermore, pollution includes health risks, contributing to respiratory ailments and cardiovascular diseases. It is critical to tackle these pollution challenges to protect the environment and public health [4]. The HMs are dangerous to food chains and can cause long-term health problems in people. These metals and other pollutants do not break down easily; hence, they persist in the environment consistently. To tackle these harmful substances, the use of biological methods is needed. Researchers study how microbes interact with HMs to create safe and sustainable cleanup methods such as bioremediation [5].Bioremediation is a process that uses living organisms, mainly microorganisms and plants, to clean up environ-mental pollutants. The goal is to break down harmful substances in waste and make them less toxic or harmless [6]. This helps restore contaminated sites to their original condition. Bioremediation can decrease the effects of various pollutants, including pesticides, industrial solvents, HMs, and other chemical compounds [7]. The field covers basic research such as studying how organisms break down substances and practical uses such as assessing pollution and predicting how it behaves in the environment. Methods such as probiotics effectively clean toxins and HMs from foods [8,9]. These methods can remove harmful elements such as As, Hg, Pb, and Cd and pesticides. Bioremediation techniques can be used to clean water, soil, lagoons, waste streams, and sludge. Using natural and specially designed microorganisms with appropriate engineering, correct conditions for effective bioremediation can be created [10,11]. Transferring research findings from the laboratory to industrial uses is usually slow and the outcomes can vary significantly from those seen under controlled labor-atory conditions. Hence, only a few industrial methods for HM bioremediation have successfully been adopted. This challenge has shifted future studies on technology transfer toward assessing potentials for scaling these processes and verifying the effectiveness of the linked systems. To successfully implement bioremediation at a larger scale, it is critical to investigate biochemical and physical factors such as compositions of the media and growth conditions [12]. As the global population grows and living standards improve, demands for agricultural production are expected to double by 2050. However, industrial growth in developing nations has resulted in significant contamination of farmland with HMs. Key HMs, including Cd, As, Cu, Hg, Pb, and Cr, are identified as major contributors to soil pollution in China, increasing international awareness of soil degradation through initiatives such as "World Soil Day" and the "International Year of Soils" in 2015. In 2017, the UN Environment Assembly called World Health Organization (WHO) and Food and Agriculture Organization (FAO) to report on global soil pollution and assess risks by 2021. China's strategic plan established in 2016 has aimed to rehabilitate 700,000 hectares of polluted agricultural lands by 2020, targeting to restore and certify 95% of the nation’s contaminated lands as safe for agricultural use by 2030 [13]. This report aims to highlight the urgent need to address increasing levels of pollution caused by industrialization and assess their significant effects on environmental health, the food industry, and public safety. Additionally, this study aimed to investigate the potential of bioremediation as an innovative solution for using microorganisms to effectively clean up hazardous pollut-ants; thereby, helping in restoring the contaminated eco-systems.

2. Results and Discussion

2.1. Toxic heavy metals

The presence of HMs such as Cd, Cr, Pb, Hg, As, Cu, and Ni in wastewater poses significant risks to human health and aquatic life. Developing countries are parti-cularly affected by HM contamination, which increases environmental, health, and economic concerns. Exposure to these metals can damage the central nervous system, decrease energy levels, and hurt vital organs. Long-term exposure can cause degenerative processes similar to neurological disorders such as Alzheimer's and Parkinson's diseases [14].

Cd, a rare metal extensively used in various industries, includes significant health hazards, including damage to vital organs and the development of conditions such as Itai-Itai disease. Mining and refining activities, air pollution, and the use of sewage sludge are the primary contributors to water and soil contaminations with Cd. Exposure to Cd primarily occurs through the consumption of contaminated foods and water, inhalation, and tobacco smoke. The metal can bioaccumulate in plants and animals and has been associated with various forms of cancers and osteoporosis. Even at low exposure levels, typical in the general non-smoking populations, Cd can result in renal tubular dysfunction and bone toxicity. Recent studies have suggested correlations between increased Cd exposure and hormone-linked cancers such as breast cancer, as well as effects on neurodevelopment. Plants absorb Cd through their root systems, while grain and vegetable products serve as the primary diet sources. Special attention should be paid to vulnerable groups, including infants, vegetar-ians, smokers, and fertile women with low-iron status faci-litating gastrointestinal absorption of Cd and individuals with preexisting renal dysfunction. Moreover, Cd is highly toxic to humans can cause reproductive system failure and brain and kidney damage, and is linked to cancers. It can enter the food chain and accumulate in organisms. The study aimed to use natural microorganisms to completely remove Cd from the environment [14, 15].

Cr, showing various oxidation states from -2 to +6, is used in industrial processes such as metal refining, chrome-plating, and stainless-steel production. This results in the release of Cr(VI) into the atmosphere, leading to potential exposure through polluted air or contaminated drinking water. The International Agency for Research on Cancer categorizes Cr(III) & (VI) as Group 1 carcinogens, highlighting their significant health hazards. Cr(III) occurs naturally in rocks and soil and can enter human diets through plant intakes. Toxic effects of Cr are due to dermal and respiratory exposures. Furthermore, Cr(VI) is an extre-mely sensitized agent that causes skin allergies [16,17].

Pb, a toxic metal, is commonly detected in soil due to human activities and natural sources. Its presence in soil poses a significant risk to human health, especially through food consumption, with cereals and vegetables are the primary sources of exposure. Despite regulatory efforts in developed nations to control and prohibit the use of Pb in gasoline and certain materials since the 1970s, concerns about Pb exposure from foods are still reported. Exten-sively, Pb is used in various industries, including batteries, paints, automobile fuels, semiconductors, and electro-plating. Prolonged exposure to Pb can lead to serious health problems such as nephropathy, abdominal pain, weakness, and male infertility. It is present in dust, soil, old paints, and leaded gasoline, affecting behaviors, cognitive performance, growth, and overall health. Histo-rical and continued uses of Pb have resulted in its presence in the food chain. Urban agriculture in developing nations, critical for food security, may contribute to higher Pb levels in leafy vegetables due to land shortage and previous waste disposal. While adults primarily encounter Pb exposure through foods, infants are majorly exposed through drinking water, soil ingestion, and dust. Pb in drinking water often originates from water pipes or Pb solders in taps. Additionally, certain shellfish, chocolate products, sweet potatoes, canned fruits, and hot sauces imported from Mexico may contain increased Pb concentrations. Exposure to Pb has been associated with a variety of adverse effects on the cardiovascular system in adults. The effect associated with the lowest Pb exposure includes increases in systolic blood pressure. Polymor-phisms in aminolevulinic dehydratase gene may play a role in mediating these effects. Relatively low exposure to lead has been associated with functional renal deficits such as decreases in glomerular filtration rate, increased serum creatinine levels, and decreased creatinine clearance [15,18].

Technically, Zn belongs to Group II–B in the periodic table. It is relatively abundant in the earth crust. Furthermore, Zn is a bluish-white lustrous metal, very malleable and moderately reactive that combines with oxygen and other non–metals. It only includes one common oxidation state (+2) and is widely used in corro-sion-resistant steel coatings, brass alloys, painting pigments, wood preservatives, dry-cell batteries, cosmet-ics, and pharmaceuticals. The industry uses a large quantity of Zn to produce die-casting, which contributes to emissions to the atmosphere water, and soil. The principal source of Zn contamination in the environment involves industrial wastes, metal plating, and acid drainage. Sewage and animal wastes constitute a source of Zn in the soil. Additionally, Zn is in contrast to the other two HMs discussed previously; it is an essential trace element for animals, plants, and bacteria necessary for life. It plays an important role in the catalytic activity of proteins. Shortage can cause hair loss, skin lesions, and diarrhea in humans, while in plants it can cause chlorosis, impaired growth, and malformation of leaves and stems. Excess exposures to Zn can lead to loss of weight, impaired immune system, and blockage of essential reactions in the cell [14].

Hg, the only liquid metal, is one of the most THMs in three distinct forms elemental, inorganic, and organic, each with various toxicity levels. Moreover, Hg includes signifi-cant industrial uses due to its ability to form amalgams with other metals (e.g. in gold extraction or dental amal-gams). The inorganic forms can present health risks to individuals through occupational exposure, while methyl Hg, the most prevalent organic form, can be harmful when ingested through the diet. Furthermore, Hg is released into the environment through natural human activities such as coal combustion and industrial processes. The acceptable levels of Hg in drinking water and soil are 0.002 mg.l-1 and 0.010-0.3 mg.kg-1, respectively. Hg is extensively used in various industries such as electronics, dentistry, wood processing, and pharmaceuticals, emphasizing the import-ance of responsible use and disposal to minimize its environmental effects and safeguard human health. These compounds include diverse effects on the lungs, kidneys, nerves, skin, eyes, and digestive and immune systems. Hg can cause the immune system to attack the body. Severe hopelessness, extreme fatigue, insomnia, hair loss, vision impairment, memory loss, restlessness, tremors, sudden anger outbursts, brain injuries, and lung and kidney damage are possible consequences of certain contamin-ations [19, 20].

As is a dangerous toxin detected in groundwater and an industrial pollutant. The permissible limit of As in primary drinking water and soils is 0.05 mg.l-1 and 150 mg.kg-1, respectively. This toxic metal can affect humans through drinking water, inhalation of polluted air, and intake of contaminated foods. Long-term exposure to As can cause cardiovascular and blood diseases, neurotoxicity and nephrotoxicity, dermatitis, and various types of cancers. More than 200 million people worldwide are exposed to its chronic effects. Treatment methods for arsenicosis include chelation therapy, phytopreparations, antioxidants, and appropriately selected diets. Exogenous antioxidants such as Zn and Sn are convenient for As detoxification. In addition, it is noteworthy that the toxic effects of As adversely affect vital cellular functions such as oxidative phosphorylation and ATP synthesis [19–21].

Cu is essential for enzymes and electron transport but can be harmful at higher concentrations. The acceptable limits for Cu in primary and secondary drinking waters and soils are 1.3 mg.l-1, 1.0 mg.l-1, and 2100 mg.kg-1, respect-ively. Exceeding these limits can lead to adverse health effects. Cu is released into the environment from various sources and can exist in various forms in water. Microorganisms have developed mechanisms to maintain Cu balance, including sequestration, absorption, and efflux. Genes linked to resistance to Cu may be detected on plasmids and chromosomes [19, 22].

Ni, a strong, shiny white metal in the form of Ni2+ cations, is commonly detected in a state and used in various uses such as stainless-steel production, catalysis, and coin production. Ni is naturally present in small quantities in soils and essential for the metabolic activities of plants and many bacteria. However, some Ni and Pb compounds can be poisonous to health problems, including cancers, respiratory problems, and heart diseases. Continuous exposure to allergens through inhalation can result in various severe health problems such as allergic skin conditions, respiratory cancers, and negative effects on fertility and hair. These allergens have been identified as immunotoxic, neurotoxic, and genotoxic agents, presen-ting a significant threat to overall health. The allowed quantity of Ni in soil ranges from 5-500 mg.kg-1. Additionally, Ni can enter surface water through waste-water drains. While animals need Ni in small quantities, exceeded quantities can be toxic. Ni is essential for certain bacterial enzymes, but excessive Ni can be poisonous by binding to proteins and nucleic acids, often inhibiting enzyme activity as well as DNA replication, transcription and translation [19,20,22]. Table 1 summarizes THM characteristics.

2.2. Roles of biotechnology in bioremediation

Bioremediation refers to the removal of environmental pollutants from air, water, soil and flue gasses in natural or artificial settings [3]. Thus, bioremediation, a state-of-the-art and environmentally sustainable technology, uses organic microorganisms to remove pollutions. Hence, biotechnology is a method of using scientific engineering of microorganisms to improve the efficiency of organisms to serve humans and remediate environmental toxic substances [23]. A significantly developed environmen-tally biotechnol-ogical technology is necessary to remove the contaminated materials and reestablish the natural resources. Nowadays, recent advancements in the emerging novel methods of biotechnology support the bioremediation process for the protection of the natural environment through recycling waste materials. Later, use of the most advanced technology to boost the bioremedi-ation process is discussed. One of the biotechnology approaches includes use of genetic engineering. For example, it is possible to create an organism or genetically engineered microbes (GEM) that produce all the necessary enzymes or use all the degradation pathways for the bioremediation processes [24]. Thus, use of engineered microorganisms is more cost-effective than that use of alternative methods is. Usually, GEMs are created by introducing stronger proteins into bacteria through biotechnology or genetic engineering to enhance a desired trait. Biodegradation of oil spills, halobenzoates naphtha-lenes, toluenes, trichloroethylene, octanes and xylenes has been carried out via GEMs such as bacteria, fungi and algae [25]. Deterioration of polychlorinated biphenyls (PCB) is controlled by groups of genes that were observed in the genetic materials of two various organisms. Use of genetic engineering for achieving recombination between Pseudomonas pseudoalcaligenes KF707 and Burkholderia cepacia LB400 bph genes may increase the degradation rate of PCBs and stimulate remediation of toluene and benzene [25]. Another use of biotechnology is the blending between metallothionein (MT) isolated from rats, IgA protease protein isolated from Neisseria gonorrhoeae and lpp-ompA fusion vehicle to create bacterial cell walls with metal ion-binding polypeptides [26]. Another branch of biotechnology concerns the use of transgenic plants in bioremediation processes by introducing genes of interest from various sources (other plants, microorganisms, or animals) to improve the plant ability to remove toxic pollutants [27]. Thus, this translocation process enhances the phytoremediation capacity of plants. Biotechnolog-ically induced microorganisms are stronger than naturally occurring ones and may degrade contaminants faster because they can rapidly accommodate new pollutants they encounter or co-metabolize.

2.3. Cleaning up of the heavy metal contamination

HMs are naturally occurring elements that can enter the human food supply through industrial and agricultural activities, posing health risks due to their accumulation in living organisms. These metals are persistent pollutants that need regulation in water and food sources. The industrial wastewater release, containing HMs, is dangerous and traditional remediation methods are often ineffective and costly. While developed countries have regulations, several developing nations lack adequate frameworks to address the issue. Innovative solutions such as bioremediation with microorganisms are urgently needed to combat HM pollutions in water and soil [28, 29]. Specific microbial species help in environmentally sustainable cleanup and enhance human health. Probiotics such as lactic acid bacteria (LAB), Bifidobacteria and Saccharomyces boulardii detoxify HMs through attach-ment, precipitation and complex formation. Gram-positive bacteria, particularly Bacillus, are effective in this process, while Gram-negative bacteria are less effective. In addition, LAB can bioadsorb HMs and convert toxic elements into safer forms such as transforming methylated Hg. Studies show that Lactobacilli can convert Cr into less harmful forms, binding to HMs at the cell wall, regardless of their viability [28]. Probiotics, particularly strains such as Lactobacillus and Bifidobacterium, are effective in protecting foods from HMs and offer health benefits such as treating diseases, improving metabolism, and enhancing immune function. They can decrease HM toxicity by binding and adsorbing metals affected by pH levels with strains such as L. rhamnosus showing protective effects against HM poiso-ning, especially in vulnerable popul-ations. Additionally, probiotics may improve cognitive function and lessen oxidative stress in animal models. These bacteria present a promising cost-effective strategy for detoxifying HMs in foods and individuals [29, 30].

In a review, Goyal et al. (2019) investigated microbial-assisted remediation of HMs as a cost-effective approach, highlighting Bacillus genus for its significant biochemical capabilities in the bioremediation of Cd2+ and Ni2+. Bacillus achieves this through processes such as biosor-ption and bioaccumulation. Additionally, it has demons-trated potential to decrease toxicity of these metals in eukaryotic cells and mice, similar to the effects observed with probiotic genera such as Lactobacillus and Bifidobacterium [31]. Hadiani et al. (2018) showed that S. cerevisiae removed 88.9% of Hg²⁺ ions from water at pH 5.45 and an initial concentration of 79.8 µg/l, demonst-rating its potential for low Hg treatments. A 2019 study detected optimal conditions for As bioremediation at pH 5.0, achieving peak bioabsorption efficiencies of 66.2% for As(III) and 15.8% for As(V) with a concentration of 95.0 µg/l and inoculum size of 32.5 × 10⁷ cfu. Massoud et al. (2019) reported 70% Pb removal from milk and similar results were detected for Cd in 2020, indicating S. cerevisiae effectiveness against various HMs [29, 32]. A study by Ameen et al. (2020) showed that L. plantarum, a metal-resistant LAB from Alexandria's Mediterranean Seacoast, Egypt, effectively removed Ni2+ and Cr2+. Optimal Cr removal occurred at 100 ppm Cr2+ with a 3% inoculum, while Cd and Pb uptake were highest at pH 2.0 and 22 °C within 1 h. Its biosorption behavior followed the Langmuir isotherm model, making L. plantarum a promising solution for treating HM-contaminated waste-water [33]. In 2022, Afsharian et al. studied L. acidophilus ATCC 4356 ability to remove water contaminants in microgravity. They detected that heat pretreatment improved As absorption, while sodium hydroxide treatment was more effective for Cd and Pb without affecting Hg removal. The study highlighted that metal complexes with L. acidophilus were released under normal gravity and overall simulated microgravity enhanced the strain's HM bioreduction [34]. Traditional methods can be costly and ineffective, increasing the interest in eco-friendly options. Recent studies by Wahid et al. (2023) highlighted the effectiveness of probiotic bacteria, particularly Pediococcus pentosaceus, in metal removals and pathogen decreases. Two yogurt strains, L. acido-philus, and L. plantarum, were assessed for survival in acidic environments. The two survived simulated gastric conditions, with L. acidophilus showing antibiotic resist-ance except to gentamicin and co-amoxiclav. L. plantarum included a higher Pb removal rate of 42.70% for HMs than L. acidophilus of 35.50%. Cd removal rates were 11.50% for L. plantarum and 3.50% for L. acidophilus, indicating the strains' bioremediation potentials [35]. Mushtaq et al. (2023) investigated the probiotic characteristics of LAB capable of biosorbent Pb, a toxic pollutant. Eight strains from carnivores’ feces and human breast milk achieved a 70% Pb biosorption rate and were still viable under simulated gastrointestinal conditions. Significantly, Levila-ctobacillus brevis was analyzed, revealing optimal biosorption conditions at pH 6 after 60 m with a cell density of 1 g/l [36]. Salavatifar and Khosravi (2024) studied how microgravity affected HM biosorption by S. cerevisiae. They detected that microgravity increased biosorption rates for Hg (97%) and Pb (72.5%) but not for Cd or As. Metal-yeast complexes were still stable in gastrointestinal conditions with Pb binding decreasing initially in gastric conditions but recovering in the intestinal environment. The kinetics followed a pseudo-second-order model and all metals fitted the Langmuir isotherm. These results suggest that microgravity could enhance biosorption efficiency, benefiting detoxification in the food industry and astronaut health [37].

2.4. Microbial-based removal techniques (biorem-ediation)

 Currently, the quantity of toxins in the environment (air, water, and soil) is increasing, and traditional methods of controlling these pollutants are ineffective. Thus, improvements in modern remediation techniques can be an opportunity to reform bioremediation and waste removal from the environment [25]. Relatively, microbial techni-ques and methods evolved for achieving successful biorem-ediation with engineered microorganisms. For example, recombination in E. coli is introduced by insertion or deletion to improve the bioremediation characteristics of the microorganism [38]. Bioremediation of wastes using recombinant microbes is expensive but effectively removes all the wastes from the environment. Compared to other physical and chemical approaches, the use of microbial metabolism capabilities to deteriorate or eliminate environmental toxins is an economically secure option. These manipulated microorganisms are more effective than natural strains due to their higher degradation capacity and ability to promptly adapt to various pollutants such as substrates or cometabolites [39]. As previously stated, numerous techniques are available to treat this microbial contamination from water, soil, and other environments. For example, microbes decreasing HMs and improving soil fertility and plant development have made them a preferred source for bioremediation. However, climate statuses can be effective in the degradation of HMs by the microbes. Higher humidity maintains anaerobic conditions and slows the rate of decomposition. In cold statuses, microbial decomposition of HMs is slow, as metabolic activities are inhibited as the microbial transport pathways are frozen by sub-zero water [40]. While, in hot climates, the rate of HM solubility increases, which increases their availability and the rate of microbial biodegradation [41]. However, the power of microbial biodegradation is calculated by the metal or pollutant chemical structure, concentration, bioavailability, toxicity, and stability [42]. Later, microbial-based pollutant removal techniques are discussed. Recombinant micro-organisms and genetically modified microbes have been used as an effective technique for the breakdown of pollution. Recombinant E. coli is one of the model organisms used in bioremediation. E. coli is known as a “working horse” of molecular biology for its rapid growth rate in defined chemical environments and the diverse tools available for its genetic modifications [43]. Technically, E. coli includes favorite cloning genes and can introduce DNA molecules into cells. Due to the functions of E. coli for rapid growth and high-level protein expression, the micro-organism leads to protein production [38]. Several studies have shown that enteric E. coli develops phenol and p-cresol when grown on natural media (peptone and casein media) [44]. Further studies emphasized the ability of E. coli to break down and use a diverse range of aromatic acids such as phenylacetic acid (PA), hydroxyphenylacetic acid (HPA), and phenyl propionic acid (PP) [39]. Genetically modified E. coli is used as a highly effective agent in bioremediation processes. The introduction of a gene into bacteria results in the conversion of the bacteria into a distinct strain that can effectively remove hydrocarbon pollutants from the surrounding environment. This system is capable of removing Ni from the aqueous system, reducing Cr(IV) to Cr(III) as an important reaction in the bioremediation of hazardous Cr species in aerobic environmental statuses [45]. These highlights showed that E. coli is useful for the degradation of HMs. The persistent presence of HM contamination in the ecosystem has become a significant global concern, posing risks to environmental integrity and human health due to their tendency to bioaccumulate and resistance to biodegradation. Numerous microbes belonging to various taxonomic categories, including bacteria, fungi, and algae, have been documented to enzymatically transform metal ions into less harmful forms or to remove them from the environment in an environ-mentally sustainable way. Therefore, microbial species include important roles in the detoxication of HMs and toxins in food through bioremediation. Microbial species are used for bioremediation of water, soil, and air from HMs [46].

Water is essential for the sustenance of life on the planet and it is vital to eliminate any form of contaminations from this precious resource. Demands for water have surged significantly because of industrialization, primarily due to its essential role in industrial production processes [25]. Thus, microbial techniques are needed to remove HMs from polluted water and contaminated soil. Biofilm-mediated bioremediation presents a compelling strategy for the removal of environmental pollutants due to its significant adaptability, substantial biomass and superior ability to absorb, immobilize and degrade contaminants. Biofilms are aggregates of single or mixed microbial cells that adhere to a living cell or are inert to a surface in an aqueous environment [47]. Microorganisms that form biofilms demonstrate significant resilience when subjected to severe environmental stressors. They are capable of competing effectively for nutrients, show enhanced tolerance to pollutants in comparison to free-floating planktonic cells, and create a protective habitat for other cells. Biofilm communities possess the ability to adsorb and metabolize organic pollutants and HMs, facilitated by a precisely regulated gene expression pattern that is affected by quorum sensing [47]. Contrary to organic pollutants, HM remediation is distinctive as biological processes cannot eliminate these contaminants from the environment. Naturally, HMs are detected in the environment as minor constituents of geochemical cycles, suggesting that microbial communities may interact with and use these metals. When HMs are introduced as pollutants, their concentrations frequently exceed natural levels. There is a possibility of the occurrence of mixed contaminations [48]. However, several studies have demonstrated the ability of the biofilms to repopulate after subsequent desorption cycles with the removal of Cu, Zn, and Cd in successive sorption–desorption cycles [49]. In packed-bed bioreactors designed for the treatment of Hg-contaminated wastewater, mixed culture biofilms demonstrated greater capacity for Hg retention and higher diversity when exposed to fluctuating Hg concentrations, in contrast to monoculture biofilms [50]. Findings indicated that biofilms with greater diversity showed enhanced efficiency in the bioremediation of metals, highlighting their significance in the advancement and use of biofilm-based solutions. A moving bed sand filter, engineered for the treatment of industrial wastewater with increased levels of HMs, facilitated the development of biofilms composed of metal-resistant bacteria [51]. It has been reported that biofilms not only function as biosorption materials but also decrease the pH sufficiently to promote bioprecipitation of certain HMs [51]. A separate investigation used a mixed-species biofilm composed of sulfate-reducing bacteria, lessening the oxidation-decrease potentials to facilitate the precipitation of metal sulfides, including Cu, Zn, Ni and Fe, as well as the simultaneous precipitation of As. Treatment achieved the removal of 98% of Zn, Cu, and Ni, as well as 82 and 78% of Fe, respectively [52]. The total absorptive capacity of the biofilms was inhibited by the resistance of certain bacteria within the community such as Stenotrophomonas spp., which demonstrated resistance to Cr(VI) [53].

Soil pollution due to HMs is a global threat to human health and food production safety. Except for uncommon geogenic origins, HM pollutions are inadvertently introduced to soils through anthropogenic activities such as mining, smelting, warfare and military training, electronic industries, fossil fuels, waste disposal, agrochemical uses and irrigation [46]. The presence of HMs in polluted soils disrupts the natural ecosystem services and poses risks to human health through the food chain. Thus, accumulation of HMs in the soil causes ecosystem malfunction, water degradation and food contamination [46]. Numerous in-situ and ex-situ remediation methods have been established to manage, remediate and rehabilitate soils contaminated with HMs. These techniques include surface capping, soil flushing, electrokinetic extraction, solidification, vitrification, and phytoremediation [46]. These methods can be categorized into five distinct types of physical, chemical, electrical, thermal and biological remediation. Alternatively, they may be divided into three major groups of containment-based approaches (e.g. capping and encapsulation), transfo-rmation-based strategies (including stabilization and immobilization) and transport-based techniques (such as extraction and removal) [54]. These techniques vary significantly in effectiveness and costs in field practices. The HMs in soil are present in various forms, including dissolved ions such as Cu2+, Cd2+, CrO42−, Cr2O72− and MoO42−, as well as organic complexes where metals such as Cu2+, Pb2+ and Hg2+ are bound to dissolve organic matter within the soil solution. Additionally, there are exchangeable ions, including Cu2+, Zn2+, Cd2+, Ni2+ and Pb2+, which are adsorbed onto solid soil particles. Furthermore, HM can be detected as (co)precipitates within the soil matrix, represented by compounds such as Cd3(PO4)2, ZnS, PbCO3 and HgSO4 [55].

In-situ remediation eliminates the need for excavating and transporting contaminated soil to off-site treatment facilities. Hence, soil disturbance is minimized, the risk of exposure to contaminants for workers and the surrounding community decreases and the overall treatment costs may considerably decrease. While, the ex-situ remediation techniques entail the excavation of contaminated soil from its original site, followed by treatment at the same location or a various facility [46]. Technology for in-situ extraction and migration of potentially toxic elements in soil encompasses the removal of pollutants from the soil using physical, chemical, biological, or a combination of these methods, aimed at diminishing the risk of soil contamin-ation. This approach differs from in-situ immobilization remediation as it actively decreases the concentration of harmful pollutants in the soil, necessitating a subsequent appropriate treatment for the extracted materials. Recent uses of in-situ extraction for As-contaminated soils have included techniques such as phytoextraction, microbial extraction, and electrokinetic migration [56]. Common ex-situ soil remediation technologies include ex-situ land farming, ex-situ bio piles, ex-situ windrow, soil washing, ex-situ composting, ex-situ ion exchange, ex-situ solid-ification/stabilization, pyrolysis, thermal desorption, ultrasonic technology, and microwave technology. Due to the complexity of contaminated soils and pollutants, more than one remediation technology is usually needed to lessen the total concentration or available content of the target pollutant to an acceptable level [57]. Another technique for bioremediation includes encapsulation. The encapsulation process can be divided into three significant stages. The initial stage involves the creation of an encapsulating solution that contains at least one active ingredient, which may be an encapsulating material or a carrier. In the subsequent stage, microorganisms or materials intended for encapsulation are brought into contact with the encapsulating solution. The final stage, which may occur concurrently with the second, involves the formation of capsules through the use of the encapsulation method [58]. Applicability of these individual techniques in a specific soil remediation project is set primarily by the contamination site geography, contamination characteristics, remediation goal, cost-effectiveness, financial budget, implementation readiness, time needs, and public acceptability.

2.5. Bioremediation of heavy metals in the food industry

Rapid industrialization significantly affects water, food, feeds, and weather quality. Industries such as chemicals, food processing, and metallurgy industries release large quantities of waste, including toxic substances, into the environment. Additionally, agricultural pesticides, chem-ical fertilizers, and emissions from transportation contri-bute to harmful pollutions, which threaten food safety [59]. The HM pollution includes significant threats to food safety. Research has shown HM accumulates in various environmental sources such as water, rice, vegetables, and fish. Accumulation of these metals in human organs and tissues can lead to several health problems, including kidney damage, cardiovascular problems, and neurological disorders [59, 60]. Various methods have been suggested to solve the problem of HM pollution in the environment. Bioremediation has emerged as a promising solution for addressing HM pollution by using living organisms to decrease environmental metal concentrations. This method has received significant attention due to its effectiveness. Organisms such as plants, fungi, and microorganisms of yeasts, bacteria, algae, and cyanobacteria are frequently used in HM bioremediation. Microorganisms are particularly favored because they are easy to use and adapt well to various environments [61].

2.5.1. Roles of microorganisms in bioremediation

One of the major benefits of using microorganisms to remove toxic elements includes their safety in human aspects. An example of such microorganisms is S. cerevisiae, commonly known as bakery yeast, which is extensively used in the food industry. Advantages that have led to its popularity in the food industry include its safety for human use, large-scale production as a byproduct of the fermentation industry, and low costs of the growth media [29].

2.5.2. Use of catalase enzyme in bioremediation

Catalase is an enzyme addressed for its function in decomposing hydrogen peroxide into water and oxygen. This enzyme primarily controls the metabolism of hydrogen peroxide. Catalase is a widely occurring enzyme detected in almost all living organisms. It boasts one of the highest turnover rates within the enzymes, capable of breaking down over a million molecules of hydrogen peroxide per molecule of the enzyme. Catalase plays a critical role in several biotechnological uses, including bioremediation. Catalase has been identified as one of the most abundant and readily accessible enzymes of microbial origin, making it a valuable indicator in bioremediation processes, particularly for the remediation of crude oil-contaminated soil, as its activity is highly affected by hydrocarbon pollution. Additionally, catalase facilitates the breakdown of hydrogen peroxide into water and oxygen, providing essential oxygen during the aerobic bioremed-iation of wastes; thus, serving as a vital source of oxygen for aerobic microorganisms. Its use extends to the removal of hydrogen peroxide from bleaching industry effluents, enabling the reuse of water in subsequent dyeing proc-esses. This demonstrates its significant role in the bioremediation of wastewater from textile industries. In the food industry, catalase is used in cheese production, removal of glucose from egg whites for bakery uses, as an antioxidant enzyme system with glucose oxidase, in food packaging, and for assessing milk quality [62].

2.5.3. Remediation processes using geotextiles

In recent years, crude oil and its byproducts have significantly affected environmental degradation with contaminations from activities such as exploration, transportation, and accidents. Soil contamination from petroleum products harms soil, groundwater, and air quality. Various soil cleanup methods, including washing, oxidation, microwave heating, and bioremediation, have been used. Absorbent geotextiles have been interested in water purification due to their high oil absorption, low chemical content, energy efficiency, and ease of use. Natural fibers such as kenaf, plant fibers, milkweed floss, wool, and cotton have been used to separate oil from seawater [63].

2.6. Mechanisms of bioremediation

Numerous bacterial strains have developed a variety of distinct mechanisms to adapt, interact, acclimate and thrive in mineral-rich environments, particularly those containing HMs. These mechanisms include the uptake of HMs onto the cell surface through biosorption, intracellular seques-tration via accumulation, extracellular sequestration as insoluble compounds through precipitation and production of metabolites that solubilize and chelate metal comp-ounds, leading to leaching (Figure 1) [64].

2.6.1. Bioadsorption/biosorption

Biosorption or bioadsorption refers to non-specific active or passive physicochemical interactions between inorganic and organic metals, minerals and cellular materials [65]. The biosorption mechanism includes key processes such as surface adsorption, physisorption, chemisorption, ion exchange and surface complexation. In surface adsorption, there is an electrical attraction between negatively charged ligands on the cell wall and positively charged metal ions in the media, often resulting in an exchange reaction. Physical adsorption relies majorly on Van der Waals forces, while chemical adsorption involves attraction between the adsorbents and adsorbates. Working together or independently, these mechanisms lead to metal adsorption on the microbial cell surface. The HM binding occurs in two stages of interactions between reactive groups on the bacterial surface and metal ions, followed by metal deposition [66]. Studies on spectroscopic and chemical modifications have revealed that cellular radicals, including hydroxyl, carboxyl, sulfate, sulfhydryl (thiol), thioether, phosphate, phosphonate, phosphodiester, amino, imine, amide, imidazole and carbonyl (ketone), show a significant ability to bind metals. Most functional groups that bind metals are detected on the bacterial cell wall. Ionization of these groups provides a negative charge to the bacterial surface, enabling the attachment of cationic metals [67].

Living or unloving biological materials, including microorganisms, are used to remove or immobilize HMs and toxins from contaminated environments, including soil and water, in a process known as biosorption. Detoxifying environments that have been polluted by industrial activities, agricultural runoff and other sources of contamination can be the benefits of biosorption [68]. Bacteria, fungi and algae can bind HMs and toxins through various mechanisms, including ion exchange, physical adsorption and complexation. Functional groups of microbial surfaces such as carboxyl, hydroxyl and amino groups can interact with metal ions [69]. Detoxifying soils contaminated with HMs by biosorption can improve safety of crops grown in these areas. Elimination of HMs accumulated in food products can be used by biosorption as well [69].

2.6.1.1. Physical adsorption/physisorption

Adsorption occurs due to non-specific, quick, and reversible attraction forces such as Van der Waals forces. Alternatively, it can occur through electrostatic adsorption, resulting from Coulombic attraction between charged solute particles and bacterial cell surface [70].

2.6.1.2. Ion exchange

In the ion exchange mechanism, metal cations attach to vacant sites that were formerly occupied by other cations. Divalent metal ions are adsorbed through an exchange with polysaccharide counter ions detected on the bacterial cell wall and outer membrane. This process is affected by several factors such as the types and number of sites on the cell surface and their ionization patterns, which are ultimately governed by the pH and pKa values of the respective groups. Protonated amine groups are positively charged and become neutral upon deprotonation. Addition of protons neutralizes phosphate, carboxyl and sulfate groups. In their deprotonated state, these become negatively charged [71].

2.7. Complexation

Metal remediation can occur through the formation of complexes at the bacterial cell periphery, where reactive radicals on the cell wall interact with metal ions in solutions. Amino, carboxyl, hydroxy, thiol, phosphate and hydroxyl-carboxyl groups coordinate with HM ions to form complexes. These complexes, often referred to as coordination compounds or chelates, involve metal cations bonded to ligands. Complexation processes have been addressed for various metals such as Mg, Ca, Cd, Cu, Zn and Hg in certain bacterial species. Environmental factors such as temperature, pH, and composition of wastewater with bacterial strain types affect biosorption. The use of dead bacterial cells is increasingly favored as they are less susceptible to metal toxicity and do not need nutrients, decreasing operational costs. Biomass from fermentation industries can be used for biosorption and metal loading on non-living biomasses occurs rapidly. Bioadsorption, a fast simple process, can use live or dead bacterial cells or exopolymers to remove HMs from aqueous solutions. The process is efficient, metals are adsorbed quickly and biomasses can be reused [72, 73].

2.7.1. Bioaccumulation

Bioaccumulation refers to the accumulation of metals within bacterial cells through the uptake of non-metabolic metals using the same carrier pathways as essential metals. This process involves the binding of metal ions to reactive radicals on the bacterial cell wall and their internal regions via energy-independent mechanisms. Then, metals diffuse into the cytoplasm through energy-dependent or independent processes. Transport pathways for essential ions such as sodium, potassium, and magnesium are used for the transport of HMs across microbial membranes for intracellular accumulation. Cation transport systems bind with HM ions that share similar ionic radius and charge with essential metal ions [73].

Microorganisms can uptake toxic metals within their cellular structures in the bioaccumulation process. Through bioaccumulation, they can detoxify environments by sequestering harmful substances. Bacterial mechanisms of action include conversion of HMs to less harmful states, absorption and bioaccumulation of HMs, and extracellular precipitation by the production of exopolysaccharides or other compounds that bind HMs [74]. Rhizosphere microbes can assist in breaking down organic pollutants or converting HMs into forms that are easier for plants to uptake or detoxify by phytoremediation synergy [75]. Microbes help ensure that crops growing, having lower levels of harmful substances by cleaning up contaminated soils using bioaccumulation division of bioremediation. Figure 2 classifies roles of microorganisms in detoxification.

2.7.1.1. Intracellular transport

Normally, HMs enter bacterial cells through channels, secondary carrier proteins and primary active transporters on the cell membrane. Channels are α-helical proteins that facilitate passive diffusion of metals based on the concentration gradients. These proteins, parts of the major intrinsic proteins superfamily, help transport As and Hg in various bacterial species such as E. coli, Corynebacterium, Streptomyces coelicolor, Serratia and Pseudomonas [76, 77].

2.8. Intracellular fate of bioaccumulated heavy metals

Growing bacterial cells can continuously eliminate metals through internal detoxification processes. These processes include biotransformation and reduction of metals via enzymes, methylation, sequestration through metal-organic complex formation and production of metal chelators such as metallothioneins. These mechanisms help bacteria defend against metal toxicity [78].

2.8.1. Bioprecipitation

Bacteria remove metals from solution through precipitation, a defense mechanism independent of cellular metabolism. This occurs due to chemical interactions between the bacterial surface and metal ions with precipitation happening via reduction, sulfide formation or phosphate formation, depending on the species and environment.

2.8.1.1. Metal precipitation as phosphates

In this precipitation method, the enzyme phosphatase releases inorganic phosphate (Pi) from cellular organic phosphates such as glycerol-2-phosphate. Then, Pi precipitates metals or radionuclides as phosphates on the bacterial cell surface. For example, immobilized cells of Citrobacter spp. precipitated Cu, Cd, Pb and U from glycerol-2-phosphate enriched solutions. The phosphatase enzyme catalyzed the cleavage of glycerol-2-phosphate, releasing hydrogen phosphates that precipitate metals extracellularly as insoluble metal phosphates [65].

2.8.1.2. Reduction

Many bacterial strains can decrease hazardous selenite and selenate to elemental selenium, which precipitates as a red deposit. Desulfomicrobium norvegicum biofilm precipitates selenium with sulfur [79], while Alteromonas (now Shewanella) putrefaciens reduces U(VI) to U(IV) carbonate [80]. Shewanella oneidensis MR-1 and Geobacter species can reduce Hg(II) to Hg(0) [81].

2.8.1.3. Metal precipitation as sulfides

Sulfate-reducing bacteria are anaerobic organisms that use sulfate as a terminal electron acceptor, reducing it to sulfide. This sulfide combines with metals in the cell and the environment to form insoluble metal sulfides, protecting the bacteria from metal toxicity. These bacteria create reducing conditions that can reduce metals such as U(VI) and Cr (VI) [82]. Enzymatic sulfide formation has been observed in Salmonella typhimurium and Klebsiella planticola, where metal-sulfide precipitation occurs due to sulfide production from thiosulfate [83].

Rapid industrialization and events such as uncontrolled agrochemicals lead to high soil HM concentrations. Non-bridgeable HM persistent in nature causes harmful effects on human health and disrupts the environment and agricultural food products. Natural organisms such as microbes are useful tools for detoxifying HMs and toxins in food through bioremediation involving various biological methods [48] as follows.

2.9. Biotransformation [(oxidation-reduction (redox reaction)]

Microorganisms can transform THMs and organic pollutants into less toxic forms through oxidation-reduction reactions. This process facilitates the conversion of harmful substances into further benign compounds, enhancing detoxification [84]. However, factors such as temperature, pH, nutrient availability, and the presence of co-contaminants can affect microbial activity and efficacy. Moreover, certain HMs can be toxic to the microbes, potentially limiting their effectiveness [85]. Biotransformation through redox reactions is a vital microbiological process. Detoxification of HMs and toxins makes biotransformation an essential tool in bioremediation to ensure food safety and environmental health.

2.10. Biomineralization

Microorganisms can facilitate precipitation of HMs as mineral forms, effectively immobilizing them and reducing their bioavailability and toxicity in contaminated environments, including soils and aquatic systems, known as biomineralization [86]. Biomineralization can help detoxify contaminants in agricultural soils; thereby, decreasing the presence of harmful metals in crops in the context of food production. This process is critical for maintaining food safety and protecting human health. Identifying and engineering specific microbial strains that show high efficiency in HM detoxification as well as understanding mechanisms underlying biomineralization are essential research interests [86].

2.11. Bioprecipitation 

The primary mechanism of bioprecipitation involves the transformation of soluble metal ions into insoluble precipitates through microbial metabolic activities by metabolic byproducts (e.g. sulfides or phosphates) that react with HMs [87], leading to their removal from solutions. This process can clean contaminated water and soil systems. Bioprecipitation can help detoxify metals through the intervention of specific microbial strains for food products contaminated with HMs such as Cd, Pb, and Hg. Soil and water treatments are used to decrease metal contamination before crops are grown or harvested by bioprecipitation use. This enhances the safety of the food produced [88].

2.12. Bioleaching

Extraction of HMs from minerals or contaminated materials by microbes is known as bioleaching, which facilitates the release of metals into solution through microbial activity, making it easier to remove HMs from contaminated sites [85]. Bioleaching methods can potentially be used to detoxify HMs and other toxins in food products. Harmful substances degrading or transforming them into less toxic forms by microbes can enhance food safety. Bioleaching as a broader strategy for bioremediation may involve phytoremediation (using plants) or other microbiological techniques to improve detoxification efficacy.

2.12.1. Sulfide bioleaching

Sulfide-producing microorganisms such as Acidithiobacillus and Leptospirillum can solubilize HMs from complex minerals using sulfide bioleaching [89]. It is particularly effective for extracting metals such as Cu and Zn while minimizing environmental effects. The HMs and toxins can enter the food chain through contaminated soil, water, and food processing practices. The use of sulfide bioleaching and associated microbial techniques can help lessen these risks [89].

2.12.2. Pyrite leaching

Certain microorganisms can oxidize pyrite (FeS2) to solubilize metals during the mineral recovery processes [90]. This method is often used in mining to recover valuable metals while managing metal contamination of soil appropriate for food safety. Oxidation of pyrite in the presence of water and oxygen leads to sulfuric acid production and can release iron and other associated HMs (e.g. Pb, As, and Cd) that may be bound to the pyrite matrix. Bacteria and archaea (e.g., Acidithiobacillus spp.) can oxidize pyrite to facilitate the leaching process. Acceleration of the pyrite breaking down using the released iron and sulfur for their metabolic processes, creating an environment that enhances metal solubility and bioavailability [91]. In the context of food safety and agriculture, pyrite leaching with the assistance of microbes detoxifies soils contaminated with HMs to improve the safety and quality of agricultural products.

2.13. Heterotrophic bacterial leaching

Heterotrophic bacteria can leach metals from contaminated substrates using organic matter, potentially helping in the detoxification of metal-rich wastes or soils and other environmental toxins such as those present in foods [92]. Heterotrophic bacteria rely on other organic compounds for nutrition. They metabolize organic matter and can degrade various pollutants such as HMs, hydrocarbons, pesticides, and other harmful substances. Mechanisms of action include bioaccumulation; in which, bacteria can absorb HMs through their cell walls. These metals accumulate within the cells and can later be harvested or removed. Biotransformation includes the conversion of toxic forms of HMs into less harmful states; they may reduce soluble metal ions to insoluble forms, rendering them less bioavailable [85]. Extracellular precipitation is a way; by which, bacteria can produce exopolysaccharides that bind to HMs, causing them to precipitate and settle out of solutions [93]. In food production and processing, heterotrophic bacterial leaching can be used to remediate crops contaminated by HMs and toxins, decreasing the risks associated with consuming contaminated foods. This process may involve agricultural soils or water used in irrigation; thereby, ensuring a safer food supply. Identifying specific strains of heterotrophic bacteria that show high efficacy in removing HMs and other toxins. Assistance in genetic engineering and synthetic biology can enhance the capabilities of these microorganisms [94].

2.14. Biosurfactant production

Biosynthesis of surfactant-like substances by microorganisms such as bacteria and fungi refers to biosurfactant production. Biosurfactants can decrease surface tension and facilitate solubilization, emulsification, and dispersion of hydrophobic compounds, including HMs and organic pollutants due to amphiphilic characteristics [95]. Biosurfactants can play several important roles associated with the detoxification of HMs and toxic substances in foods. Solubility of HMs such as Pb, Cd, Hg, and organic toxins (e.g. pesticides and harmful industrial byproducts) by biosurfactants in water as solubilization makes them further accessible to microorganisms for uptake and transformation. Other characteristics of biosurfactants are microbial growth promotion, bioavailability enhancement of pollutants, stabilization of pollutant-microbe interface, and environmental remediation. Certain bacterial species of Pseudomonas, Rhodococcus, Bacillus, and Serratia are significant producers of biosurfactants. Yeasts such as Candida spp. and filamentous fungi such as Aspergillus spp. can also produce these compounds [96]. Biosurfactants can potentially be used in the food industry by detoxification and food preservation.

2.15. Biomethylation 

Microorganisms can convert inorganic metals into less toxic volatile forms through biomethylation, which facilitates the removal of HMs from ecosystems. Bacteria and fungi include enzymes that can facilitate the transfer of a methyl group (-CH₃) to HMs (e.g. Hg, As, and Pb) or other organic compounds [97]. Biomethylation is useful in foods to ensure that food products are free from HM decontamination through detoxification and other contaminants. The composition of microbial communities and specific strains or consortia of microbes are selected to promote desired transformations of HMs and enhance bioremediation outcomes.

2.16. Biovolatilization

Transformation of metals into gaseous forms by microbes known as biovolatization allows for the detoxification and removal of metals from the environment and foods. The HMs such as Hg and As can be converted into volatile organometallic compounds such as methylmercury, which can then be volatilized into the atmosphere by bacteria and fungi [98]. Biovolatization may be significant when bioavailability of HMs can decrease; thereby, minimizing their uptake by plants and hence their presence in the food supply.

2.17. Microbe-assisted phytoremediation

This approach uses plants and microorganisms (microbes) in combination to clean up contaminated environments, particularly from HMs and toxins. The natural ability of plants to absorb and detoxify pollutants is a sustainable effective bioremediation strategy. Plants uptake HMs and other toxins through their roots and then store them in their tissues or transform them into less harmful substances. This process is known as phytoremediation [99]. Microbes in rhizospheres can transform HMs into further soluble forms, making them further available for uptake by plants. Detoxification by microbes degrades or transforms harmful organic pollutants such as pesticides and industrial chemicals, decreasing their toxicity and symbiotic relationships; in which, plants form mutualistic relationships with beneficial microbes (e.g. mycorrhizal fungi and nitrogen-fixing bacteria) to increase the plant's tolerance and accumulation of HMs [100]. Phytoremediation assisted by bacteria can include agricultural implications and decrease food contamination from HMs and other pollutants.

2.18. Bioremediation technologies

Bioremediation technologies can broadly be classified as ex-situ or in-situ technologies (Figure 3).

 2.18.1. Ex-situ treatments

Ex-situ bioremediation involves relocating pollutants from contaminated sites for treatment. Factors affecting method choice include treatment costs, pollution type and depth, contamination extent, site location and geology. Techniques include piling, bioreactors, landfarming and windrows. Advantages include effective biodegradation, detoxification of pollutants and decrease of treatment time. Limitations include difficulties with chlorinated hydrocarbons and specific soil treatment needs [6, 26, 39].

Biopiles are engineered, aerated composted piles used for treating surface contamination from petroleum hydrocarbons. They represent an advanced version of land farming, helping manage contaminant losses through leaching and volatilization. Biopiles substitute an environment for indigenous aerobic and anaerobic microorganisms and introducing effective microbes can enhance pollutant remediation efficiency [101].

A bioreactor accelerates bioremediation of the soil and water pollutants by creating a controlled three-phase system (solid, liquid and gas), enhancing biodegradation, compared to in-situ methods. Contaminated soil often needs pretreatment such as washing before mixing with water and additives in the bioreactor. The process involves agitation to promote contacts with microorganisms and optimizes conditions for pollutant degradation. After treatment, water is separated from solids, which may need additional disposal or treatment if pollutants persist [3].

Landfarming is an efficient method for treating contaminated soil by spreading and cultivating it to enhance microbial activity, particularly for the upper 10–35 cm of the surface soil. It maintains a neutral pH with agricultural lime and is effective for ex-situ and in-situ bioremediations of contaminants such as aliphatic hydrocarbons, polycyclic aromatic hydrocarbons and PCBs. This cost-effective approach needs minimal equipment and maintenance [102].

Windrow bioremediation restores contaminated soil by regularly turning materials to enhance hydrocarbon-degrading bacterial activity. This method improves aeration, moisture and nutrient distribution, leading to faster bioremediation through assimilation and mineralization. Studies show windrow methods generally outperform pile systems in removing hydrocarbons but may be less effective in soils with toxic volatiles and can produce methane emissions from anaerobic zones if aeration is inadequate [6, 102].

 2.18.2. In-situ treatment

Natural attenuation and bioremediation are methods to treat polluted environments; in which, microorganisms contribute to pollutant degradation. Natural attenuation is a cost-saving method to treat polluted environments [103]. Natural attenuation is based on natural processes to clean up and attenuate pollutions. It encompasses processes that lead to decreases of the mass, toxicity, mobility or volume of the contaminants without human intervention, where naturally occurring environmental processes break down and decrease the concentration of contaminants in the envi-ronment via microorganisms [104]. Natural attenuation is the most interesting process because the contaminants can be transformed into less harmful products. This method contains the addition of electron acceptors, electron donors, or nutrients to stimulate naturally occurring microbial populations (biostimulation or introduction of nutrients and chemicals to stimulate indigenous microorganisms) or can introduce specific microorganisms that aimed at enhancing biodegradation of the target compounds (bioaugmentation or inoculation with exogenous microorganisms) [105].

Biostimulation or stimulated aerobic biodegradation is the process of eliminating factors that limit the biodegradation of contaminants [106]. It involves modification of the environment to stimulate existing bacteria capable of bioremediation. This was first defined by Perfumo as the addition of nutrients, oxygen or other electron donors and acceptors to the coordinated site to increase population or activity of naturally occurring microorganisms for bioremediation. The key benefit of biostimulation includes that bioremediation is carried out by the natural micro-organisms, which are well adapted to the subsurface environment and spatially spread in the subsurface [107].

Bioaugmentation is a method used for the bioremoval of HM contaminated sites using specific or genetically engineered bacteria that are proficient in fighting against HM contaminants [108]. Addition of microbial biomasses to contaminated areas can significantly improve their biodegradation performance. In addition to save time and costs, this method can be adapted to the green environment [109].

Bioventing is an in-situ technology that includes stimulating indigenous microorganisms through the addition of gas to destroy contaminants; it uses low airflow rates to provide adequate oxygen to maintain the microbial activity [110]. Therefore, it decreases release of volatile contaminants into the air. Benefits of bioventing vary. First, it offers an environmental friendly solution, minimizing needs of excavation and disposal of the contaminated soil. Second, it is often more cost-effective than that the traditional methods are, needing less labors and specialized equipment [111].

Bioslurping is a novel technology that combines two remediation methods of bioventing and vacuum-enhanced free-product recovery. Bioslurping, known as dual-phase extraction, is an in-situ technique that uses vacuum-enhanced dewatering technologies to remediate contaminated sites [112]. Bioslurping use elements of, bioventing and free-product recovery to address two separate contaminant media. Bioslurping can enhance free-product recovery and is a cost-efficient technology. Depending on the site conditions, operation and maintenance time of bioslurping can vary from a few months to years [113].

Biosparging is an in-situ remediation technology that injects air or gases into the contaminated area to stimulate aerobic biological activities for the encouragement of aerobic biodegradation. This recovery method is used to eliminate residual contamination in the site and target chemical compounds that can be degraded under aerobic conditions [114]. Two primary factors may limit the effectiveness of biosparing, including (1) permeability of the contaminated site and (2) biodegradability of the contaminants [115].

Biofiltration is a pollution control system using bioreactors containing living materials to capture and biologically destroy contaminants. This includes pollutant removal via physical separation, adsorption on the filter media and biodegradation and biotransformation by microbes, forming biofilms on the filter media [116]. Biofiltration-based studies have shown significant results in the removal of volatile organic compounds and HMs [117]. A significant advantage of biofiltration techniques is that the contaminants are converted into biodegradable wastes without deriving secondary pollutants within a specific time [118].

Chemical and physical waste cleanup methods are used for the contamination removals; however, these approaches are costly and include harmful environmental effects. Therefore, increases in use of bioremediation have been recorded, leading to development of genetically engineered microbes. Genetic engineering refers to the direct manipulation of DNA to alter characteristics of an organism in a particular way. These designed engineered microorganisms appeared more effective than normal strains and include superior debasement adaptability as well as capacity to rapidly adapt to various contaminants. Use of engineered microorganisms is safer and more cost-effective, compared to other methods [119]. Bioremediation in combination with nanotechnology has been investigated as further effective for the removal of environmental contamination. This combined method can include a broader range of potential uses with decreased costs and lowest negative effects on the environment. Degradation of contaminants using nanoparticle catalysts is a common use of nanotechnology in this field [120].

2.19. Factors limiting bioremediation technologies

Bioremediation includes several benefits, compared to conventional methods such as landfilling or incineration [121]. However, there are limitations to bioremediation; chemicals such as HMs, radionuclides and chlorinated compounds are not susceptible to biodegradation. Moreover, microbial systems may produce toxic metabolites from the contaminants (Table 2) [122].

3. Conclusion

Various methods are reported to remove contamination, one of which is bioremediation. Use of microorganisms to diminish or decrease concentration of dangerous wastes is called bioremediation. This word refers to a group of methods that use biological systems such as indigenous or genetically modified microbes to restore or clean-up contaminated environments. A majority of indigenous bacteria are capable of successfully restoring the environment by oxidizing, immobilizing and/or converting pollutants into carbon dioxide and water. Such a biological action system includes various uses such as cleanup of the contaminated material (e.g. foods and water). Bioremediation is an easy, cost and timesaving method to treat polluted environments, compared to the conventional methods such as landfilling or incineration. This method is a commonly accepted substitute for cleaning up the environment as it particularly promises to reach clean-up objectives at an economical rate with milder chances of the contaminant spread to other media.

Further studies: Enhancing regulatory frameworks and increasing public awareness about HM exposure is critical for the prevention. Investigation of the innovative technologies for monitoring and removing HMs from wastewater can significantly decrease associated risks. Collaborative efforts within the governments, industries and communities are vital for creating cleaner environments and protecting future generations from contaminations. Focusing on advanced technologies and public education is a key to minimizing health risks.

4.Acknowledgements

This study was spported by Shahid Beheshti University of Medical Sciences (grant no. 43012694).

5.Conflict of Interest

The authors report no conflict of interest.

6.Author contributions

All authors participated in project administration and writing of the primary draft of the manuscript, providing critical revision and editing. All authors approved the final version of the manuscript.

7. Ethical Code:

This project is approved via

 IR.SBMU.RETECH.REC.1403.517 ethical code.

References

  1. Zaynab M, Al-Yahyai R, Ameen A, Sharif Y, Ali L, Fatima M, Khan KA, Li S. Health and environ-mental effects of heavy metals. J King Saud Univ Sci. 2022. 34(1): 101653.

https://doi.org/10.1016/j.jksus.2021.101653

  1. Budi HS, Catalan Opulencia MJ, Afra A, Abdelbasset WK, Abdullaev D, Majdi A, Taherian M, Ekrami HA, Mohammadi MJ. Source, toxicity and carcinogenic health risk assessment of heavy metals. Rev Environ Health. 2024. 39(1): 77-90.

https://doi.org/10.1515/reveh-2022-0096

  1. Rahman Z, Singh VP. Bioremediation of toxic heavy metals (THMs) contaminated sites: concepts, applica-tions and challenges. Environ Sci Pollut Res Int. 2020. 27(22): 27563-27581.

https://doi.org/10.1007/s11356-020-08903-0

  1. Mushtaq M, Arshad N, Rehman A, Javed GA, Munir A, Hameed M, Javed S. Levilactobacillus brevis MZ3840-11 and Levilactobacillus brevis MW362779 can miti-gate lead induced hepato-renal damage by regulating visceral dispersion and fecal excretion. World J Microbiol Biotechnol. 2024. 40(2): 74.

https://doi.org/10.1007/s11274-023-03818-7

  1. Kondakindi VR, Pabbati R, Erukulla P, Maddela NR, Prasad R. Bioremediation of heavy metals-contaminated sites by microbial extracellular polymeric substances- A critical view. Environ Chem Ecotoxicol. 2024, 6:408-421.

https://doi.org/10.1016/j.enceco.2024.05.002

  1. Azubuike CC, Chikere CB, Okpokwasili GC. Biorem-ediation techniques–classification based on site of appli-cation: principles, advantages, limitations and prospects. World J Microbiol Biotechnol. 2016. 32:1-18.

https://doi.org/10.1016/B978-0-443-21911-5.00003-9

  1. Panigrahi S, Velraj P, Rao T.S. Functional microbial diversity in contaminated environment and application in bioremediation, in Microbial diversity in the genomic era. 2019. Elsevier. 359-385.

https://doi.org/10.1016/B978-0-12-814849-5.00021-6

  1. Mirmahdi R.S, Zoghi A, Mohammadi F, Khosravi-Darani K, Jazaiery S, Mohammadi R, Rehman Y. Biode-contamination of milk and dairy products by probiotics: Boon for bane. Ital. J. Food Sci. 2021. 33(SP1): 78-91.

https://doi.org/10.15586/ijfs.v33iSP2.2053

  1. Sarlak Z, Khosravi-Darani K, Rouhi M, Garavand F, Mohammadi R. Sobhiyeh MR. Bioremediation of org-anophosphorus pesticides in contaminated foodstuffs using probiotics. Food Control. 2021. 126:108006.

https://doi.org/10.2174/1389557519666190318102201

  1. Massoud R, Khosravi‐Darani K, Sharifan A, Asadi G, Zoghi A. Lead and cadmium biosorption from milk by Lactobacillus acidophilus ATCC 4356. Food Sci. Nutr. 2020. 8(10): 5284-91.

https://doi.org/10.1016/j.foodcont.2021.108006

  1. Attarianshandiz M. Enhancing immunity against carcin-ogens through probiotics: A literature review. J Food Sci Nutr The. 2023. 9(1): 013-23.

https://doi.org/10.17352/jfsnt.000043

  1. Fernández PM, Viñarta SC, Bernal AR, Cruz EL, Figueroa LI. Bioremediation strategies for chromium removal: current research, scale-up approach and future perspectives. Chemosphere. 2018. 208:139-148.

https://doi.org/10.1016/j.chemosphere.2018.05.166

  1. Shakti P, Pandey AK. Agricultural soil contamination due to industrial discharges: challenges for public health protection and food security. In Bioremediation of Emerging Contaminants from Soils, 2024. Elsevier. 21-42.

https://doi.org/10.1016/B978-0-443-13993-2.00002-5

  1. Dan S, Sharma A. Microbial Bioremediation of Heavy Metals: Emerging Trends and Recent Advances. Res J Biotechnol. 2020. 15: 164-178.
  2. Fowler B.A, Alexander J, Oskarsson A. Toxic metals in food, in Handbook on the Toxicology of Metals. 2015. Elsevier. 123-140.

https://doi.org/10.1016/B978-0-444-59453-2.00006-8

  1. Iyer M, Anand U, Thiruvenkataswamy S, Babu HW, Narayanasamy A, Prajapati VK, Tiwari CK, Gopalak-rishnan AV, Bontempi E, Sonne C, Barceló D. A review of chromium (Cr) epigenetic toxicity and health hazards. Sci Total Environ. 2023. 882: 163483.

https://doi.org/10.1016/j.scitotenv.2023.163483

  1. DesMarias T.L, Costa M. Mechanisms of chromium-induced toxicity. Curr Opin Toxicol. 2019. 14:1-7.

https://doi.org/10.1016/j.cotox.2019.05.003

  1. Kumar A, Kumar A, MMS CP, Chaturvedi AK, Shab-nam AA, Subrahmanyam G, Mondal R, Gupta DK, Malyan SK, Kumar SS, Khan S. Lead toxicity: health hazards, influence on food chain and sustainable remed-iation approaches. Int J Environ Res Public Health, 2020. 17(7): 2179.

https://doi.org/10.3390/ijerph17072179

  1. Senthil Kumar P, Gunasundari E. Bioremediation of Heavy Metals. In: Varjani S, Agarwal A, Gnansounou E, Gurunathan B. Bioremediation: Applications for Environmental Protection and Management. Energy, Environment and Sustainability. Singapore, 2018: 165-195.

https://doi.org/10.1007/978-981-10-7485-1_9

  1. Dixit R, Wasiullah X, Malaviya D, Pandiyan K. Singh UB, Sahu A, Shukla R, Singh BP, Rai JP, Sharma PK, Lade H. Bioremediation of heavy metals from soil and aquatic environment: an overview of principles and criteria of fundamental processes. Sustainability. 2015. 7(2): 2189-2212.

https://doi.org/10.3390/su7022189

  1. Bjørklund G, Rahaman MS, Shanaida M, Lysiuk R, Oliynyk P, Lenchyk L, Chirumbolo S, Chasapis CT. Peana M, Natural dietary compounds in the treatment of arsenic toxicity. Molecules. 2022. 27(15): 4871.

https://doi.org/10.3390/molecules27154871

  1. Jobby R, Desai N. Bioremediation of heavy metals. Biodegradation and bioremediation. Environ Eng Sci. 2017. 8: 201-220.

https://doi.org/10.1080/15569543.2023.2283768

  1. Mohanta TK, Mohanta YK, Mohanta N. Role of bio-technology in bioremediation, in Biotechnology: Con-cepts, Methodologies, Tools and Applications. 2019. IGI Global. 1-34.

https://doi.org/10.4018/978-1-5225-8903-7.ch001

  1. Pant G, Garlapati D, Agrawal U, Prasuna RG. Mathi-mani T, Pugazhendhi A. Biological approaches prac-tised using genetically engineered microbes for a sustai-nable environment: a review. J Hazard Mater. 2021. 405: 124631.

https://doi.org/10.1016/j.jhazmat.2020.124631

  1. Sharma S, Pathania S, Bhagta S, Kaushal N, Bhardwaj S, Bhatia RK, Walia A. Microbial remediation of polluted environment by using recombinant E. coli: a review. Biotechnol Environ. 2024. 1(1): 8.

https://doi.org/10.1186/s44314-024-00008-z

  1. Abo-Alkasem M.I, Hassan N.m.H, Abo Elsoud M.M. Microbial bioremediation as a tool for the removal of heavy metals. Bull Natl Res Cent. 2023. 47(1): 31.

https://doi.org/10.1186/s42269-023-01006-z

  1. Hakeem K, Bhat R, Qadri H. Bioremediation and Biotechnology. Cham, 2020.

https://doi.org/10.1007/978-3-030-35691-0

  1. Ansari F, Lee CC, Rashidimehr A, Eskandari S, Ashaolu TJ, Mirzakhani E, Pourjafar H, Jafari SM. The role of probiotics in improving food safety; detoxification of heavy metals and chemicals. Toxin Rev, 2024. 43(1): 63-91.

https://doi.org/10.1016/j.ejbt.2018.11.003

  1. Massoud R, Hadiani MR, Hamzehlou P, Khosravi-Darani K. Bioremediation of heavy metals in food industry: Application of Saccharomyces cerevisiae. Electron J Biotechnol. 2019. 37:56-60.

https://doi.org/10.1016/j.ejbt.2018.11.003

  1. Homayouni Rad A, Pourjafar H, Mirzakhani E. A com-prehensive review of the application of probiotics and postbiotics in oral health. Front Cell Infect Microbiol. 2023, 13:1120995.

https://doi.org/10.3389/fcimb.2023.1120995

  1. Goyal P, Belapurkar P, Kar A. A review on in vitro and in vivo bioremediation potential of environmental and probiotic species of Bacillus and other probiotic micro-organisms for two heavy metals, cadmium and nickel. Biosci. Biotechnol. Res. Asia. 2019. 16(1): 01-13.

http://dx.doi.org/10.13005/bbra/2714

  1. Massoud R, Sharifan A, Khosravi-Darani K, Asadi G. Cadmium bioremoval by Saccharomyces cerevisiae in milk. J Med Microbiol Infect Dis. 2020. 8(1): 29-33.

https://doi.org/10.29252/JoMMID.8.1.29

  1. Ameen F, Hamdan A, El-Naggar M. Assessment of the heavy metal bioremediation efficiency of the novel marine lactic acid bacterium, Lactobacillus plantarum MF042018. Sci Rep. 2020.10:314.

https://doi.org/10.1038/s41598-019-57210-3

  1. Afsharian Z, Salavatifar M, Khosravi-Darani K. Impact of simulated microgravity on bioremoval of heavy-metals by Lactobacillus acidophilus ATCC 4356 from water. Heliyon. 2022. 8(12): e12307.

https://doi.org/10.1016/j.heliyon.2022.e12307

  1. Wahid M, Awais MU, Talat R, Mehmood A, Haq I, Al Farraj DA, Mohammed YH, Banach A. Antibiotic susceptibility and bioremediation potential of probiotic bacteria against lead and cadmium isolated from yogurt. Pak J Pharm Sci. 2023. 36(3): 969-972.

https://doi.org/10.36721/PJPS.2023.36.3.SP.969-972.1

  1. Mushtaq M, Arshad N, Hameed M, Munir A, Javed GA, Rehman A. Lead biosorption efficiency of Levilacto-bacillus brevis MZ384011 and Levilacto-bacillus brevis MW362779: A response surface based approach. Saudi J Biol Sci. 2023. 30(2): 103547.

https://doi.org/10.1016/j.sjbs.2022.103547

  1. Salavatifar M, Khosravi‐Darani K. Investigation of the simulated microgravity impact on heavy metal biosorp-tion by Saccharomyces cerevisiae. Food Sci Nutr. 2024. 12(5): 3642-3652.

https://doi.org/10.1002/fsn3.4034

  1. Ali S, Bukhari D.A, Rehman A. Call for biotechnol-ogical approach to degrade plastic in the era of COVID-19 pandemic. Saudi J Biol Sci. 2023. 30(3): 103583.

https://doi.org/10.1016/j.sjbs.2023.103583

  1. Bala S, Garg D, Thirumalesh BV, Sharma M, Sridhar K,. Inbaraj BS, Tripathi M. Recent strategies for bioremedi-ation of emerging pollutants: a review for a green and sustainable environment. Toxics, 2022. 10(8): 484.

https://doi.org/10.3390/toxics10080484

  1. Sharma P, Singh SP, Parakh SK, Tong YW. Health hazards of hexavalent chromium (Cr (VI)) and its micro-bial reduction. Bioeng. 2022. 13(3): 4923-4938.

https://doi.org/10.1080/21655979.2022.2037273

  1. Mahmoud GAE. Microbial Scavenging of Heavy Metals Using Bioremediation Strategies. In: Kumar V, Prasad R, Kumar M. Rhizobiont in Bioremediation of Hazardous Waste. 2021. Singapore. 265-289.

https://doi.org/10.1007/978-981-16-0602-1_12

  1. Ambaye TG, Vaccari M, Franzetti A, Prasad S, Formi-cola F, Rosatelli A, Hassani A, Aminabhavi TM, Rtimi S. Microbial electrochemical bioremediation of petro-leum hydrocarbons (PHCs) pollution: Recent advances and outlook. Chem Eng J. 2023. 452:139372.

https://doi.org/10.1016/j.cej.2022.139372

  1. Adamczyk P.A, Reed J.L. Escherichia coli as a model organism for systems metabolic engineering. Curr Opin Syst Biol. 2017. 6:80-88.

https://doi.org/10.1016/j.coisb.2017.11.001

  1. Bălă GP, Râjnoveanu RM, Tudorache E, Motișan R, Oancea C. Air pollution exposure -the (in) visible risk factor for respiratory diseases. Environ Sci Pollut Res. 2021. 28(16): 19615-19628.

https://doi.org/10.1007/s11356-021-13208-x

  1. Mohamed MS, El-Arabi NI, El-Hussein A, El-Maaty SA, Abdelhadi AA. Reduction of chromium-VI by chro-mium-resistant Escherichia coli FACU: a prospective bacterium for bioremediation. Folia Microbiol. 2020. 65:687-696.

https://doi.org/10.1007/s12223-020-00771-y

  1. Liu L, Li W, Song W, Guo M. Remediation techniques for heavy metal-contaminated soils: Principles and applicability. Sci Total Environ. 2018. 633:206-219.

https://doi.org/10.1016/j.scitotenv.2018.03.161

  1. Mishra S, Huang Y, Li J, Wu X, Zhou Z, Lei Q, Bhatt S, Chen S. Biofilm-mediated bioremediation is a powerful tool for the removal of environmental pollutants. Chemosphere. 2022. 294:133609.

https://doi.org/10.1016/j.chemosphere.2022.133609

  1. Verma S, Kuila A. Bioremediation of heavy metals by microbial process. Environ Technol Innov. 2019. 14: 100369.

https://doi.org/10.1016/j.eti.2019.100369

  1. Ruhal R, Kataria R. Synthetic biology approaches for bioremediation of metals. In: Bioremediation of Toxic Metal -(loid)s. 2022. CRC Press. 292-306.

https://doi.org/10.1201/9781003229940-19

  1. Mahto KU, Kumari S, Das S. Unraveling the complex regulatory networks in biofilm formation in bacteria and relevance of biofilms in environmental remediation. Crit Rev Biochem Mol Biol. 2022. 57(3): 305-32.

https://doi.org/10.1080/10409238.2021.2015747

  1. Bai H, Liu D, Zheng W, Ma L, Yang S, Cao J, Lu X, Wang H, Mehta N. Microbially-induced calcium carbo-nate precipitation by a halophilic ureolytic bacterium and its potential for remediation of heavy metal-contaminated saline environments. Int Biodeter Biodegr. 2021. 165:105311.

https://doi.org/10.1016/j.ibiod.2021.105311

  1. Chen J, Zhang G, Ma C, Li D. Antimony removal from wastewater by sulfate-reducing bacteria in a bench-scale upflow anaerobic packed-bed reactor. Acta Geochimica. 2020. 39:203-215.

https://doi.org/10.1007/s11631-019-00382-6

  1. An Q, Deng SM, Zhao B, Li Z, Xu J, Song JI. Simultaneous denitrification and hexavalent chromium removal by a newly isolated Stenotrophomonas malto-philia strain W26 under aerobic conditions. Environ Chem. 2021. 18(1): 20-30.

https://doi.org/10.1071/EN20097

  1. Zhang H, Yuan X, Xiong T, Wang H, Jiang L. Bioremediation of co-contaminated soil with heavy metals and pesticides: Influence factors, mechanisms and evaluation methods. Chem Eng J. 2020. 398: 125657.

https://doi.org/10.1016/j.cej.2020.125657

  1. Kim R.Y, Yoon JK, Kim TS, Yang JE, Owens G, Kim KR. Bioavailability of heavy metals in soils: definitions and practical implementation-a critical review. Environ Geochem Health. 2015. 37:1041-1061.

https://doi.org/10.1007/s10653-015-9695-y

  1. Liao X, Li Y, Miranda-Avilés R, Zha X, Anguiano JH, Sánchez CD, Puy-Alquiza MJ, González VP, Garzon LF. In situ remediation and ex situ treatment practices of arsenic-contaminated soil: An overview on recent advances. J Hazard Mater Adv. 2022. 8:100157.

https://doi.org/10.1016/j.hazadv.2022.100157

  1. Koul B, Taak P. Biotechnological strategies for effective remediation of polluted soils. 2018. Singapore.

https://doi.org/10.1007/978-981-13-2420-8

  1. Valdivia-Rivera S, Ayora-Talavera T, Lizardi-Jiménez MA, García Cruz U. Encapsulation of microorganisms for bioremediation: techniques and carriers. Rev Environ Sci Biotechnol. 2021. 20(3): 815-838.

https://doi.org/10.1007/s11157-021-09577-x

  1. Sardar K, Ali S, Hameed S, Afzal S, Fatima S, Shakoor MB, Bharwana SA, Tauqeer HM. Heavy metals contam-ination and what are the impacts on living organisms. Greener J Environ Manage. 2013. 2(4): 172-179.

https://doi.org/10.15580/GJEMPS.2013.4.060413652

  1. Owolabi J, Hekeu M. Heavy metal resistance and antibiotic susceptibility pattern of bacteria isolated from selected polluted soils in Lagos and Ota, Nigeria. Int J Appl Sci. 2014. 4(6): 6-12.
  2. Halder S, Bioremediation of heavy metals through fresh water microalgae: a review. Sch Acad J Biosci, 2014, 2(11): 825-830.

https://doi.org/10.36347/sajb.2014.v02i11.016

  1. Kaushal J, Mehandia S, Singh G, Raina A, Arya SK. Catalase enzyme: Application in bioremediation and food industry. Biocatal Agric Biotechnol. 2018. 16:192-199.

https://doi.org/10.1016/j.bcab.2018.07.035

  1. Rezaei A, Rowshanzamir M, Hejazi SM. Banitalebi-Dehkordi M, Application of superabsorbent geotextiles to decontaminate and improve crude oil-contaminated soil. Transp Geotech. 2023. 38:100910.

https://doi.org/10.1016/j.trgeo.2022.100910

  1. Sreedevi P, Suresh K, Jiang G. Bacterial bioremediation of heavy metals in wastewater: a review of processes and applications. J Water Process Eng. 2022. 48: 102884.

https://doi.org/10.1016/j.jwpe.2022.102884

  1. Qin H, Hu T, Zhai Y, Lu N, Aliyeva J. The improved methods of heavy metals removal by biosorbents: A review Environ Pollut. 2020. 258:113777.

https://doi.org/10.1016/j.envpol.2019.113777

  1. Priya AK, Gnanasekaran L, Dutta K, Rajendran S, Balakrishnan D, Soto-Moscoso M. Biosorption of heavy metals by microorganisms: Evaluation of different underlying mechanisms. Chemosphere. 2022. 307: p.135957.

https://doi.org/10.1016/j.chemosphere.2022.135957

  1. Singh S, Kumar V, Datta S, Dhanjal DS, Sharma K, Samuel J, Singh J. Current advancement and future prospect of biosorbents for bioremediation. Sci Total Environ. 2020. 709: 135895.

https://doi.org/10.1016/j.scitotenv.2019.135895

  1. Ismail I, Moustafa T. Biosorption of heavy metals. Heavy metals: Sources, toxicity and remediation techniques, 2016. Nova Science Publishers, 131-174.

https://doi.org/10.2166/wst.2013.718

  1. Javanbakht V, Alavi S.A, Zilouei H. Mechanisms of heavy metal removal using microorganisms as biosorbent. Water Sci. Technol. 2014. 69(9): 1775-1787.

https://doi.org/10.1016/j.btre.2016.12.006

  1. Yılmaz Ş, Ecer Ü, Şahan T. An optimization study for bio-removal of lead from aqueous environments by alkali modified Polyporus Squamosus. MANAS J Eng. 2021. 9: 1-9.

https://doi.org/10.51354/mjen.804338

  1. Nascimento JM, Otaviano JJ, Sousa HS, Oliveira JD. Biological Method of Heavy Metal Management: Biosorption and Bioaccumulation. Heavy Metals in the Environment: Management Strategies for Global Pollution. 2023. ACS Publications, 315-60.

https://doi.org/10.1021/bk-2023-1456.ch016

  1. Al-Ansari MM, Benabdelkamel H, AlMalki RH,. Rahman AM, Alnahmi E, Masood A,.Ilavenil S, Choi KC. Effective removal of heavy metals from industrial effluent wastewater by a multi metal and drug resistant Pseudomonas aeruginosa strain RA-14 using integrated sequencing batch reactor. Environ Res. 2021. 199: 111240.

https://doi.org/10.1016/j.envres.2021.111240

  1. Jeyakumar P, Debnath C, Vijayaraghavan R, Muthuraj M. Trends in bioremediation of heavy metal contamin-ations. Environ Eng Res. 2023. 28(4): 220631.

https://doi.org/10.4491/eer.2021.631

  1. Gupta P, Diwan B. Bacterial exopolysaccharide mediated heavy metal removal: a review on biosynthesis, mechanism and remediation strategies. Biotechnol Rep. 2017. 13: 58-71.

https://doi.org/10.1016/j.btre.2016.12.006

  1. Eze K.C, Obasi NP, Inyang OA, Ewa SC. Danger of Contaminants in Foods through Phytoremediation, in Phytoremediation in Food Safety. 2024. CRC Press. 83-98.

https://doi.org/10.1201/9781032683751

  1. Diep P, Mahadevan P, Yakunin A.F. Heavy metal removal by bioaccumulation using genetically engineer-ed microorganisms. Front Bioeng Biotechnol. 2018. 6: 157.

https://doi.org/10.3389/fbioe.2018.00157

  1. Ghosh S, Sarkar B, Khumphon J, Thongmee S. Genetically Modified Microbe Mediated Metal Bioaccumulation: A Sustainable Effluent Treatment Strategy. In Industrial Wastewater Reuse: Applications, Prospects and Challenges. 2023. Singapore: Springer Nature Singapore. 215-229.

https://doi.org/10.1007/978-981-99-2489-9_11

  1. Tayang A, Songachan LS. Microbial bioremediation of heavy metals. Curr Sci. 2021. 120(6): 00113891.

https://doi.org/10.18520/cs/v120/i6/1013-1025

  1. Xiong J, Wang H, Yao J, He Q, Ma J, Yang J, Liu C, Chen Y, Huangfu X, Liu H. A critical review on sulfur reduction of aqueous selenite: Mechanisms and applica-tions. J Hazard Mater. 2022. 422: 126852.

https://doi.org/10.1016/j.jhazmat.2021.126852

  1. Kuippers G, Morris K, Townsend LT, Bots P, Kvashnina K, Bryan ND, Lloyd JR. Biomineralization of uranium-phosphates fueled by microbial degradation of isosacchar-inic acid (ISA). Environmental Science & Technology. 2021. 55(8): 4597-606.

https://doi.org/10.1021/acs.est.0c03594

  1. Zhang X, Guo Y, Liu G, Liu Y, Shi J, Hu L, Zhao L, Li Y, Yin Y, Cai Y, Jiang G. Superoxide mediated extracellular mercury reduction by aerobic marine bacterium Alteromonas Sp. KD01. Environ Sci Technol. 2023. 57(49): 20595-604.

https://doi.org/10.1021/acs.est.3c04777

  1. Janot N, Dunham-Cheatham SM, Lezama Pacheco JS, Cerrato JM, Alessi DS, Noël V, Lee E, Pham DQ. Suvorova E, Bernier-Latmani R, Williams KH. Redu-cing Conditions Influence U (IV) Accumulation in Sedi-ments during In Situ Bioremediation. ACS Earth Space Chem. 2024. 8(2): 148-58.

https://doi.org/10.1021/acsearthspacechem.3c00271

  1. Zhang S. Song M, Zhang J, Wang H. Cysteine and thiosulfate promoted cadmium immobilization in strain G303 by the formation of extracellular CdS. Sci Total Environ. 2024. 923: 171457.

https://doi.org/10.1016/j.scitotenv.2024.171457

  1. Young SD. Chemistry of heavy metals and metalloids in soils. Heavy metals in soils: Trace metals and metall-oids in soils and their bioavailability. 2013. Dordrecht: 51-95.

https://doi.org/10.1007/978-94-007-4470-7_3

  1. Dong Y, Zan J, Lin H. Bioleaching of heavy metals from metal tailings utilizing bacteria and fungi: Mechanisms, strengthen measures and development prospect. J Environ Manage. 2023. 344: 118511.

https://doi.org/10.1016/j.jenvman.2023.118511

  1. Kumari D, Qian XY, Pan X, Achal V, Li Q, Gadd GM. Microbially-induced carbonate precipitation for immob-ilization of toxic metals. Adv Appl Microbiol. 2016. 94:79-108.

https://doi.org/10.1016/bs.aambs.2015.12.002

  1. Lin H, Zhou M, Li B, Dong Y. Mechanisms, application advances and future perspectives of microbial-induced heavy metal precipitation: a review. Int Biodeter Biodegr. 2023. 178: 105544.

https://doi.org/10.1016/j.ibiod.2022.105544

  1. Hou D, O’Connor D, Igalavithana AD, Alessi DS, Luo J, Tsang DS, Sparks DL, Yamauchi Y, Rinklebe J, Ok YS. Metal contamination and bioremediation of agricul-tural soils for food safety and sustainability. Nat Rev Earth Environ. 2020. 1(7): 366-381.

https://doi.org/10.1038/s43017-020-0061-y

  1. Sarkodie E.K, Juang L, Li K, Yang J, Guo Z, Shi J, Deng Y, Liu H, Jiang H, Liang Y, Yin H. A review on the bioleaching of toxic metal (loid) s from contaminated soil: Insight into the mechanism of action and the role of influencing factors. Front Microbiol. 2022. 13:1049277.

 https://doi.org/10.3389/fmicb.2022.1049277

  1. Pineda Y.S, Devries SL, Steiner NC, Block-Cora KA. Bioleaching of gold in mine tailings by Alcaligenes faecalis. Minerals. 2023. 13(3): p. 410.

https://doi.org/10.3390/min13030410

  1. Spietz RL, Payne D, Szilagyi R, Boyd ES. Reductive biomining of pyrite by methanogens. Trends Microbiol. 2022. 30(11): 1072-1083.

https://doi.org/10.1016/j.tim.2022.05.005

  1. Newsome L, Falagán C. The microbiology of metal mine waste: Bioremediation applications and implica-tions for planetary health. Geo Health. 2021. 5(10): e20-20GH000380.

https://doi.org/10.1029/2020GH000380

  1. Yin KQ. Wang MLV, Chen L. Microorganism remedi-ation strategies towards heavy metals. Chem Eng J. 2019. 360: 1553-1563.

https://doi.org/10.1016/j.cej.2018.10.226

  1. Zhang W, Zhang H, Xu R, Qin H, Liu H, Zhao K. Heavy metal bioremediation using microbially induced carbo-nate precipitation: Key factors and enhancement strategies. Fronti Microbiol. 2023. 14: 1116970.

https://doi.org/10.3389/fmicb.2023.1116970

  1. Cameotra S.S, Makkar R.S. Biosurfactant-enhanced bioremediation of hydrophobic pollutants. Pure App Chem. 2010. 82(1): 97-116.

https://doi.org/10.1351/PAC-CON-09-02-10

  1. Hussein H.A, Khan K.M, Abdullah M.A. Mass produc-tion and factors affecting biosurfactant productivity using bioreactors in Green Sustainable Process for Chemical and Environmental Engineering and Science, 2021. Elsevier. 379-398.

https://doi.org/10.1016/B978-0-12-823380-1.00015-0

  1. Akhtar M.S, Chali B, Azam T. Bioremediation of arsenic and lead by plants and microbes from contamin-ated soil. J Plant Res. 2013. 1(3): 68-73.

https://doi.org/10.12691/plant-1-3-4

  1. Srivastava S, Agrawal S, Mondal M.K. Biological based methods for the removal of volatile organic compounds and heavy metals. In: An Innovative Role of Biofil-tration in Wastewater Treatment Plants (WWTPs). 2022. Else-vier. 331-346.

https://doi.org/10.1016/B978-0-12-823946-9.00022-X

  1. Oladoye P.O, Olowe O.M, Asemoloye M.D. Phyto-remediation technology and food security impacts of heavy metal contaminated soils: A review of literature. Chemosphere. 2022. 288:132555.

https://doi.org/10.1016/j.chemosphere.2021.132555

  1. Bhantana P, Rana MS, Sun XC, Moussa MG, Saleem MH, Syaifudin M, Shah A, Poudel A, Pun AB, Bhat MA, Mandal DL. Arbuscular mycorrhizal fungi and its major role in plant growth, zinc nutrition, phosphorous regulation and phytoremediation. Symbiosis. 2021. 84: 19-37.

https://doi.org/10.1007/s13199-021-00756-6

  1. Mani D, Kumar C. Biotechnological advances in bioremediation of heavy metals contaminated eco-systems: an overview with special reference to phyto-remediation. Int J Sci Environ Technol. 2014. 11: 843-872.

https://doi.org/10.1007/s13762-013-0299-8

  1. Tyagi B, Kumar N. Bioremediation: Principles and applications in environmental management. In Biorem-ediation for Environmental Sustainability. 2021. Else-vier. 3-28.
  2. Chaturvedi A, Saraswat P, Gupta A, Prasad M, Ranjan R. Detoxification of Sewage Sludge by Natural Attenuation and Application as a Fertilizer in Sustain-able Management and Utilization of Sewage Sludge. 2022. Springer, Cham. 263-80.

https://doi.org/10.1007/978-3-030-85226-9_13

  1. Thornton SF, Spence MJ, Bottrell SH, Spence KH. Natural attenuation of dissolved petroleum fuel const-ituents in a fractured Chalk aquifer: contaminant mass balance with probabilistic analysis. Q J Eng Geol Hydrogeol. 2024. 57(2): qjegh2023-116.

https://doi.org/10.1144/qjegh2023-116

  1. Tian L, Guan W, Ji Y, He X, Chen W, Alvarez PJ, Zhang T. Microbial methylation potential of mercury sulfide particles dictated by surface structure. Nat. Geo-sci. 2021. 14(6): 409-416.

https://doi.org/10.1038/s41561-021-00735-y

  1. Abdulsalam S, Omale A.B. Comparison of biostimulation and bioaugmentation techniques for the remediation of used motor oil contaminated soil. Braz Arch Biol Technol. 2009. 52(3): 747-754.

https://doi.org/10.1590/S1516-89132009000300027

  1. Juárez-Maldonado A, Tortella G, Rubilar O, Fincheira P, Benavides-Mendoza A. Biostimulation and toxicity: The magnitude of the impact of nanomaterials in microorganisms and plants. J Adv Res. 2021. 31: 113-126.

https://doi.org/10.1016/j.jare.2020.12.011

  1. Ahmad I, Abdullah N, Koji I, Yuzir A, Mohamad SE, Show PL, Cheah WY, Khoo KS. The role of restaurant wastewater for producing bioenergy towards a circular bioeconomy: A review on composition, environmental impacts and sustainable integrated management. Environ Res. 2022. 214 (Pt 2): 113854.

https://doi.org/10.1016/j.envres.2022.113854

  1. Muter O. Current Trends in Bioaugmentation Tools for Bioremediation: A Critical Review of Advances and Knowledge Gaps. Microorganisms, 2023. 11(3): 710.

https://doi.org/10.3390/microorganisms11030710

  1. Khan A, Zytner R. Degradation Rates for Petroleum Hydrocarbons Undergoing Bioventing at the Meso-Scale. Bioremediation J. 2013. 17(3): 159-172.

https://doi.org/10.1080/10889868.2013.807772

  1. Mosco M, Zytner R. Large Scale Bioventing Degradation Rates of Petroleum Hydrocarbons and Determination of Scale-up Factors. Bioremediation J. 2017. 21(3-4): 149-62.

https://doi.org/10.1080/10889868.2017.1312265

  1. Song P, Xu D, Yue J, Ma Y, Dong S, Feng J. Recent advances in soil remediation technology for heavy metal contaminated sites: A critical review. Sci Total Environ. 2022. 838:156417.

https://doi.org/10.1016/j.scitotenv.2022.156417

  1. Macaulay B, Rees D. Bioremediation of oil spills: A review of challenges for research advancement. J Environ Sci Stud. 2014. 8: p. 9-37.
  2. Hsia KF, Chen CC, Ou JH, Lo KH, Sheu YT, Kao CM, Treatment of petroleum hydrocarbon-polluted ground-water with innovative in situ sulfate-releasing biobarrier. J Clean Prod. 2021. 295:126424.

https://doi.org/10.1016/j.jclepro.2021.126424

  1. Fragkou E, Antoniou E, Daliakopoulos I, Manios T, Theodorakopoulou M, Kalogerakis N. In situ aerobic bioremediation of sediments polluted with petroleum hydrocarbons: a critical review. J Mar Sci Eng. 2021. 14(9): 1003.

https://doi.org/10.3390/jmse9091003

  1. Sinha P, Mukherji S. Biofiltration Process for Treatment of Water and Wastewater. Trans Ind Nat Acad Eng. 2022. 7(4): 069-91.

https://doi.org/10.1007/s41403-022-00360-0

  1. Taherzadeh MJ. Bioengineering to tackle environ-mental challenges, climate changes and resource recov-ery. Bioeng. 2019. 10(1): 698-699.

https://doi.org/10.1080/21655979.2019.1705065

  1. Pachaiappan R, Cornejo-Ponce L, Rajendran K, Manavalan K, Femilaa Rajan V, Awad F. A review on biofiltration techniques: recent advancements in the removal of volatile organic compounds and heavy metals in the treatment of polluted water. Bioeng. 2022. 13(4): 8432-8477.

https://doi.org/10.1080/21655979.2022.2050538

  1. Rafeeq H, Afsheen N, Rafique S, Arshad A, Intisar M, Hussain A. Bilal M, Iqbal HM. Genetically engin-eered microorganisms for environmental remediation. Chemosphere. 2023. 310:136751.

https://doi.org/10.1016/j.chemosphere.2022.136751

  1. Singh Y, Saxena MK. Insights into the recent advances in nano-bioremediation of pesticides from the contaminated soil. Front Microbiol. 2022. 13: 982611.

https://doi.org/10.3389/fmicb.2022.982611

  1. Mishra M., Singh SK. Kumar A. Environmental factors affecting the bioremediation potential of microb-es. In Microbe Mediated Remediation of Environmental Contaminants, Woodhead publishing series in Food Science, Technology and Nutrition. 2021. 47-58.

https://doi.org/10.1016/B978-0-12-821199-1.00005-5

  1. Dzionek A, Wojcieszyńska D, Guzik U. Natural carriers in bioremediation: A review. Electron J Biotech-nol. 2016. 23:28-36.

https://doi.org/10.1016/j.ejbt.2016.07.003