Cover page


Review Article


Approach to Neurometabolic Diseases from a Pediatric Neurological Point of View

Parvaneh KARIMZADEH

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 1-16
https://doi.org/10.22037/ijcn.v9i1.7979

How to Cite This Article: Karimzadeh P. Approach to Neurometabolic Diseases from a Pediatric Neurological Point of View. Iran J Child Neurol. 2015 Winter;9(1): 1-16.

 

Abstract
Objective
Neurometabolic disorders are an important group of diseases that mostly are
presented in newborns and infants.
Neurological manifestations are the prominent signs and symptoms in this group
of diseases. Seizures are a common sign and are often refractory to antiepileptic drugs in untreated neurometabolic patients.
The onset of symptoms for neurometabolic disorders appears after an interval of
normal or near normal growth and development.Additionally, affected children
may fare well until a catabolic crisis occurs.
Patients with neurometabolic disorders during metabolic decompensation have
severe clinical presentation, which include poor feeding, vomiting, lethargy,
seizures, and loss of consciousness.
This symptom is often fatal but severe neurological insult and regression in
neurodevelopmental milestones can result as a prominent sign in patients who
survived.
Acute symptoms should be immediately treated regardless of the cause.
A number of patients with neurometabolic disorders respond favorably and, in
some instances, dramatically respond to treatment.
Early detection and early intervention is invaluable in some patients to prevent
catabolism and normal or near normal neurodevelopmental milestones.
This paper discusses neurometabolic disorders, approaches to this group of
diseases (from the view of a pediatric neurologist), clinical and neurological
manifestations, neuroimaging and electroencephalography findings, early
detection, and early treatment.

 

References

  1. Filiano JJ. Neurometabolic Disease in the Newborn. ClinPerinatol 33, 2006, 411-479.
  2. Van Karnebeek CDM, Stockler S. Treatable inborn errors of metabolism causing intellectual disability: A systematic literature review. Molecular Genetics and Metabolism, 105, 2012, 369-381.
  3. Abdel-Salam GMH, Abdel-Kader AA, Effat L, Gouda A, Hindawy A, El-Gammal MA. Clinical, Electroencephalographic (EEG), Neuroradiological and Molecular Correlations in Late-Detected Phenylketonuria (PKU) Patients.Egypt J. Neurol. Psychiat. Neurosurg., 2005, 42(2):391-406.
  4. Karimzadeh P, Ahmadabadi F, Jafari N, Shariatmadari F, Nemati H, Ahadi A, KarimiDardashti S, Mirzarahimi M, Dastborhan Z, ZareNoghabi J. Study on MRI Changes in Phenylketonuria in Patients Referred to Mofid Hospital/ Iran. Iran J Child Neurol. 2014 spring 8(2):53-56.
  5. Karimzadeh P, Jafari N, Ahmad Abadi F, Jabbedari S, Taghdiri MM, Alaee MR, Ghofrani M, Tonekaboni SH, NejadBiglari H. Propionic Acidemia: Diagnosis and Neuroimaging Findings of This Neurometabolic Disorder. Iran J Child Neurol. 2014 Winter; 8(1):58-61.
  6. Ozand PT, Rashed M, Gascon GG, Youssef NG, Harfi H, Rahbeeni Z, et al. Unusual presentations of propionic acidemia. Brain Dev 1994;16(Suppl):46-57 .
  7. Karimzadeh P, Jafari N, Jabbehdari S, Taghdiri MM, Nemati H, Saket S, Alaee MR, Ghofrani M, Tonakebni SH. Methylmalonicacidemia: Diagnosis and Neuroimaging Findings of This Neurometabolic Disorder (An Iranian Pediatric Case Series). Iran J Child Neurol. 2013 Summer; 7(3): 63-66.
  8. Radmanesh A, Zaman T, Ghanaati H, Molaei S, Robertson RL, ZamaniAA. Methylmalonicacidemia: brain-imaging findings in 52 children and a review of the literature. PediatrRadiol 2008 Oct;38(10):1054-6.
  9. Karimzadeh P, Ahmadabadi F, Jafari N, Jabbehdari S, Alaee MR, Ghofrani M, Taghdiri MM, Tonekaboni SH. Biotinidase Deficiency: A Reversible Neurometabolic Disorder (An Iranian Pediatric Case Series). Iran J Child Neurol. 2013 Autumn; 7(4):47- 52.
  10. Karimzadeh P, Pirzadeh Z, Ahmadabadi F, Jafari N, Jabbehdari S, Nemati H, Ghofrani M, Taghdiri MM, Tonekaboni SH, Mohammad-Reza Sharbatdaralaei. Glutaricaciduria type 1: diagnosis and neuroimaging findings of this neurometabolic disorder in an Iranian pediatric case series. International Journal of Developmental Disabilities 05/2014; DOI: 10.1179/2047387714Y.0000000039.
  11. Bernier FP, Boneh A, Dennett X, Chow CW, Cleary MA, Thorburn DR. Diagnostic criteria for respiratory chain disorders in adults and children. Neurology. 2002;59:1406–11.
  12. Wolf NI, Smeitink AM. Mitochondrial disorders: A proposal for consensus diagnostic criteria in infants and children. Neurology. 2002;59:1402–5.
  13. Majamaa K, Moilanen JS, Uimonen S, et al. Epidemiology of A3243G, the mutation for mitochondrial encephalomyopathy, lactic acidosis, and strokelike episodes: Prevalence of the mutation in an adult population. Am J Hum Genet. 1998;63:447–54.
  14. Lamont PJ, Surtees R, Woodward CE, Leonard JV, Wood NW, Harding AE. Clinical and laboratory findings in referrals for mitochondrial DNA analysis. Arch Dis Child, Arch Dis Child. 1998 Jul;79(1):22-7.
  15. Ferri R. Neurometabolic Disorders.Arch Neurol. 2005;62:1045-1046.
  16. PapettiL, ParisiP, LeuzziV, NardecchiaF, NicitaF, UrsittiF, Marra F, Paolino MC, Spalice A. Metabolic epilepsy: an update. Brain & Development. 2013 Oct;35(9):827-41.
  17. Salpietro V, Phadke R, Saggar A, Hargreaves IP, Yates R, Fokoloros C, Mankad K, Hertecant J, Ruggieri M, McCormick D, KinaliM. Zellweger syndrome and secondary mitochondrial myopathy. Eur J Pediatr. 2014 Oct 7 (Article in press).
  18. Schulz A, Kohlschütter A. NCL Disorders: Frequent Causes of Childhood Dementia. Iran J Child Neurol.2013 Winter; 7(1):1-8.
  19. Jalanko A, Braulke T. Neuronal ceroidlipofuscinoses. Biochim Biophys Acta 2009;1793:697-709.
  20. Karimzadeh P, Jafari N, NejadBiglari H, Jabbeh Dari S, Ahmad Abadi F, Alaee MR, Nemati H,  SaketS, Tonekaboni SH, Taghdiri MM, Ghofrani M. GM2-Gangliosidosis (Sandhoff and Tay-Sachs disease): Diagnosis and Neuroimaging Findings(An Iranian Pediatric Case Series). Iran J Child Neurol. 2014 Summer;8(3): 55-60.
  21. Vanier MT, Duthel S, Rodriguez-Lafrasse C, et al. Genetic heterogeneity in Niemann-Pick C disease: a study using somatic cell hybridization and linkage analysis. Am J Hum Genet. 1996;58(1):118-125.
  22. Vanier MT. Niemann-Pick disease type C. Orphanet J Rare Dis. 2010;5:16.
  23. Meikle PJ, Hopwood JJ, Clague AE, et al. Prevalence of lysosomal storage disorders. JAMA. 1999; 281(3):249-254.
  24. Patterson MC, Hendriksz CJ, Walterfang M, et al. Recommendations for the diagnosis and management of Niemann-Pick disease type C: an update. Mol Genet Metab. 2012 ;106(3):330-3.
  25. Karimzadeh P, Tonekaboni SH, Ashrafi MR, Shafeghati Y, Rezayi A, Salehpour S, Ghofrani M, Taghdiri MM, Rahmanifar A, Zaman T, Aryani O, Shoar BN, Shiva F, Tavasoli A, and Houshmand M. Effects of Miglustat on Stabilization of Neurological Disorder in Niemann-Pick Disease Type C: Iranian Pediatric Case Series.J Child Neurol. 2013 Dec;28(12):1599-606.
  26. Gropman A. Imaging of Neurogenetic and Neurometabolic Disorders of childhood.Current Neurology and NeuroscienceReport. 2004. 4:139-146.
  27. Youssef-Turki IB, Kraoua I, Smirani S, Mariem K, Rhouma HB, Rouissi A, Gouider-Khouja N. Epilepsy Aspects and EEG Patterns in Neuro-Metabolic Diseases, Journal of Behavioral and Brain Science.2011. 1: 69-74.
  28. PapettiL, ParisiP, LeuzziV, Nardecchiac F, NicitaF, UrsittiF, MarraF, PaolinoMC,SpaliceA. Metabolic epilepsy: an update. Brain & Development. Article in press.Nov 2012.

Comparison of Serum Zinc Levels among Children with Simple Febrile Seizure and Control Group: A Systematic Review

Mohammad Mehdi NASEHI, Roya SAKHAEI, mahmood MOOSAZADEH*, Maryam ALIRAMZANY

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 17-24
https://doi.org/10.22037/ijcn.v9i1.5634

How to Cite This Article: Nasehi MM, Sakhaei R, Moosazadeh M, Aliramzany M. Comparison of Serum Zinc Levels among Children with Simple Febrile Seizure and Control Group: A Systematic Review. Iran J Child Neurol. 2015 Winter;9(1):17-24 .

Abstract
Objective
Several factors are involved in the etiology of febrile seizure (FS), among them
is zinc (Zn), which has been discussed in various studies. The present systematic review compares Zn levels in children with FS and a control group.

Materials & Methods
We searched keywords of febrile seizure, febrile convulsion, children, childhood,
fever, trace elements, risk factor, predisposing, zinc, Zn, and epilepsy in the
following databases: SCOPUS, PubMed, and Google Scholar. The quality of
research papers was assessed using a checklist. Data was extracted from primary
studies based on demographic variables and amounts of Zn in case and control
groups.

Results
Twenty primary studies were entered in the present study. Of which, eighteen
studies, reported that Zn serum levels were significantly lower in the case group
(patients with FS) than the control group.

Conclusion
The present systematic review indicated that Zn is one factor for predicting FS.
A low level of this element among children can be regarded as a contributing
factor for FS, a conclusion with a high consensus among different studies carried
out in different parts of the world.

 

References

  1. Heydarian F, Ashrafzadeh F, Ghasemian A. Serum ZINC level in Patients with simple febrile seizure. Iran J Child Neurology 2010; 14(2):41-44.
  2. Mahyar A, Pahlavan AA, Varasteh-Nejad A. Serum zinc level in children with febrile seizure. Acta Medica Iranica 2008; 46(6): 477-80.
  3. Kunda GK, Rabin F, Nandi ER, Sheikh N, Akhter S. Etiology and Risk Factors of Febrile Seizure – An Update. Bangladesh J Child Health 2010; 34 (3):103-112.
  4. Abbaskhaniyan A, Shokrzadeh M, Rafati MR, Mashhadiakabr M, Arab A, Yazdani J. Survey and Relation of Serum Magnesium Level in Children with Seizure. J Mazand Univ Med Sci 2012; 2(90): 43-49.
  5. Abaskhanian A, Vahid Shahi K, Parvinnejad N. The Association between Iron Deficiency and the First Episode of Febrile Seizure. J Babol Univ Med Sci 2009; 11(3):32-36.
  6. Amiri M, Farzin L, Moassesi ME, Sajadi F. Serum Trace Element Levels in Febrile Convulsion. Biol Trace Elem Res 2010; 135:38–44.
  7. Fetveit A. Assessment of febrile seizures in children. Euro J Pedia 2008; 167:17-27.
  8. Sadeghzadeh M, Khoshnevis P, Mahboubi E. Iron Status and Febrile Seizure- A Case Control Study in Children Less Than 3 Years. Iran J Child Neurol 2012; 6(4):27-31.
  9. Salehiomran MR, Mahzari M. Zinc Status in Febrile Seizure: A Case-Control Study. Iran J Child Neurol. 2013; 7(4):20-23.
  10. Shiva S, Barzegar M, Zokaie N, Shiva Sh. Dose Supplemental Zinc Prevents Recurrence of Febrile Seizures?. Iran J Child Neurol 2011; 5(4):11-14.
  11. Sadeghzadeh M, Khoshnevis Asl P, Mousavinasab N, Koosha A, Norouzi M. The Relation Between Serum Zinc Level and Febrile Seizures in Children Admitted to Zanjan Valie-Asr Hospital. ZUMS Journal 2011; 19 (74):17-24.
  12. Waruiru C, Appleton R. febrile seizures: an update. Archive Dis Child 2004; 89: 751-6.
  13. Moosazadeh M, Nekoei-moghadam M, Emrani Z, Amiresmaili M. Prevalence of unwanted pregnancy in Iran: A systematic review and meta-analysis. Int J Health Plann Mgmt, 2013; Published online in Wiley Online Library, DOI: 10.1002/hpm.2184. available at: http://onlinelibrary.wiley.com/journal/10.1002/%28ISSN%291099-1751/earlyview.
  14. Elm EV, Altman DG, Egger M, Pocock SJ, Gotzsche PC, Vandenbroucke P, et al. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies. Preventive medicine 2007; 45(4): 247-251.
  15. Shea BJ, Grimshaw JM, Wells GA, Boers M, Andersson N, Hamel C, et al. Development of AMSTAR: a measurement tool to assess the methodological quality of systematic reviews. BMC medical research methodology 2007; 7(1):10.
  16. Garty BZ, Olomucki R, Lerman-Sagie T, Nitzan M. Cerebrospinal fluid zinc concentration in febrile children’s. Arch Dis Child 1995; 73(4):338-41.
  17. Papierkowski A, Mroczkowska-Juchkiewics A, Pawlowska KA, Pasternak K. Magnesium and zinc levels in blood serum and cerebrospinal fluid in children with febrile convulsions. Pol Merkuriusz Le 1999; 6(33): 138-40.
  18. Burhanoglu M, Tutuncuoglu S, Coker C, Tekgul H, Ozgur T. Hypozincemia in febrile convulsion. Eur J Pediatr 1996; 155(6):498-501.
  19. Kumar L, Chaurasiya OS, Gupta AH. Prospective Study of Level of Serum Zinc In Patients of Febrile Seizures, Idiopathic Epilepsy, and CNS Infections. People’s Journal of Scientific Research 2011; 4(2):1-4.
  20. Palliana RR, Singh DK, Borade A. ZINC deficiency as a risk factor febrile seizure. Pediatrics 2008; 121(2):358-365.
  21. Ehsanipour F, Talebitaher M, Vahid Harandi N, Kani K. Serum Zinc Level in Children with Febrile Convulsion and its Comparison with that of Control Group. Iran J Pediatr 2009; 19)1):65-68.
  22. Kafadar I, Akıncı AB, Pekun F, Adal E. The Role of Serum Zinc Level in Febrile Convulsion Etiology. J Pediatr Inf 2012; 6: 90-93.
  23. Lee JH, Kim JH. Comparison of Serum Zinc Levels Measured by Inductively Coupled Plasma Mass Spectrometry in Preschool Children with Febrile and Afebrile Seizures. Ann Lab Med 2012; 32:190-193.
  24. Margaretha L, Masloman N. Correlation between serum zinc level and simple febrile seizure in children. Paediatr Indones 2010; 50(6): 326-330.
  25. Mollah MAH, Rakshit SC, Anwar KS, Arslan MI, Saha N, Ahmed S, et al. Zinc concentration in serum and cerebrospinal fluid simultaneously decrease in children with febrile seizure: Findings from a prospective study in Bangladesh. Acta Pædiatrica 2008; 97:1707–1711.
  26. Mollah MAH, Ranjan DP, Tarafdar SA, et al. Zinc in CSF of patients with febrile convulsion. Ind J Pediatr 2002; 69: 859-61.
  27. Modarresi MR, Shahkarami SMA, Yaghini O, Shahbi J, Mosaiiebi D, Mahmoodian T. The relationship between Zinc deficiency and Febrile Convulsion in Isfahan, Iran. Iran J Child Neurology 2011; 5(2):27-31.
  28. Tutuncuoglu S, Kutukculer N, Kepe L, Coker C, Berdeli A, Tekgul H. Proinflammatory cytokines, prostaglandins and zinc in febrile Convulsions. Pediatrics International 2001; 43: 235–239.
  29. Talebian A, Vakili Z, Talar SA, Kazemi SM, Mousavi GA. Assessment of the Relation between Serum Zinc & Magnesium Levels in Children with Febrile Convulsion. Iranian Journal of Pathology 2009; 4 (4), 157 – 160.
  30. Gunduz Z, Yavuz I, Koparal M, Kumandas S, Saraymen R. Serum and cerebrospinal fluid zinc level in children with febrile convulsion. Acta Paediatr Jpn 1996; 38(3): 237-41.
  31. Cho WJ, Son BH, Kim SW. Levels of Sodium and Zinc Concentration in Febrile Convulsion. J Korean Child Neurol Soc 1999; 7(2):214-219.
  32. Cho HS, Shin JH, Seo JY, Lee CA, Kim SH, Chae KY. The Levels of Zinc and Neuron-specific Enolase in Febrile Convulsion. Korean J Pediatr 2004; 47(10):1087-1092.
  33. Ganesh R, Janakiraman L, Meenakshi B. Serum zinc levels are low in children with simple febrile seizures compared with those in children with epileptic seizures and controls. Ann Trop Paediatr 2011; 31(4):345-9.
  34. Ganesh R, Janakiraman L. Serum zinc levels in children with simple febrile seizure. Clin Pediatr (Phila) 2008; 47(2):164-6.
  35. Okposio MM, Sadoh WE, Ofovwe GE, Onyiriuka AN. Serum zinc level in Nigerian children with febrile convulsion. Journal of Pediatric Neurology 2012; 10(3):187-191.
  36. Itoh M, Ebadi M. The selective inhibition of hippocampal glutamic acid decarboxylase in zinc induced epileptic seizures. Neurochem Res 1982; 7(10): 1287-98.
  37. Peters S, Koh J, Choi W. Zinc selectively blocks the action of N-methyl-D-aspartate on cortical neurons. Science 1987; 236 (4801): 589-93.

Research Article


Diagnostic accuracy of frozen section in Central nervous system lesions, a 10-year study.

Maliheh KHODDAMI*, Ali AKBARZADEH, Afshin MORDAI, Farahnaz BIDARI-ZEREHPOUSH, Hamid ALIPOUR, Sara SAMADZADEH, Bijan ALIPOUR

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 25-30
https://doi.org/10.22037/ijcn.v9i1.6073

How to Cite This Article: Khoddami M, Akbarzadeh A, Mordai A, Bidari Zerehpoush F, Alipour H, Samadzadeh S, Alipour B.Diagnostic Accuracy of Frozen Section of Central Nervous System Lesions: A 10-Year Study. Iran J Child Neurol. 2015 Winter;9(1):25-30.

 

Abstract

Objective

Definitive diagnosis of the central nervous system (CNS) lesions is unknown prior to histopathological examination. To determine the method and the endpoint for surgery, intraoperative evaluation of the lesion helps the surgeon.

In this study, the diagnostic accuracy and pitfalls of using frozen section (FS) of

CNS lesions is determined.

Materials & Methods

In this retrospective study, we analyzed the results of FS and permanent diagnoses of all CNS lesions by reviewing reports from 3 general hospitals between March 2001 and March 2011.

Results

273 cases were reviewed and patients with an age range from 3 to 77 years of age were considered. 166 (60.4%) had complete concordance between FS and permanent section diagnosis, 83 (30.2%) had partial concordance, and 24 cases (9.5%) were discordant. Considering the concordant and partially concordant cases, the accuracy rate was 99.5%, sensitivity was 91.4%, specificity was 99.7%, and positive and negative predictive values were 88.4% and 99.8%, respectively.

Conclusion

Our results show high sensitivity and specificity of FS diagnosis in the evaluation of CNS lesions. A Kappa agreement score of 0.88 shows high concordance for FS results with permanent section. Pathologist’s misinterpretation, small biopsy samples (not representative of the entire tumor), suboptimal slides, and inadequate information about tumor location and radiologic findings appear to be the major causes for these discrepancies indicated from our study.

 References

  1. Taxy JB, Anthony G. Biopsy interpretation: the frozen section. 1st ed. China: Lippincott Williams & Wilkins; 2010. P.301-3.
  2. Somerset HL, Kleinschmidt-DeMasters BK. Approach to the intraoperative consultation for neurosurgical specimens. Adv Anat Pathol 2011; 18:446-9. doi: 10.1097/ PAP.0b013e3182169934.
  3. Regragui A, Amarti Riffi A, Maher M, El Khamlichi A, Saidi A. Accuracy of Intraoperative diagnosis in central nervous system tumors: report of 1315 cases. Neurochirurgie 2003; 49(2-3 Pt 1):67-72.
  4. Plesec TP, Prayson RA. Frozen section discrepancy in the evaluation of central nervous system tumors. Arch Pathol Lab Med 2007; 131:1532-40.
  5. Savargaonkar P, Farmer PM. Utility of intra-operative consultations for the diagnosis of central nervous system lesions. Ann Clin Lab Sci 2001; 31:133-9.
  6. Talan-Hraniloviæ J, Vuèiæ M, Ulamec M, Belicza M. Intraoperative frozen section analysis in of the central nervous system and pituitary gland pathology. Acta Clin Croat 2005; 44:217-21.
  7. Roessler K, Dietrich W, Kitz K. High diagnostic accuracy of cytologic smears of central nervous system tumors. A 15-year experience based on 4,172 patients. Acta Cytol 2002; 46:667-74.
  8. Ud Din N, Memon A, Idress R, Ahmad Z, Hasan S. Central Nervous System Lesions: Correlation of  Intraoperative and Final Diagnoses, Six Year Experience at a Referral Centre in a Developing Country, Pakistan. Asian Pac J Cancer Prev 2011; 12:1435-7.
  9. Burger PC, Scheithauer BW. Tumors of the Central Nervous System. In: AFIP Atlas of Tumor Pathology Series 4. Washington DC: American Registry of Pathology; 2007.
  10. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO Classification of Tumours of the Central Nervous System. Acta Neuropathol. 2007; 114: 97–109. doi: 10.1007/s00401- 007-0243-4


Spinal Fluid Lactate Dehydrogenase Level Differentiates between Structural and Metabolic Etiologies of Altered Mental Status in Children

Nahid KHOSROSHAHI, Parastoo ALIZADEH, Mehdi KHOSRAVI, Peyman SALAMATI, Kamyar KAMRANI*

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 31-36
https://doi.org/10.22037/ijcn.v9i1.5456

How to Cite This Article: Khosroshahi N, Alizadeh P, Khosravi M, Salamati P, Kamrani K. Spinal Fluid Lactate Dehydrogenase Level Differentiates between Structural and Metabolic Etiologies of Altered Mental Status in Children. Iran J Child Neurol. 2015 Winter;9(1):31-36.

Abstract
Objective
Altered mental status is a common cause of intensive care unit admission in
children. Differentiating structural causes of altered mental status from metabolic etiologies is of utmost importance in diagnostic approach and management of the patients. Among many biomarkers proposed to help stratifying patients with altered mental status, spinal fluid lactate dehydrogenase appears to be the most promising biomarker to predict cellular necrosis.


Materials & Methods
In this cross sectional study we measured spinal fluid level of lactate
dehydrogenase in children 2 months to 12 years of age admitted to a single center intensive care unit over one year. Spinal fluid level of lactate dehydrogenase in 40 pediatric cases of febrile seizure was also determined as the control group.


Results
The study group included 35 boys (58.3%) and 25 girls (41.7%). Their mean
age was 2.7+/-3 years and their mean spinal fluid lactate dehydrogenase level
was 613.8+/-190.4 units/liter. The control group included 24 boys (55.8%) and
19 girls (44.2%). Their mean age was 1.3+/-1.2 years and their mean spinal
fluid lactate dehydrogenase level was 18.9+/-7.5 units/liter. The mean spinal
fluid lactate dehydrogenase level in children with abnormal head CT scan was
246.3+/-351.5 units/liter compared to 164.5+/-705.7 in those with normal CT
scan of the head (p=0.001).

Conclusion
Spinal fluid lactate dehydrogenase level is useful in differentiating structural and
metabolic causes of altered mental status in children.

 References

  1. Fesk SK. Coma and confusional states: emergency diagnosis and management. Neurol Clin 1998; 16: 237- 56.
  2. Cucchiara BL, Kanser SE, Wolk DA, et al. Early impairment in consciousness Predicts mortality after hemispheric ischemic stroke. Crit care med 2004; 32: 241-5.
  3. Teasdale G, Jennett B. Assessment of coma and impaired consciousness: a practical scale. Lancet 1974; 2: 81-4.
  4. Wityk RJ, Stern BJ. Ischemic stroke: today and tomorrow. Crit care med 1994; 22: 1278-93.
  5. Vázquez Jorge Alejandro, Adducci Maria del Carmen, Monzón Daniel Godoy, Iserson Kenneth V. Lactic dehyrogenase in cerebrospinal fluid may differentiate between structural and non-strucfiular central nervous system lesion in patient with diminished levels of consciousness. The Journal of Emergency Medicine2009; 37(1): 93–97.
  6. Kärkelä J, Pasanen M, Kaukinen S, Mörsky P, Harmoinen A. Evaluation of hypoxic brain injury with spinal fluid enzymes, lactate, and pyruvate. Crit Care Med. 1992 Mar; 20(3):378-86. 2007: pp. 835. ISBN 0-7817-7087-4.
  7. DV Kamat, BP Chakravorty. Comparative values of CSF-LDH isoenzymes in neurological disorders. Indian Journal of Medical Sciences 1999; 53 (1): 1-6.
  8. Pollak AN, Gupton CL. Emergency Care and Transportation of the Sick and Injured. Boston: Jones and Bartlett 2002: pp. 140. ISBN 0-7637-1666-9.
  9. Nayak BS, Bhat R. Cerebrospinal fluid lactate dehydrogenase and glutamine in meningitis. Indian J Physiol Pharmacol. 2005 Jan; 49(1):108-10.
  10. A Twijnstra, A P van Zanten, A A Hart, et al. al. Serial lumbar and ventricle cerebrospinal fluid lactate dehydrogenase activities in patients with leptomeningeal metastases from solid and haematological tumours. J Neurol Neurosurg Psychiatry 1987 50: 313-320.
  11. Nussinovitch M, Finkelstein Y, Politi K, Harel D, Klinger G, Razon Y, Nussinovitch U, Nussinovitch N. Cerebrospinal fluid lactate dehydrogenase isoenzymes in children with bacterial and aseptic meningitis. Translational Research 2009. 154 (4): 214-218.
  12. Feldman William E. Cerebrospinal Fluid Lactic Acid Dehydrogenase Activity. Levels in Untreated and Partially Antibiotic-Treated Meningitis. Am J Dis Child. 1975; 129(1): 77-80.
  13. Lutsar I, Haldre S, Topman M, Talvik T. Enzymatic changes in the cerebrospinal fluid in patients with infections ofthe central nervous system. Acta Paediatr 1994; 83(11):1146-1150.
  14. Kepa L, Oczko-Grzesik B, Błedowski D. Evaluation of cerebrospinal fluid and plasma lactate dehydrogenase activity in patients with purulent, bacterial meningoencephalitis. Przegl Epidemiol. 2006; 60(2):291-8.
  15. Ruzak-Skocir B, Trbojevic-Cepe M. Study of serum and cerebrospinal fluid enzymes in diagnosis and differential diagnosis of cerebrovascular diseases. Neurologija. 1990; 39(4):239-50.
  16. Nand N, Sharma M, Saini DS. Evaluation of lactic dehydrogenase in cases of meningitis. Indian J Med Sci. 1993; 47(4): 96-100.
  17. Neches William, Platt Martin. Cerebrospinal Fluid LDH in 287 Children, Including 53 Cases of Meningitis of Bacterial and non-Bacterial Etiology. Pediatrics 1968; 41:1097-1103.
  18. Engelke S; Bridgers S, Saldanha R, Trought W. Cerebrospinal Fluid Lactate Dehydrogenase in Neonatal Intracranial Hemorrhage. American Journal of the Medical Sciences 1986; 291 (6): 391-395.
  19. Parakh N, Gupta HL, Jain A. Evaluation of enzymes in serum and cerebrospinal fluid in cases of stroke. Neurology India 2002; 50 (4): 518-9.
  20. Lampl Y, Paniri Y, Eshel Y, Sarova-Pinhas I. Cerebrospinal fluid lactate dehydrogenase levels in early stroke and transient ischemic attacks. Stroke 1990; 21: 854-857.
  21. Hall Robert T., Kulkarni Prakash B., Sheehan Michael B., Rhodes Philip G. Cerebrospinal Fluid Lactate Dehydrogenase in Infants with Perinatal Asphyxia. Developmental Medicine & Child Neurology 1980. 22 (3): 300-307.
  22. Nussinovitch M, Volovitz B, Finkelstein Y, Amir J, Harel D. Lactic dehydrogenase isoenzymes in cerebrospinal fluid associated with hydrocephalus. Acta Paediatr 2001; 90: 972-974.

The POLG Polyglutamine Tract Variants in Iranian Patients with Multiple Sclerosis

Mehri KHATAMI, Mohammad Mehdi HEIDARI, Reza MANSOURI, Fatemeh MOUSAVI

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 37-41
https://doi.org/10.22037/ijcn.v9i1.5816

How to Cite This Article: Khatami M, Heidari MM, Mansouri R, Mousavi F. The POLG Polyglutamine Tract Variants in Iranian Patients with Multiple Sclerosis. Iran J Child Neurol. 2015 Winter; 9(1):37-41.

Abstract
Objective
Multiple Sclerosis (MS) is a common disease of the central nervous system. The interaction between inflammatory and neurodegenerative processes typically results in irregular neurological disturbances followed by progressive disability.
Mitochondrial dysfunction has been implicated in neurodegenerative disorders. 
The DNA polymerase-gamma (POLG) gene, which encodes the catalytic subunit of enzyme responsible for directing mtDNA replication, contains a poly glutamine tract (poly-Q) in the N-terminal, encoded by a CAG sequence in exon 2.

Materials & Methods
We analyzed the POLG trinucleotide repeats in 40 Iranian patients with MS (27 females and 13 males with an age range of 18–55); and 47 healthy age, gender, and ethnic matched controls were chosen by PCR-SSCP analysis. 

Results
Our results indicated that the most common allele in patients had 10 consecutive CAG repeats (10Q). Other alleles of 11and 12 trinucleotide repeats were detected.
We did not find any difference between the CAG repeat length distribution in controls and MS patients.

Conclusion
No correlation was observed in the POLG gene CAG repeat with pathogenesis of MS, but it looks that other point mutations in POLG gene may have an important role in the disease’s pathogenesis and produced more significant results.

References

  1. Baranzini SE. Revealing the genetic basis of multiple sclerosis: are we there yet? Curr Opin Genet Dev. 2011 Jun; 21(3):317-24.
  2. Hoffjan S, Akkad DA. The genetics of multiple sclerosis: an update 2010. Mol Cell Probes. 2010 Oct; 24(5):237-43.
  3. Disanto G, Berlanga AJ, Handel AE, Para AE, Burrell AM, Fries A, et al. Heterogeneity in multiple sclerosis: scratching the surface of a complex disease. Autoimmune Dis. 2010; 2011:932351.
  4. International Multiple Sclerosis Genetics C, Wellcome Trust Case Control C, Sawcer S, Hellenthal G, Pirinen M, Spencer CC, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011 Aug 11; 476(7359):214-9.
  5. Mao P, Reddy PH. Is multiple sclerosis a mitochondrial disease? Biochimica et biophysica acta. 2010 Jan; 1802(1):66-79.
  6. Inarrea P, Alarcia R, Alava MA, Capablo JL, Casanova A, Iniguez C, et al. Mitochondrial complex enzyme activities and cytochrome C expression changes in multiple sclerosis. Mol Neurobiol. 2014 Feb; 49(1):1-9.
  7. Schaller A, Hahn D, Jackson CB, Kern I, Chardot C, Belli DC, et al. Molecular and biochemical characterization of a novel mutation in POLG associated with Alpers syndrome. BMC Neurology. 2011; 11(1):4.
  8. Milone M, Brunetti-Pierri N, Tang LY, Kumar N, Mezei MM, Josephs K, et al. Sensory ataxic neuropathy with ophthalmoparesis caused by POLG mutations. Neuromuscul Disord. 2008 Aug; 18(8):626-32.
  9. Azrak S, Ayyasamy V, Zirpoli G, Ambrosone C, Bandera EV, Bovbjerg DH, et al. CAG repeat variants in the POLG1 gene encoding mtDNA polymerase-gamma and risk of breast cancer in African-American women. PLoS One. 2012; 7(1):e29548.
  10. Eerola J, Luoma PT, Peuralinna T, Scholz S, Paisan-Ruiz C, Suomalainen A, et al. POLG1 polyglutamine tract variants associated with Parkinson’s disease. Neurosci Lett. 2010 Jun 14; 477(1):1-5.
  11. Rovio A, Abel J, Ahola A, Andres A, Bertranpetit J, Blancher A, et al. A prevalent POLG CAG microsatellite length allele in humans and African great apes. Mammalian genome. 2004; 15(6):492-502.
  12. Spelbrink JN, Toivonen JM, Hakkaart GA, Kurkela JM, Cooper HM, Lehtinen SK, et al. In vivo functional analysis of the human mitochondrial DNA polymerase POLG expressed in cultured human cells. Journal of Biological Chemistry. 2000; 275(32):24818-28.
  13. Williams AJ, Paulson HL. Polyglutamine neurodegeneration: protein misfolding revisited. Trends in neurosciences. 2008; 31(10):521-8.
  14. Luoma P, Eerola J, Ahola S, Hakonen A, Hellström O, Kivistö K, et al. Mitochondrial DNA polymerase gamma variants in idiopathic sporadic Parkinson disease. Neurology. 2007; 69(11):1152-9.
  15. Heidari MM, Houshmand M, Hosseinkhani S, Nafissi S, Scheiber-Mojdehkar B, Khatami M. Association between trinucleotide CAG repeats of the DNA polymerase gene (POLG) with age of onset of Iranian Friedreich’s ataxia patients. Neurol Sci. 2008 Dec; 29(6):489-93.
  16. Heidari MM, Khatami M, Talebi AR. The POLG Gene Polymorphism in Iranian Varicocele-Associated Infertility Patients. Iran J Basic Med Sci. 2012 Mar; 15(2):739-44.
  17. Jensen M, Leffers H, Petersen JH, Nyboe Andersen A, Jorgensen N, Carlsen E, et al. Frequent polymorphism of the mitochondrial DNA polymerase gamma gene (POLG) in patients with normal spermiograms and unexplained subfertility. Hum Reprod. 2004 Jan; 19(1):65-70.
  18. Taanman JW, Schapira AH. Analysis of the trinucleotide CAG repeat from the DNA polymerase gamma gene (POLG) in patients with Parkinson’s disease. Neurosci Lett. 2005 Mar 7; 376(1):56-9.
  19. Wong LJ, Naviaux RK, Brunetti-Pierri N, Zhang Q, Schmitt ES, Truong C, et al. Molecular and clinical genetics of mitochondrial diseases due to POLG mutations. Hum Mutat. 2008 Sep; 29(9):E150-72.
  20. Kumleh HH, Riazi GH, Houshmand M, Sanati MH, Gharagozli K, Shafa M. Complex I deficiency in Persian multiple sclerosis patients. Journal of the Neurological Sciences. 2006 4/15/; 243(1–2):65-9.
  21. Ebers GC, Sadovnick AD, Dyment DA, Yee IML, Willer CJ, Risch N. Parent-of-origin effect in multiple sclerosis: observations in half-siblings. The Lancet. 2004; 363(9423):1773-4.
  22. Harding A, Sweeney M, Miller D, Mumford C, Kellar-Wood H, Menard D, et al. Occurrence of a multiple sclerosis-like illness in women who have a Leber’s hereditary optic neuropathy mitochondrial DNA mutation. Brain: a journal of neurology.1992; 115(4):979-89.
  23. Ahari SE, Houshmand M, Panahi MS, Kasraie S, Moin M, Bahar MA. Investigation on mitochondrial tRNA (Leu/Lys), NDI and ATPase 6/8 in Iranian multiple sclerosis patients. Cell Mol Neurobiol. 2007 Sep; 27(6):695-700.
  24. Mahad DJ, Ziabreva I, Campbell G, Lax N, White K, Hanson PS, et al. Mitochondrial changes within axons in multiple sclerosis. Brain: a journal of neurology. 2009 May; 132(Pt 5):1161-74.

Exon Deletion Pattern in Duchene Muscular Dystrophy in North West of Iran

Mohammad BARZEGAR, Parinaz HABIBI, Mortaza Mortaza BONYADY, Vahideh TOPCHIZADEH, Shadi SHIVA*

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 42-48
https://doi.org/10.22037/ijcn.v9i1.6224

How to Cite This Article: Barzegar M, Habibi P, Bonyady M, Topchizadeh V, Shiva Sh. Exon Deletion Pattern in Duchene Muscular Dystrophy in North West of Iran. Iran J Child Neurol. 2015 Winter; 9(1): 42-48.

Abstract
Objective
Duchene and Becker Muscular Dystrophy (DMD/ BMD) are x-linked disorders that both are the result of heterogeneous mutations in the dystrophin gene. The frequency and distribution of dystrophin gene deletions in DMD/ BMD patients show different patterns among different populations. This study investigates the deletion rate, type, and distribution of this gene in the Azeri Turk population of North West Iran.

Materials &Methods
In this study, 110 patients with DMD/ BMD were studied for intragenic deletions in 24 exons and promoter regions of dystrophin genes by using multiplex PCR.

Results
Deletions were detected in 63 (57.3%) patients, and around 83% localized in the mid-distal hotspot of the gene (on exons 44–52), 21 cases (33.3 %) with singleexon deletions, and 42 cases (66.6%) with multi-exonic deletions. The most frequent deleted exons were exon 50 (15 %) and exon 49 (14%). No deletion was detected in exon 3.

Conclusion
This study suggests that the frequency and pattern of dystrophin gene deletions in DMD/ BMD in the Azeri Turk population of North West Iran occur in the same pattern when compared with other ethnic groups.

References

  1. Emery AE. Clinical and molecular studies in Duchenne muscular dystrophy. Prog Clin Biol Res 1989; 306:15-28.
  2. Moser H. Duchenne muscular dystrophy: pathogenic aspects and genetic prevention. Hum Genet 1984; 66(1):17-40.
  3. Emery AE. Population Frequencies of inherited neuromuscular diseases: a world survey Neuromuscul Disord 1991; I (I):19-29.
  4. Bushby KM, Thmabyayah M, Gardner M D. Prevalence and incidence of Becker muscular dystrophy. Lancet 1991; 337(8748):1022-1024.
  5. Koenig M, Hoffman EP, Bertelosn CJ, Monaco AP, Feener C, Kunkel LM. Complete cloning of the Duchenne muscular dystrophy (DMD) DNA and preliminary genomic organization of the DMD gene in normal and affected individuals. Cell 1987; 50:509-517.
  6. Den Dunnen JT, Grootscholten PM, Bakker E, Blonden LA, Ginjaa rHB, Wapenaar MC, Van Paasen HM, Van Broeckhoven C, Pearson PL, Van Ommen GJ. Topography of the Duchenne muscular dystrophy (DMD) gen. Am J Hum Gennet 1989; 45(6):835-847.
  7. Monaco AP, Bertlson CJ, Liechti-Gallati S, Moser H, Kunkel L.M. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus.Genomics1988; 2:90-95.
  8. Forrest SM, Cross GS, Flint T, Speer A, Robson KJ, Davies KE. Further studies of gene deletions that cause Duchenne and Becker muscular dystrophies. Genomics 1988; 2(2):109-114.
  9. Hux Y, Ray PN, Murphy EG, Thompson MV, Worton RG. Duplicational mutation at the Duchenne muscular dystrophy locus: its frequency, distribution, origin, and phenotype-genotype correlation. Am J Hum Genet 1990; 46:682-695.
  10. Roberts RG, Bobrow M, Bentley DR. Point mutations in the dystrophin gene. Proc Nat Acad Sci USA1992; 89(6):2331-2335.
  11. Mukherjee M, Chaturvedi LS, Srivastava S, Mittal RD, Mital B. Denovo mutations in sporadic deletional Duchenne muscular dystrophy (DMD) cases. Exp Mol Med 2003; 35(2):113-117.
  12. Brooke MG, Griggs RC, Mendell GR, Fenichel GM, Shumate JB, Pellegrino RJ. Clinical Trial in Duchenne Dystrophy. Design of the protocol. Muscle& Nerve 1981; 4: 186-197.
  13. Beggs AH. Multiplex PCR for identifying dystrophin gene deletions In: Dracopoli NC, Haines JL, Korf BR, Moir DT, Morton CC, Seidman CE, et al. Current protocol in human genetics. 1st ed. New York, John Wiley & Sons 2000;unit 9.3
  14. Prior TW, Bridgeman SJ. Experience and strategy for the molecular testing of Duchenne muscular dystrophy, J Mol Diagn 2005; 7(3):317-26.
  15. Chamberlain JS, Gibbs RA, Ranier JE, Nguyen PN, Caskey CT. Deletion screening of the Duchenne Muscular Dystrophy locus via multiplex DNA amplification. Nucleic Acids Res1988;16(23):11141-56.
  16. Beggs AH, Kunkel LM. Improved diagnosis of Duchenne/ Becker muscular dystrophy. J Clin Invest1990; 85:613-619.
  17. Lai Kent KS ,Ivan FM, Tony MF, Lydia YL et al. Detecting exon deletions and duplications of DMD gene using Multiple Ligation dependent Probe Amplification (MLPA).Clinical Biochemistry (2006);39:367-372.
  18. Haider MZ, Bastaki L, Habib Y, Moosa A. Screening 25 dystrophin gene exons for deletions in Arab children with duchenne muscular dystrophy. Hum Hered. 1998; 48; 61- 66.
  19. Hassan MJ, Mahmood S, Ali G, Bibi Nm, Washeed I, Rafiq MA, Ansar M, Ahmad W, et al. Intragenic deletions in the dystrophin gene in 211 Pakistani Duchenne muscular dystrophy patients. Pediatr Int 2008; 50: 162- 166.
  20. Sbiti A, El Kerch F, Sefiani A. Analysis of dystrophin gene deletions by multiplex PCR in Moroccan patients. J Biomed biotechnol 2002; 2:158-160.
  21. Akbari M et al. Molecular diagnosis of Duchenne/Becker Muscular Dystrophy: Analysis of exon deletion and carrier detection. Yakhte Medical Journal 2010; 2(3):421- 428.
  22. Galehdari H, Pedram M, Momen AA, Mohammadian GH R, Taherian E. Study of exon deletion in the dystrophin gene in individuals being diagnosed with Duchenne Muscular Dystrophy in Ahvaz. Sci Med J 2011; 10(4):373-382.
  23. Jabbarpourbonyadi M, Barzgar M, Ayremlo H, Khandagi R, Esmaiili M. Screening and genetic diagnosis of Duchenne-Becker muscular dystrophy in East Azerbaijan by multiplex-PCR. Medical Journal of Tabriz University of Medical Sciences and health services2006; 28(1):33- 39.
  24. Ulgenalp A, Giray O, Bora E, Hizli T, Kurul S, Sagin- Saylam G, Karasoy H, Uran N, Dizdarer G, Tutuncuoglu S, Drink E, Ozkinay F, Ercal D. Deletion analysis and clinical correlations in patients with Xp21 linked muscular dystrophy, The Turkish journal of pediatrics 2004;46:333-338.
  25. Onengut S, Kavaslar GN, Battaloglu E, Serdaroglu P, Deymeer F, Ozdemir F, Ozdemir C. Calafell F, Tolun A. Deletion pattern in the dystrophin gene in Turks and a comparison with Europeans and Indians. Ann Hum Genet 2000; 64:33-40.
  26. Baumbach J, Chamberlain JS, Ward PA, Farwell NJ, Caskey CT. Molecular and clinical correlations of deletions leading to Duchenne and Becker muscular dystrophies. Neurology 1989; 39(4):465-474.
  27. Koenig M1, Beggs AH, Moyer M, Scherpf S, Heindrich K, Bettecken T, Meng G, Müller CR, Lindlöf M, Kaariainen H, et al. The molecular basis for Duchenne versus Becker muscular dystrophy: correlation of severity with type of deletion. Am J Hum Genet 1989; 45(4):498-506.
  28. Helderman-van den E, Straathof CSM, Aartsma A, Dendunnen JT, Verbist BM, Bakker E, Verschuuren JJJM, Ginjaar HB. Becker muscular dystrophy patients with deletions around exon 51; a promising outlook for exon skipping therapy in Duchenne patients. Neuromuscular disorders 2010; 20:251-254.   
  29. Artsma-Rus A, Fokkema I, Verschuuren J, Ginjaar L, Deutekom GV, Ommen GJV et al. Theoretic applicability of antisense-mediated exon skipping for duchenne muscular dystrophy mutations. hummutat 2009; 30:293-9.
  30. Van Deutekom JC, Janson AA, Ginjaar LB, Frankhuizen WS, Artsma-Rus A, Bremmer-Bout Mattie et al. Local dystrophin restoration with antisense oligonucleotide PRO051. N Engl Med 2007; 357:2677-86.

Comparative Efficacy of Zonisamide and Pregabalin as an Adjunctive Therapy in Children with Refractory Epilepsy

Mohammad Mahdi TAGHDIRI, Mohammad Kazem BAKHSHANDEH BALI, Parvaneh KARIMZADEH*, Seyed Hassan TONEKABONI, Mohammad GHOFRANI

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 49-55
https://doi.org/10.22037/ijcn.v9i1.6054

How to Cite This Article: Taghdiri MM, Bakhshandeh Bali MK, Karimzadeh P, Ashrafi MR, Tonekaboni SH, Ghofrani M. Comparative Efficacy of Zonisamide and Pregabalin as an Adjunctive Therapy in Children with Refractory Epilepsy. Iran J Child Neurol. 2015 Winter;9(1):49-55.

Abstract
Objective
Approximately one third of epileptic children are resistant to anticonvulsant drugs. This study evaluates the effectiveness, safety, and tolerability of pregabalin as adjunctive therapy in epileptic children relative to Zonisamide.

Materials & Methods
From April 2012 to November 2012,121 children were referred to Mofid
Children’s Hospital with intractable epilepsy and enrolled in the study. The patients were divided into two groups (A and B) randomly. Group A was treated with Zonisamide and group B was treated with Pregabalin in addition to prior medication. We assessed seizure frequency and severity during a 4-week interval from the beginning of the drug treatment and compared the efficacy of each in these two groups.

Results
Group A consists of 61 patients, 26 (42.6%) girls, and35 (57.4%) boys with an age range from 1.5 months–14 years (mean, 73.9± 44.04 months). Group B consists of 60 patients, 31(51.7%) girls, 29 (48.3%) boys with an age range from 6 months–16 years (mean, 71±42.9 months). Age, gender, seizure onset, seizure frequency, seizure type, and previous antiepileptic medications showed that there was no significant difference between the groups (P>0.05). Zonisamide and pregabalin reduced more than 50% of seizure intensity in 40.2%; 45.8% of patients also had a seizure frequency decline between35.8–44.4%, respectively and there was no significant superiority between these two novel anticonvulsants (P>0.05).

Conclusion
In this survey both pregabalin and Zonisamide were impressive for seizure control in children with intractable epilepsy and well sustained with mild complications that were completely reversible.

 

Frequency, Causes, and Findings of Brain CT Scans of Neonatal Seizure at Besat Hospital, Hamadan, Iran

Fateme EGHBALIAN*, Bahman RASULI, Farnaz MONSEF

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 56-63
https://doi.org/10.22037/ijcn.v9i1.5418

How to Cite This Article: Eghbalian F, Rasuli B, Monsef F. Frequency, Causes, and Findings of Brain CT Scans of Neonatal Seizure at Besat Hospital, Hamadan, Iran. Iran J Child Neurol. 2015 Winter;9(1):56-63.

Abstract

Objective

Neonatal seizures are the most common neurological symptoms and often signal an underlying serious neurologic condition. This study determines the frequency of neonatal seizure, predisposing factors, and brain computed tomography (CT) scan findings.

Materials & Methods

In a descriptive cross-sectional study, we evaluated all neonates with seizures who had been hospitalized in Besat hospital from 2007–2012. All data were gathered with questionnaires and used to compare with statistical tests by SPSS (ver 16).

 

Results

141 (4.08%) neonates (M:F; 1:2.2) were diagnosed with neonatal seizures. From the total number of 3,452 neonatal hospitalization, 78% of neonates with seizures were less than 10 days old and 60.3% of infants were born from natural vaginal delivery. As the most common cause, hypoxic-ischemic encephalopathy in this study was associated with 31.3% (n=44) of neonatal seizures and with the highest mortality rate (n=6). Among admitted neonates with seizures, the overall mortality rate was 12.8% (18 cases). A total of 33.3% of patients (47 cases) had abnormal CT scan reports and 24.8% (35 cases) of patients were not evaluated with a CT scan. Hypoxic-ischemic encephalopathy (47%) and local ischemic changes (25.5%) were the most common findings in the CT scans of neonates with seizures.

Conclusion

There was a significant correlation between neonatal seizures and delivery circumstances (p-value < 0.05). Therefore, with improvement of obstetric and delivery circumstances, early detection of predisposing factors and other rare conditions, and rapid effective treatment of these contributing factors, the rate of neonatal seizure in this period can be reduced.

References

  1. Scher MS. Seizure in the newborn infant: diagnosis, treatment, and outcome. Clin perinatal 2005; 24: 735-772.
  2. Lanska MJ, Lanska DJ, Baumann RJ, Kryscio RJ. A population-based study of neonatal seizures in Fayette County, Kentucky. Neurology 1995; 45:724-32.
  3. Ronen GM, Penney S, Andrews W. The epidemiology of clinical neonatal seizures in Newfoundland: a populationbased study. J Pediatr 1999; 134:71-5.
  4. Saliba RM, Annegers JF, Waller DK, et al. Incidence of neonatal seizures in Harris County, Texas, 1992-1994. Am J Epidemiol 1999; 150:763-9.
  5. Volpe JJ. Neonatal seizures. In: Neurology of the newborn. 5th ed. Philadelphia: WB Saunders. 2008. P. 203-44.
  6. Fanaroff AA, Martin RJ, Neonatal. Prenatal medicine. 8th ed. New York: Mosby, 2006; 956-976.
  7. Scher MS, Aso K, Beggarly ME, et al. Electrographic seizures in preterm and full-term neonates: clinical correlates, associated brain lesions, and risk for neurologic sequelae. Pediatrics 1993; 91:128-34.
  8. Tekgul H, Gauvreau K, Soul J, et al. The current etiologic profile and neurodevelopmental outcome of seizures in term newborn infants. Pediatrics 2006; 117:1270-80.
  9. Rogalski LI, Minokoshi M, Silveira DC, Cha BH, Holmes GL. Recurrent neonatal seizures: relationship of pathology to the electroencephalogram and cognition. Brain Res Dev Brain Res 2001; 129:27-38.
  10. Cilio MR, Sogawa Y, Cha BH, Liu X, Huang LT, Holmes GL. Long-term effects of status epilepticus in the immature brain are specific for age and model. Epilepsia 2003; 44:518-28.
  11. Lynch M, Sayin U, Bownds J, Janumpalli S, Sutula T. Long-term consequences of early postnatal seizures on hippocampal learning and plasticity. Eur J Neurosci 2000; 2:2252-64.
  12. Ni H, Jiang YW, Bo T, Wang JM, Wu XR. c-Fos, N-methyl-Diaspartate receptor 2C, GABA-A-a1 immunoreactivity, seizure latency, and neuronal injury following single or recurrent neonatal seizures in hippocampus of Wistar rat. Neurosci Lett 2005; 380:149-54.
  13. Bo T, Jiang Y, Cao H, Wang J, Wu X. Long-term effects of seizures in neonatal rats on spatial learning ability and N-methyl-D-aspartate receptor expression in the brain. Brain Res Dev Brain Res 2004; 152:137-42.
  14. Cornejo BJ, Mesches MH, Coultrap S, Browning MD, Benke TA. A single episode of neonatal seizures permanently alters glutamatergic synapses. Ann Neurol 2007; 61:411-26.
  15. Sanchez RM, Dai W, Levada RE, Lippman JJ, Jensen FE. AMPA/kainate receptor-mediated downregulation of GABAergic synaptic transmission by calcineurin after seizures in the developing rat brain. J Neurosci 2005; 25:3442-51.
  16. Huang L, Cilio MR, Silveira DC, et al. Long-term effects of neonatal seizures: a behavioral, electrophysiological, and histological study. Brain Res Dev Brain Res 1999; 118:99-107.
  17. Sayin U, Sutula TP, Stafstrom CE. Seizures in the developing brain cause adverse long-term effects on spatial learning and anxiety. Epilepsia 2004; 45:1539-48.
  18. Fanaroff AA, Martin RJ. Neonatal prenatal medicine. 7th ed. New York: Mosby. 2002.P.887-899.
  19. Bluvstein E, Judith B, Moshe B, Soloman N. Current management in child neurology. 2nd ed. Lernard Maria. 2002. P.123-128.
  20. Scafer B, Bodensteiner JB. Radiological findings in development delay. Semin Pediatr Neurol 1998 Mar: 37-8.
  21. Barkovick AJ. Pediatric neuroimaging. 3rd ed. New York: Lippincott, Williams & Wilkins, 2000: 162-205.
  22. Volpe A. Neurology of the newborn. 4th ed. Philadelphia: W.B Saunders. 2000. P.172-202.
  23. Swaiman KF, Ashwal S. Pediatric neurology. Principles and practice. 3rd ed. New York: Mosby. 1999.P. 183-188.
  24. Haaga JR, Lanzieri CF. Computed tomography and magnetic resonance imaging of the whole body. Third ed. New York: Mosby, 1994.
  25. Girard N, Raybaud C. Neonates with Seizures: What to Consider, How to Image. Magnetic Resonance Imaging Clinics of North America 2011 Nov;19(4); 685-708.
  26. Christopher P. Hess, A. James Barkovich. Seizures: Emergency Neuroimaging. Neuroimaging Clinics of North America 2010 Nov;20(4):619-637.
  27. Memon S, Memon MH. Spectrum and immediate outcome of seizures in neonates. JCPSP 2006; 16(11):717-20.
  28. Denizmen A, Filizy Y. Our cases of seizures in neonatal unit. J Pediatr 1997; 6(1):33-6.
  29. Park W, Kim DY, Jung CZ. Clinical study of neonatal seizure. J Korean Child Neurol Soc 1998; 6(1):71-82.
  30. Dehdashtian M, Momen AA, Ziaei T, Moradkhani S. Evaluation of seizure etiology in convulsive neonates admitted to Imam Khomeini and Abozar hospitals of Ahvaz 2004-2007. Jundishapur Scientific Medial Journal 2009;8(2): 163.
  31. Mirza RM, Posti A. Evaluation of causes of neonatal seizure in Ali Asghar and Alavi hospital in Ardabil city. Scientific journal of Ardabil university of medical sciences 2000:50-55.
  32. Kohelet D, Shoehart R, Lusky A, et al. Israel Neonatal Network, risk factors for neonatal seizures in very low birth weight infants: population-based study. J Pediatr 2005sep;16(8):245-251.
  33. Kumar A, Gupta A, Talukdar B. Clinico-Etiological and EEG profile of neonatal seizures. Ind J Pediatr. 2007; 74(1):33-7.
  34. Joseph VJ. Neonatal seizure. In: Joseph VJ. Neurology of the newborn. 5th ed. Philadelphia: WB Saunders. 2008.P.216- 223.
  35. Calciolari G, Perlman JM, Volpe JJ. Seizures in the neonatal intensive care unit of the 1989s. Types, Etiologies, Timing. Clin Pediatr (Phila)  2000 Mar; 27(3): 119-23.
  36. Mwaniki M, Mathenge A, Gwer S, et al. Neonatal seizure in a rural Kenyan District 1 hospital: aetiology, incidence and outcome of hospitalization. BMC medicine 2010; 8(16):18-25.
  37. Scher MS, Painter MJ, Bergman I, et al. EEG diagnosis of neonatal seizure: clinical correlation and outcome. Pediatr Neurol 1986; 5:17-24.
  38. Gabremariam A, Gutema Y, Leurl A, et al. Early-onset neonatal seizure: type, risk factors, and short outcome. Ann Trop Pediatr  2006; 26(2); 127-31.
  39. Taghdiri MM, Emdadi M, Eghbalian F, Tavasoli AR. Radioimaging in Convulsion Neonates Based on Brain CT Without Contrast. Scientific journal of Hamadan university of medical sciences 2005:50-55.
  40. Ronen GM, Penney S, Andrews W. The epidemiology of clinical neonatal seizures in Newfoundland: a population based study. Epilepsia 1995; 36(suppl 4):28.
  41. Omenea JA, Longeb AC, and Okoloa AA. seizures in the Nigerian neonate: perinatal factors. Int J Gynaecol Obstet 1981;19: 295-299.
  42. John RH, Charles FL. Computed tomography and magnetic resonance imaging the whole body. 3rd ed. New York: Mosby. 1994.P. 163-165.
  43. Stevenson KD, Sunshine P. The management of neonatal seizures. Semin Pediatr Neurol 1999; 27: 72-82.
  44. Koren G, Warwicke B, Rajchgot R, Donn S. Intravenous paraldehyde for seizure control in newborn. Ann Neurol 2002; 36: 108-110.
  45. Painter M, Pippenger G, Wasterlain C. Phenobarbital and Phenytoin in neonatal seizure. J Pediatr 2001; 91: 153-160.
  46. Rennie J, Boylan G. Neonatal seizure and their treatment. Vol 16. New York: Lippincott Williams & Wilkins. 2003.P.177-181.

Comparative Study of Sustained Attentional Bias on Emotional Processing in ADHD Children to Pictures with Eye-Tracking

Ebrahim PISHYAREH, Mehdi EHRANI-DOOST, Javad MAHMOODI-GHARAIE, Anahita KHORRAMI, Saeid Reza RAHMDAR

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 64-70
https://doi.org/10.22037/ijcn.v9i1.5589

How to Cite This Article: Pishyareh E, Tehrani-doost M, Mahmoodi-gharaie J, Khorrami A, Rahmdar SR. A Comparative Study of Sustained
Attentional Bias on Emotional Processing in ADHD Children to Pictures with Eye-Tracking. Iran J Child Neurol. 2015 Winter;9(1):64-70.

Abstract

Objective

ADHD children have anomalous and negative behavior especially in emotionally related fields when compared to other. Evidence indicates that attention has an impact on emotional processing. The present study evaluates the effect of emotional processing on the sustained attention of children with ADHD type C.

Materials & Methods

Sixty participants form two equal groups (each with 30 children) of normal and ADHD children) and each subject met the required selected criterion as either a normal or an ADHD child. Both groups were aged from 6–11-years-old. All pictures were chosen from the International Affective Picture System (IAPS) and presented paired emotional and neutral scenes in the following categories: pleasant-neutral; pleasant-unpleasant; unpleasant-neutral; and neutral–neutral. Sustained attention was evaluated based on the number and duration of total fixation and was compared between the groups with MANOVA analysis.

Results

The duration of sustained attention on pleasant in the pleasant-unpleasant pair was significant. Bias in duration of sustained attention on pleasant scenes in pleasant-neutral pairs is significantly different between the groups.

Conclusion

Such significant differences might be indicative of ADHD children deficiencies in emotional processing. It seems that the highly deep effect of emotionally unpleasant scenes to gain the focus of ADHD children’s attention is responsible for impulsiveness and abnormal processing of emotional stimuli.

 

References

1. Sadock, B.J., H.I. Kaplan, and V.A. Sadock, Kaplan & Sadcok’s Synopsis of Psychiatry: Behavioral Sciences, Clinical Psychiatry. Lippincott; Williams & Wilkins. 2003.

2. Cormier, E., Attention deficit/hyperactivity disorder: a review and update. Journal of Pediatric Nursing, 2008. 23(5): p. 345-357.

3. Casey, R.J., Emotional competence in children with externalizing and internalizing disorders. Emotional development in atypical children 1996: p. 161-183.

4. Cadesky, E.B., V.L. Mota, and R.J. Schachar, Beyond words: How do children with ADHD and/or conduct problems process nonverbal information about affect? Journal of the American Academy of Child & Adolescent Psychiatry 2000. 39(9): p. 1160-1167.

5. Norbury, C., et al., Eye-movement patterns are associated with communicative competence in autistic spectrum disorders. Journal of Child Psychology and Psychiatry 2009. 50(7): p. 834-842.

6. Vike E. The Perception of Emotional Expressions in Incarcerated Youth 2002.

7. Oades, R., Frontal, temporal and lateralized brain function in children with attention-deficit hyperactivity disorder: a psychophysiological and neuropsychological viewpoint on development. Behavioural Brain Research 1998; 94(1): p. 83-95.

8. Singh SD, et al. Recognition of facial expressions of emotion by children with attention-deficit hyperactivity disorder. Behavior Modification 1998. 22(2): p. 128-142.

9. Yuill N, Lyon J. Selective difficulty in recognizing facial expressions of emotion in boys with ADHD. European child & adolescent psychiatry  2007;16(6):398-404.

10. LeDoux J. Emotion circuits in the brain. Annual review of neuroscience 2000;12(1):155-184.

11. Da Fonseca, D., et al., Emotion understanding in children with ADHD. Child Psychiatry & Human Development 2009; 40(1):111-121.

12. Guevremont DC, Dumas MD. Peer relationship problems and disruptive behavior disorders. Journal of Emotional and Behavioral Disorders 1994; 2(3):164.

13. Tucha L, et al. Vigilance and sustained attention in children and adults with ADHD. Journal of Attention Disorders 2009;12(5): 410.

14. Barry, T.D., Robert D. Lyman, and L.G. Klinger, Academic Underachievement and Attention-Deficit/Hyperactivity Disorder: The Negative Impact of Symptom Severity on School Performance. Journal of School Psychology 2002;40(3):259-283.

15. Douglas VI. Stop, look and listen: The problem of sustained attention and impulse control in hyperactive and normal children. Canadian Journal of Behavioural Science/Revue canadienne des sciences du comportement. 1972; 4(4):259.

16. Lang P, Bradley M, Cuthbert B. International affective picture system (IAPS): Technical manual and affective ratings. NIMH Center for the Study of Emotion and Attention, 1997.

17. Watts SE, Weems . CF. Associations among selective attention, memory bias, cognitive errors and symptoms of anxiety in youth. Journal of abnormal child psychology 2006; 34(6):838-849.

18. Nummenmaa L, Hyo¨na J, Calvo MG. Eye Movement Assessment of Selective Attentional Capture by Emotional Pictures. Emotion 2006; 6(2):257-268.

19. Kiss M, et al. Efficient attentional selection predicts distractor devaluation: event-related potential evidence for a direct link between attention and emotion. J Cogn Neurosci 2007. 19(8): p. 1316-22.

20. Kinsbourne M, Bemporad B. Lateralization of emotion: A model and the evidence. The psychobiology of affective development, 1984: p. 259-291.

21. Da Fonseca D, et al. Emotion understanding in children with ADHD. Child psychiatry and human development 2009; 40(1): 111-121.

22. Calvo MG, Lang PJ. Gaze patterns when looking at emotional pictures: Motivationally biased attention . Motivation and Emotion 2004; 28:221-243.

23. Armony JL, Dolan RJ. Modulation of spatial attention by fear-conditioned stimuli: an event-related fMRI study. Neuropsychologia 2002; 40(7): p. 817-826.

24. Kanske, P., Exploring Executive Attention in Emotion: ERP and FMRI Evidence2008: Max Planck Inst. For Human Cognitive and Brain Sciences.

25. Whalen CK, Henker B. Attention-deficit/hyperactivity disorders. Handbook of child psychopathology 1998; 3: 181-211.

26. Shaywitz BA, Shaywitz SE. Comorbidity: A critical issue in attention deficit disorder. Journal of Child Neurology 1991; 6(1 suppl): S13.

27. Pennington BF,Ozonoff S. Executive functions and developmental psychopathology. Journal of Child Psychology and Psychiatry 1996. 37(1): p. 51-87.

28. Vike E. The Perception of Emotional Expressions in Incarcerated Youth 2001.

29. Deans P, et al. Use of Eye Movement Tracking in the Differential Diagnosis of Attention Deficit Hyperactivity Disorder (ADHD) and Reading Disability Psychology 2010; 1: 238-246.

31. Caseras X, et al., Biases in visual orienting to negative and positive scenes in dysphoria: An eye movement study. Journal of abnormal psychology 2007; 116(3): 491.

32. Eizenman M, et al. A naturalistic visual scanning approach to assess selective attention in major depressive disorder. Psychiatry research 2003; 118(2):117-128.

33. Kellough J, et al. Time course of selective attention in clinically depressed young adults: An eye tracking study. Behaviour Research and Therapy 2008; 46(11): 1238-1243.

34. Krauel K, et al. Emotional memory in ADHD patients with and without comorbid ODD/CD. Journal of Neural Transmission 2009; 116(1):117-120.

Causes and Associated Factors of Headaches among 5 to 15-year-old Children Referred to a Neurology Clinic in Kashan, Iran

Ahmad TALEBIAN, Babak SOLTANI*, Mostafa HAJI REZAEI

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 71-75
https://doi.org/10.22037/ijcn.v9i1.5679

How to Cite This Article: Talebian A, Soltani B, Haji Rezaei M. Causes and Associated Factors of Headaches among 5 to 15-year-old Children
Referred to a Neurology Clinic in Kashan, Iran. Iran J Child Neurol. 2015 Winter;9(1):71-75.

Abstract

Objective

Headaches are common neurologic problems for children and adolescents. They are divided into two types: primary and secondary. Primary headaches include migraines and tension-type as well as comprise the majority of headaches. We detect the causes of headaches and their associations with demographic variables among children and adolescents.

Materials & Methods

This cross-sectional study was performed on 5–15 year-old children with headaches from March 2010 to April 2012 who presented at a pediatric neurology clinic in Kashan, Iran. Diagnosis of headaches was done in accordance with the International Classification of Headache Disorders. Data regarding the type of headache, age, gender, pain severity, aura, family history, and sleep disorder were collected.

Results

One hundred fourteen children (44 male and 70 female) with headaches were enrolled in the study. The types of headaches were comprised as follows: 67 cases of migraines, 38 cases of tension-type headaches, 2 cases of cluster headaches, and 7 cases of secondary headaches. Pulsating headaches, family history of headaches, insomnia, and pain severity had higher prevalence in migrainous patients.

Conclusion

Physicians should extend their information gathering about primary and secondary headaches. Sleep disturbances and a family history of headaches were the most important factors associated with migraine headaches.

References

  1. Cuvellier JC, Donnet A, Guegan-Massardier E, Nachit-Ouinekh F, Parain D, Vallee L. Treatment of primary headache in children: a multicenter hospital-based study in France. J Headache Pain 2009; 10: 447-53.
  2. Lateef TM, Merikangas KR, He J, Kalaydjian A, Khoromi S, Knight E, et al. Headache in a national sample of American children: prevalence and comorbidity. J Child Neurol 2009; 24: 536-43.
  3. Zwart JA, Dyb G, Holmen TL, Stovner LJ, Sand T. The prevalence of migraine and tension-type headaches among adolescents in Norway. The Nord-Trondelag Health Study (Head-HUNT-Youth), a large populationbased epidemiological study. Cephalalgia 2004; 24: 373- 9.
  4. Isik U, Topuzoglu A, Ay P, Ersu RH, Arman AR, Onsuz MF, et al. The prevalence of headache and its association with socioeconomic status among schoolchildren in istanbul, Turkey. Headache 2009; 49: 697-703.
  5. Abu-Arafeh I, Macleod S. Serious neurological disorders in children with chronic headache. Arch Dis Child 2005; 90: 937-40.
  6. Lewis DW. Headaches in children and adolescents. Am Fam Physician 2002; 65: 625-32.
  7. The International Classification of Headache Disorders: 2nd edition. Cephalalgia 2004; 24 Suppl 1: 9-160.
  8. Tavasoli A, Aghamohammadpoor M, Taghibeigi M. Migraine and Tension-Type Headache in Children and Adolescents Presenting to Neurology Clinics. Iran J Pediatr 2013; 23: 536-540.
  9. Bruni O, Fabrizi P, Ottaviano S, Cortesi F, Giannotti F, Guidetti V. Prevalence of sleep disorders in childhood and adolescence with headache: a case-control study. Cephalalgia 1997; 17: 492-8.
  10. Isik U, Ersu RH, Ay P, Save D, Arman AR, Karakoc F, et al. Prevalence of headache and its association with sleep disorders in children. Pediatr Neurol 2007; 36: 146-51.
  11. Miller VA, Palermo TM, Powers SW, Scher MS, Hershey AD. Migraine headaches and sleep disturbances in children. Headache 2003; 43: 362-8.
  12. Lewis DW, Ashwal S, Dahl G, Dorbad D, Hirtz D, Prensky A, et al. Practice parameter: evaluation of children and adolescents with recurrent headaches: report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology 2002; 59: 490-8.
  13. Lewis DW,Koch T. Headache evaluation in children and adolescents: when to worry? When to scan? Pediatr Ann 2010; 39: 399-406.
  14. Hershey AD, Powers SW, Bentti AL, Degrauw TJ. Effectiveness of amitriptyline in the prophylactic management of childhood headaches. Headache 2000; 40: 539-49.
  15. Donald W. Headache in children and adolescent. Am Fam Physician 2002; 65: 625-33.
  16. Kroner-Herwig B, Gassmann J. Headache disorders in children and adolescents: their association with psychological, behavioral, and socio-environmental factors. Headache 2012; 52: 1387-401.
  17. Wober-Bingol C, Wober C, Wagner-Ennsgraber C, Zebenholzer K, Vesely C, Geldner J, et al. IHS criteria and gender: a study on migraine and tension-type headache in children and adolescents. Cephalalgia 1996; 16: 107-12.
  18. Abu-Arafeh I,Russel G. Prevalence of headache and migraine in schoolchildren. British medical journal 1994; 308: 765-9.
  19. Ayatollahi SM,Khosravi A. Prevalence of migraine and tension-type headache in primary-school children in Shiraz. East Mediterr Health J 2006; 12: 809-17.
  20. Fallahzadeh H,Alihaydari M. Prevalence of migraine and tension-type headache among school children in Yazd,Iran. J Pediatr Neurosci 2011; 6: 106-9.
  21. Russell MB, Iselius L, Ostergaard S, Olesen J. Inheritance of chronic tension-type headache investigated by complex segregation analysis. Hum Genet 1998; 102: 138-40.
  22. Stewart WF, Linet MS, Celentano DD, Van Natta M, Ziegler D. Age and sex specific incidence rates of migraine with and without visual aura. Am J Epidemiol 1991; 134: 1111-20.

Psychometric Properties of the Persian Version of Cerebral Palsy Quality of Life Questionnaire for Children

Farin SOLEIMANI, Roshanak VAMEGHI MD, Anoshirvan KAZEMNEJAD, Nazila AKBAR FAHIMI, Zahra NOBAKHT, Mehdi RASSAFIANI*

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 76-86
https://doi.org/10.22037/ijcn.v9i1.6060

How to Cite This Article: Soleimani F, Vameghi R, Kazemnejad A, Akbar Fahimi N, Nobakht Z, Rassafiani M. Psychometric Properties of the Persian Version of Cerebral Palsy Quality of Life Questionnaire for Children. Iran J Child Neurol. 2015 Winter;9(1):76-86.

 

Abstract

Objective

Cerebral palsy (CP) is the most common cause of chronic disability that restricts participation in daily life for children. Thereby, it is comprised of quality of life. Quality of life (QOL) measures have been a vital part of health outcome appraisals for individuals with CP and to obtain empirical evidence for the effectiveness of a range of interventions. The CP QOL-Child is a condition-specific QOL questionnaire designed for children with CP to assess well-being rather than ill-being.

Materials & Methods

Forward and backward translations of the CP QOL-Child were performed for: (1) the primary caregiver form (for parents of children with CP aged 4–12 years); and (2) the child self-report form (for children with cerebral palsy aged 9–12 years). Psychometric properties assessment included reliability, internal consistency, and item discrimination, construct validity with Gross Motor Function Classification System (GMFCS) and Manual Ability Classification System (MACS) was done. SPSS was used to analyze the results of this study.

Results

A sample of 200 primary caregivers forchildren with CP (mean = 7.7 years) and 40 children (mean = 10.2 years) completed. Internal consistency ranged from 0.61–0.87 for the primary caregivers form, and 0.64–0.86 for the child self-report form. Reliability ranged from 0.47–0.84. Item discrimination analysis revealed that a majority of the items (80%) have high discriminating power. Confirmatory factor analysis demonstrated a distinguishable domain structure as in the original English version. Moderate associations were found between lower QOL and more severe motor disability(GMFCS; r = .18–.32; p < .05 and MACS; r= .13 - .40; p < .05). The highest correlation between the primary caregiver and child forms on QOL was in the domain of functioning and consistent with the English version.

Conclusion

Content validity, item discriminant validity, internal consistency, and test-retest reliability of the Persian version of the CP QOL- Child were all acceptable. Further study of concurrent validity of this version is needed.

 

 

References

  1. Bax M, Goldstein M, Rosenbaum PL, Leviton A, Paneth N, Dan B et al. Executive Committee for the Definition of Cerebral Palsy. Proposed definition and classification of cerebral palsy, April 2005. Dev Med Child Neurol 2005; 47:571–576.
  2. Rosenbaum, P, Paneth, N, Leviton A, Goldstein M, Bax M, DamianoD, et al. A report: The definition and classification of cerebral palsy April 2006. Dev Med Child Neurol 2007, 49, 8–14.
  3. Surveillance of Cerebral Palsy in Europe. Surveillance of cerebral palsy in Europe: A collaboration of cerebral palsy surveys and registers. Dev Med Child Neurol 2000; 42, 816–824.
  4. Liptak GS, O’Donnell M, Conaway M, Chumlea WC, Wolrey G, Henderson RC, et al. Health status of children with moderate to severe cerebral palsy. Dev Med Child Neurol 2001; 43, 364–370.
  5. Maher CA, Olds T, Williams MT, Lane AE.Self-reported quality of life in adolescents with cerebral palsy.Physical & Occupational Therapy in Pediatrics 2008; 28, 41–57.
  6. Pirpiris M, Gates PE, McCarthy JJ, D’Astous J, Tylkowksi C, Sanders JO, et al. Function and well-being in ambulatory children with cerebral palsy. Journal of Pediatric Orthopedics 2006; 26, 119–124.
  7. Vargus-Adams J. Health-related quality of life in childhood cerebral palsy. Archives of Physical Medicine and Rehabilitation 2005; 86, 940–945.
  8. Varni JW, Burwinkle TM, Sherman SA, Hanna K, Berrin SJ, Malcarne VL, et al. Health-related quality of life of children and adolescents with cerebral palsy: Hearing the voices of the children. Dev Med Child Neurol 2005; 47, 592–597.
  9. Bjornson KF, McLaughlin JF.The measurement of health-related quality of life (HRQL) in children with cerebral palsy.European Journal of Neurology 2001; 8(Suppl. 5), 183–193.
  10. Waters E, Maher E, Salmon L, Reddihough D, Boyd R. Development of a condition-specific measure of quality of life for children with cerebral palsy: Empirical thematic data reported by parents and children. Child: Care, Health, and Deve 2005; 31, 127–135.
  11. Waters E, Davis E, Mackinnon A, Boyd R, Graham HK, Kai Lo S, et al. Psychometric properties of the quality of life questionnaire for children with CP. Dev Med Child Neurol 2007; 49, 49–55.
  12. Wang HY, Cheng CC, Hung JW, Ju YH, Lin JH, Lo SK. Validating the Cerebral Palsy Quality of Life for Children (CP QOL-Child) questionnaire for use in Chinese. populations. Neuropsychological Rehabilitation 2010; 20, 883–898.
  13. Mutch L, Alberman E, Hagberg B, Kodama K, PeratMV. Cerebral palsy epidemiology: Where are we now and where are we going? Dev Med Child Neurol 1992; 34: 547–51.
  14. Stanley F, Blair E, Alberman E. (eds) Cerebral Palsies: Epidemiology and Causal Pathways. London: Mac Keith. 2000.
  15. Palisano R, Rosenbaum P, Walter S, Russell D, Wood E, Galuppi B. Development and reliability of a system to classify gross motor function in children with cerebral palsy. Dev. Med. Child Neurol 1997; 39: 214–23.
  16. Wood E, Rosenbaum P. The Gross Motor Function Classification System for cerebral palsy: A study of reliability and stability overtime. Dev Med Child Neurol 2000; 42: 292–6.
  17. Sanger TD, Delgado MR, Gaebler-Spira D, Hallett M, Mink JW. Classification and definition of disorders causing hypertonia in childhood. Pediatrics 2003; 111: e89–97.
  18. Sanger TD. Pathophysiology of pediatric movement disorders.J. Child Neurol 2003; 18: S9–24.
  19. Delgado M, Albright A. Movement disorders in children: Definitions, classifications and grading systems. J Child Neurol 2003; 18: S1–8.
  20. Palisano R, Rosenbaum P, Walter S, Russell D, Wood E, Galuppi B. Development and reliability of a system to classify gross motor function in children with cerebral palsy. Developmental Medicine and Child Neurology 1997; 39:214-223.
  21. Riahi A, Rassafiani M, Binesh M. The Cross-Cultural Validation and Test-Retest and Inter-Rater Reliability of the Persian Translation of Parent Version of the Gross Motor Function Classification System for Children with Cerebral Palsy. J of Rehab 2013; 13(5), 25-30.
  22. Morris C, Galuppi BE, Rosenbaum P. Reliability of family report for the Gross Motor Function Classification System. Developmental Medicine and Child Neurology 2004; 46:455-460.
  23. Riyahi A, Rassafiani M, AkbarFahimi N, Sahaf R, Yazdani F. Cross cultural validation of the Persian version of the Manual Ability Classification System for children with cerebral palsy. International Journal of Therapy and Rehabilitation 2013; 20(1), 19-24.
  24. The International Quality of Life Assessment (IQOLA) Project. http://www.iqola.org.
  25. Stanley F, Blair E, Alberman E. (2000) Cerebral Palsies: Epidemiology and Causal Pathways. Clinics in Developmental Medicine No. 151. London: Mac Keith Press.
  26. World Health Organization (1993). Measuring Quality of Life: The development of the World Health Organization Quality of Life Instrument (WHOQOL).Geneva: Division of Mental Health, World Health Organization.
  27. Davis E, Waters E, Mackinnon A, Reddihough D, Graham HK, Mehmet-Radji O, Boyd R. (2006) Paediatric quality of life instruments: a review of the impact of the conceptual framework on outcomes. Dev Med Child Neurol 48: 311–318.
  28. Schneider JW, Gurucharri LM, Gutierrez AL, et al. Health-related quality of life and functional outcome measures for children with cerebral palsy. Dev Med Child Neurol 2001; 43(9):601-8.
  29. Majnemer A, Shevell M, Hall N, et al. Developmental and functional abilities in children with cerebral palsy as related to pattern and level of motor function. J Child Neurol 2010; 25(10):1236-41.
  30. Davis E, Shelly A, Waters E, Boyd R, Cook K, Davern M, &Reddihough D. The impact of caring for a child with cerebral palsy: Quality of life for mothers and fathers. Child: Care, Health and Dev 2010; 36, 63–73.
  31. Liu WY, Hou YJ, Wong AM, Lin PS, Lin YH, Chen CL. Relationships between gross motor functions and health-related quality of life of Taiwanese children with cerebral palsy. AJPM&R 2009; 88, 473–483.
  32. Eiser C, Morse R. Can parents rate their child’s health related quality of life? Results of a systematic review. Qual Life Res 2001; 10: 347–357.
  33. Massaro M, Pastore S, Ventura A, Barbi E. Pain in cognitively impaired children: a focus for general pediatricians. Eur J Pediatr 2013; 172(1):9-14
  34. Elbasan B, Duzgun I, Oskay D. Is There any Difference in Health Related Quality of Life, Self Care and Social Function in Children with Different Disabilities Living in Turkey? Iran J Pediatr 2013; 23, 281-288.

 

Neural Differentiation of Human Umbilical Cord Mesenchymal Stem Cells by Cerebrospinal Fluid

Shirin FARIVAR*, Zahra MOHAMADZADE, Reza SHIARI, Alireza FAHIMZAD

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 87-93
https://doi.org/10.22037/ijcn.v9i1.4257

How to Cite This Article: : Farivar S, Mohamadzade Z, Shiari R, Fahimzad AR. Neural Differentiation of Human Umbilical Cord
Mesenchymal Stem Cells by Cerebrospinal Fluid. . Iran J Child Neurol. 2015 Winter; 9(1):87-93.

 

 Abstract

Objective

Wharton’s jelly (WJ) is the gelatinous connective tissue from the umbilical cord. It is composed of mesenchymal stem cells, collagen fibers, and proteoglycans. The stem cells in WJ have properties that are interesting for research. For example, they are simple to harvest by noninvasive methods, provide large numbers of cells without risk to the donor, the stem cell population may be expanded in vitro, cryogenically stored, thawed, genetically manipulated, and differentiated in vitro. In our study, we investigated the effect of human cerebrospinal fluid (CSF) on neural differentiation of human WJ stem cells.

Material & Methods

The cells in passage 2 were induced into neural differentiation with different concentrations of human cerebrospinal fluid. Differentiation along with neural lineage was documented by expression of three neural markers: Nestin, Microtubule-Associated Protein 2 (MAP2), and Glial Fibrillary Astrocytic Protein (GFAP) for 21 days. The expression of the identified genes was confirmed by Reverse Transcriptase PCR (RT-PCR).

Results

Treatment with 100 and 200μg/ml CSF resulted in the expression of GFAP and glial cells marker on days 14 and 21. The expression of neural-specific genes following CSF treatment was dose-dependent and time-dependent. Treatment of the cells with a twofold concentration of CSF, led to the expression of MAP2 on day 14 of induction. No expression of GFAP was detected before day 14 or MAP2 before day 21, which shows the importance of the treatment period. In the present study, expression analysis for the known neural markers: Nestin, GFAP, and MAP2 using RT-PCR were performed. The data demonstrated that CSF could play a role as a strong inducer.

Conclusion

RT-PCR showed that cerebrospinal fluid promotes the expression of Nestin, MAP2, and GFAP mRNA in a dose-dependent manner, especially at a concentration of 200 μl/ml. In summary, CSF induces neurogenesis of WJ stem cells that encourages tissue engineering applications with these cells for treatments of neurodegenerative defects and traumatic brain injury.

References

  1. Gage, F. H. Mammalian neural stem cells. Science 2000 Feb 25;287(5457):1433-8.
  2. Da Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all postnatal organs and tissues. J Cell Sci 2006 Jun 1;119(Pt 11):2204- 13. Epub 2006 May 9.
  3. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells Science 1999 Apr 2;284(5411):143-7.
  4. Tse WT, Pendleton JD, Beyer WM, Egalka MC, Guinan EC. Suppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation 2003 Feb 15;75(3):389- 97.
  5. Le Blanc K. Immuno-modulatory effects of fetal and adult mesenchymal stem cells. Cytotherapy 2003;5(6):485-9.
  6. Stenderup K, Justesen J, Clausen C, Kassem M. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone 2003 Dec;33(6):919-26.
  7. Bongso A, Fong CY, Gauthaman K. Taking stem cells to the clinic: major challenges. J Cell Biochem 2008 Dec 15;105(6):1352-60. doi: 10.1002/jcb.21957.
  8. Fong CY, Chak LL, Biswas A. Human Wharton’s jelly stem cells have unique transcriptome profiles compared to human embryonic stem cells and other mesenchymal stem cells. Stem Cell Rev 2011 Mar;7(1):1-16. doi: 10.1007/s12015-010-9166-x.
  9. Troyer DL, Weiss ML. Wharton’s jelly-derived cells are a primitive stromal cell population. Stem Cells 2008 Mar; 26(3):591-9. Epub 2007 Dec 6.
  10. Yuan X, Desiderio DM. Proteomics analysis of human cerebrospinal fluid. J Chromatogr B Analyt Technol Biomed Life Sci 2005 Feb 5;815(1-2):179-89.
  11. Thompson, EJ. Cerebrospinal fluid. J Neurol Neurosurg Psychiatry 1995 Oct;59(4):349-57.
  12. Alcazar A, Regidor I, Masjuan J, Salinas M, Alvarez- Cermeno JC. Induction of apoptosis by cerebrospinal fluid from patients with primary-progressive multiple sclerosis in cultured neurons. Neurosci Lett 1998 Oct 16;255(2):75-8.
  13. Colombo JA, Napp MI. Cerebrospinal fluid from L-dopa-treated Parkinson’s disease patients is dystrophic for various neural cell types exvivo: effects of astroglia. Exp Neurol 1998 Dec;154(2):452-63.
  14. Redzic ZB, Preston JE, Duncan JA, Chodobski A, Szmydynger-Chodobska J. The choroid plexus-cerebrospinal fluid system: from development to aging. Current topics in developmental biology 2005; (71): 1-52.
  15. Bachy I, Kozyraki R, Wassef M. The particles of the embryonic cerebrospinal fluid: how could they influence brain development? Brain Res Bull 2008 Mar 18;75(2- 4):289-94. doi: 10.1016/j.brainresbull.2007.10.010.
  16. Parada C, Gato A, Bueno D. Mammalian embryonic cerebrospinal fluid proteome has greater apo-lipoprotein and enzyme pattern complexity than the avian proteome. Journal of proteome research 2005; 4(6): 2420- 2428.
  17. Martin C, Bueno D, Alonso MI, Moro JA, Callejo S, Parada C, Martin P, Carnicero E, Gato A. FGF2 plays a key role in embryonic cerebrospinal fluid trophic properties over chick embryo neuro epithelial stem cells. Dev Biol 2006 Sep 15;297(2):402-16. Epub 2006 May 19.
  18. Huttner HB, Janich P, Kohrmann M, Jaszai J, Siebzehnrubl F, Blumcke I, Suttorp M, Gahr M, Kuhnt D, Nimsky C. The stem cell markerprominin-1/CD133 on membrane particles in human cerebrospinal fluid offers novel approaches for studying central nervous system disease. Stem Cells 2008 Mar;26(3):698-705.
  19. Nordin C, Gupta RC, Sjodin I. Cerebrospinal fluid amino acids in pathological gamblers and healthy controls. Neuropsychobiology 2007;56(2-3):152-8. doi: 10.1159/000115782. Epub 2008 Feb 7.
  20. Gato A, Martin P, Alonso MI, Martin C, Pulgar MA, Moro JA. Analysis of cerebro-spinal fluid protein composition in early developmental stages in chick embryos. J Exp Zool A Comp Exp Biol 2004 Apr 1;301(4):280-9.
  21. Mitchell KE, Weiss ML. Matrix cells from Wharton’s jelly form neurons and glia. Stem Cells 2003;21(1):50-60.
  22. Marcus AJ, Woodbury D. Fetal stem cells from extra-embryonic tissues: do not discard. J Cell Mol Med 2008 Jun;12(3):730-42. doi: 10.1111/j.1582- 4934.2008.00221.x. Epub 2008 Jan 11.
  23. Miao Z, Jin J, Chen L, Zhu J, Huang W, Zhao J, Quian H, Zhang X. Isolation of mesenchymal stem cells from human placenta: comparison with human bone marrow mesenchymal stem cells. Cell Biol Int 2006 Sep;30(9):681-7. Epub 2006 Apr 22.
  24. In ‘tAnker PS, Scherjon SA, Kleijburg-van der Keur C, Noort WA, Claas FHJ, Willemze R, Fibbe WE, Kanhai HHH. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 2003;102(4):1548-49.
  25. Magatti M, De Munari S, Vertua E, Gibelli L, Wengler GS, Parolini O. Human amnion mesenchyme harbors cells with allogeneic T-cell suppression and stimulation capabilities. Stem Cells 2008 Jan;26(1):182-92. Epub 2007 Sep 27.
  26. Kang XQ, Zang WJ, Bao LJ, Li DL, Xu XL, Yu XJ. Differentiating characterization of human umbilical cord blood-derived mesenchymal stem cells in vitro. Cell Biol Int 2006 Jul;30(7):569-75. Epub 2006 Mar 6.
  27. Kern S, Eichler H, Stoeve J, Kluter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells 2006 May;24(5):1294-301. Epub 2006 Jan 12.
  28. Wagner W, Wein F, Seckinger A, Frankhauser M, Wirkner U, Krause U, et al. Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood. Exp Hematol 2005 Nov;33(11):1402-16.
  29. Jackson JS, Golding JP, Chapon C, Jones WA, Bhakoo KK: Homing of stem cells to sites of inflammatory brain injury after intracerebral and intravenous administration: a longitudinal imaging study. Stem Cell Res Ther 2010 Jun 15;1(2):17. doi: 10.1186/scrt17.
  30. Romanov YA, Svintsitskaya VA, Smirnov VN. Searching for alternative sources of postnatal human mesenchymal stem cells: candidate MSC-like cells from umbilical cord. Stem Cells 2003;21(1):105-10.
  31. Weiss ML, Medicetty S, Bledsoe AR. Human umbilical cord matrix stem cells: Preliminary characterization and effect of transplantation in a rodent model of Parkinson’s disease. Stem Cells 2006 Mar;24(3):781-92.
  32. Carlin R, Davis D, Weiss M. Expression of early transcription factors Oct4, Sox2 and Nanog by porcine umbilical cord (PUC) matrix cells. Reprod Biol Endocrinol. 2006 Feb;6(4):8.
  33. Usha Nekanti, Sumitava Dastidar, Parvathy Venugopal, Satish Totey and Malancha Ta. Increased Proliferation and Analysis of Differential Gene Expression in Human Wharton’s Jelly-derived Mesenchymal Stromal Cells under Hypoxia. Int J Biol Sci. 2010; 6(5): 499–512.

Neurometabolic Disorder Articles


Homocystinuria: Diagnosis and Neuroimaging Findings - of Iranian Pediatric patients

Parvaneh KARIMZADEH, Narjes JAFARI*, Mohammad Reza ALAI, Sayena JABBEHDARI, Habibeh NEJAD BIGLARI

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 94-98
https://doi.org/10.22037/ijcn.v9i1.7994

How to Cite This Article: Karimzadeh P, Jafari N, Alai MR, Jabbehdari S, Ahmad Abadi F, NejadBiglari H. Homocystinuria: Diagnosis and Neuroimaging Findings - of Iranian Pediatric Patients. Iran J Child Neurol. 2015 Winter;9(1):94-98.

Abstract
Objective
Homocystinuria is a neurometabolic diseases characterized by symptoms include Neurodevelopmental delay, lens dislocation, long limbs and thrombosis.

Materials & Methods
The patients who were diagnosed as homocystinuria marfaniod habits, seizure in the Neurology Department of Mofid Children’s Hospital in Tehran, Iran between 2004 and 2014 were included in our study. The disorder was confirmed by clinical andneuroimaging findings along withneurometabolic and genetic assessment fromreference laboratory in Germany. We assessed age, gender, past medical history, developmental status, clinical manifestations, and neuroimaging
findings of 20 patients with homocystinuria.

Results
A total of 75% of patients were offspring from consanguineous marriages. A total of 95% of patients had a history of developmental delay and 40% had developmental regression. A total of 75% had seizures from these 45% showed refractory seizures. Seizures among 13 patients werecontrolled with suitable homocystinuria treatment. The patients with homocystinuriawere followed for approximately 10 years and the follow-ups showed that the patients with an early diagnosis and treatment had more favorable clinical responses for growth index,
controlled refractory seizures, neurodevelopmental status, and neuroimaging findings. Neuroimaging findings include brain atrophy and/or white matter involvement.

Conclusion
According to the results of this study, we suggest that early assessment and detectionplayan important role in the prevention of disease progression and clinical signs. Homocystinuria in patients with a positive family history, developmental delays, or regression, refractory, or recurrent seizures should take precedence over other causes.

References

  1. Brustolin S1, Giugliani R, Félix TM. Genetics of homocystine metabolism and associated disorders.Braz J Med Biol Res. 2010 Jan; 43(1):1-7.
  2. Iacobazzi V, Infantino V, Castegna A, AndriaG. Hyperhomocysteinuria: Related genetic diseases and congenital defects, abnormal DNA methylation and newborn screening issues. Mol Genet Metab. 2014 September - October; 113(1-2):27-33.
  3. SelhubJ.Homocystinemetabolism.Annu Rev Nutr. 1999;19:217-46.
  4. Ali Z, Troncoso JC, Fowler DR.Recurrent cerebral venous thrombosis associated with heterozygote methylenetetrahydrofolate reductase C677T mutation and sickle cell trait without Homocysteinuria: An autopsy case report and review of literature.ForensicSci Int. 2014 Sep;242:e52-5.
  5. RozenR.Genetic modulation of Homocysteinuria. SeminThrombHemost. 2000;26(3):255-61.
  6. Palma Reis R, Sales Luis. Homocisteinemia e Doença Vascular – O Nascer de um Novo Factor de Risco. Rev Port Cardiol 1999; 18:507-14
  7. McCully KS. Homocystine theory of arteriosclerosis: development and current status. Atherosclerosis Rev 1983;11:157-246.
  8. Malinow MR. Homocystinuria: a common and easily reversible risk factor for occlusive atherosclerosis. Circulation 1990;81:2004-6.
  9. Boushey CJ, Beresford SAA, Omenn GS, Motulsky AG. A quantitive assessment of plasma homocystine as a factor for vascular disease: probable benefits of increasing folic acid intakes. JAMA 1995;472:1049-57.
  10. Carson NAJ, Neill DW. Metabolic abnormalities detectedin a survey of mentally backward individuals in NorthernIreland. Arch Dis Child 1962;37:505-13.
  11. Gerritsen T, Waisman HA. Homocystinuria, an error inthe metabolism of methionine. Pediatrics 1964;33:413-20.
  12. Mallory T. Case records of the Massachusetts GeneralHospital.Case 19471. Marked cerebral symptoms followinga limp of three months duration. N Engl J Med 1933;209:1063-6.
  13. Shih VE, Efron ML. Pyridoxine-unresponsive homocystinuria. Final diagnosis of MGH case 19471. N Engl Med1970;283:1206-8.
  14. Kang SS, Wong PWK, Malinow MR. Hyperhomocyst(e) inemia as a risk factor for occlusive vascular disease.Ann Rev Nutr 1992;12:279-98.
  15. Mudd SH, Skovby F, Levey HL, et al. The natural history of homocystinuria due to cystathionine B-synthase deficiency. Am J Hum Genet 1985;37:1-31.
  16. Fonseca N1, Vidal N, Santos J, Brito AP, Bernardino L, Silvestre I, InêsL.Hyperhomocysteinuria–case report.Rev Port Cardiol. 2003 Feb;22(2):223-30.
  17. Mudd SH, Levy HL. Disorders of transsulfuration. InStanbury JB, Wingarden JB, Fredrickson DS, Goldstein JL,Bronw MS (eds.). The Metabolic Basis of Inherited Disease.New York, McGraw-Hill 1983;522-559.
  18. Sachdeva V1, Mittal V, Pathengay A, Kekunnaya R, Gupta A, Rao BV.Isolated abducens nerve palsy with hyperhomocysteinuria: Association and outcomes.Indian J Ophthalmol. 2013 Oct;61(10):598-600.

Case Report


How to Cite This Article: Ghanizadeh A, Berk M. Beta-Lactam Antibiotics as A Possible Novel Therapy for Managing Epilepsy and Autism, A Case Report and Review of Literature. Iran J Child Neurol. 2015 Winter;9(1):99-102.

Abstract

Autism is a disorder of unknown etiology. There are few FDA approved medications for treating autism. Co-occurring autism and epilepsy is common, and glutamate antagonists improve some symptoms of autism. Ceftriaxone, a beta-lactam antibiotic, increases the expression of the glutamate transporter 1 which decreases extracellular glutamate levels. It is hypothesized that modulating astrocyte glutamate transporter expression by ceftriaxone or cefixime might improve some symptoms of autism. This case report of a child with autism and epilepsy suggests a decrease in seizures after taking cefixime.

References

  1. Selassie AW, Wilson DA, Martz GU, Smith GG, Wagner JL, Wannamaker BB. Epilepsy beyond seizure: a population-based study of comorbidities. Epilepsy Res 2014;108(2): 305-315.
  2. Sansa G, Carlson C, Doyle W, Weiner HL, Bluvstein, Barr W, et al. Medically refractory epilepsy in autism. Epilepsia 2011; 52(6): 1071-1075.
  3. Ghanizadeh A. Increased glutamate and homocysteine and decreased glutamine levels in autism: a review and strategies for future studies of amino acids in autism. Dis Markers; 2013;35(5): 281-286.
  4. Yamada T, Kawahara K, Kosugi T, Tanaka M.Nitric oxide produced during sublethal ischemia is crucial for the preconditioning-induced down-regulation of glutamate transporter GLT-1 in neuron/astrocyte co-cultures. Neurochem Res 2006:31(1): 49-56.
  5. Zeng, LH, Ouyang Y, Gazit V, Cirrito JR, Jansen LA, Ess KC, et al. Abnormal glutamate homeostasis and impaired synaptic plasticity and learning in a mouse model of tuberous sclerosis complex. Neurobiol Dis 2007; 28(2): 184-196.
  6. Zeng LH, Bero AW, Zhang B, Holtzman D. M. , Wong M. Modulation of astrocyte glutamate transporters decreases seizures in a mouse model of Tuberous Sclerosis Complex. Neurobiol Dis 2010; 37(3): 764-771.
  7. Nizzardo M, Nardini M, Ronchi D, Salani S, Donadoni C, Fortunato F, et al. Beta-lactam antibiotic offers neuroprotection in a spinal muscular atrophy model by multiple mechanisms. Experimental Neurology 2011; 229: 214–225.
  8. Harada M, Taki MM, Nose A, Kubo H, Mori K, Nishitani H and T. Matsuda. Non-Invasive Evaluation of the GABAergic/Glutamatergic System in Autistic Patients Observed by MEGA-Editing Proton MR Spectroscopy Using a Clinical 3 Tesla Instrument. J Autism Dev Disord 2010 Apr;41(4):447-54. doi: 10.1007/s10803-010-1065- 0.
  9. Polleux, F. and J. M. Lauder. Toward a developmental neurobiology of autism. Ment Retard Dev Disabil Res Rev 2004;10(4): 303-317.
  10. Maezawa I, Jin LW. Rett syndrome microglia damage dendrites and synapses by the elevated release of glutamate. J Neurosci 2010;30(15): 5346-5356.
  11. Ghanizadeh A. Targeting neurotensin as a potential novel approach for the treatment of autism. J Neuroinflammation 2010;7(1): 58.
  12. Ghanizadeh A. Transplantation of GABAergic cell line as a novel hypothesized treatment for autism. Epilepsy & Behavior 2010;19(4): 664.
  13. Kim SY, Jones TA. The effects of ceftriaxone on skill learning and motor functional outcome after ischemic cortical damage in rats. Restor Neurol Neurosci 2013;31(1):87-97. doi: 10.3233/RNN-2012-120245.
  14. Ghanizadeh A.Could fever and neuroinflammation play a role in the neurobiology of autism? A subject worthy of more research. Int J Hyperthermia 2011;27(7): 737-738.
  15. Ghanizadeh A. May lovastatin target both autism and epilepsy? A novel hypothesized treatment. Epilepsy Behav 2011;20(2): 422.
  16. Ghanizadeh A. Hydrogen as a novel hypothesized emerging treatment for oxidative stress in autism. Eur Rev Med Pharmacol Sci 2012;16(9): 1313-1314.
  17. Ghanizadeh A. Physical exercise and intermittent administration of lactulose may improve autism symptoms through hydrogen production. Med Gas Res 2012;2(1): 19.
  18. Asadabadi M, Mohammadi MR, Ghanizadeh A, Modabbernia A, Ashrafi MR, Hassanzadeh E, et al. Celecoxib as adjunctive treatment to risperidone in children with autistic disorder: a randomized, double-blind, placebo-controlled trial. Psychopharmacology (Berl) 2013 Jan;225(1):51-9. doi: 10.1007/s00213-012- 2796-8. Epub 2012 Jul 11.
  19. Wang DD, Englot DJ, Garcia PA, Lawton MT, Young WL. Minocycline- and tetracycline-class antibiotics are protective against partial seizures in vivo. Epilepsy Behav 2012;24(3): 314-318.
  20. Librizzi L, Noe F, Vezzani A, Curtis M, Ravizza T. Seizure-induced brain-borne inflammation sustains seizure recurrence and blood-brain barrier damage. Ann Neurol 2012;72(1): 82-90.
  21. Gasior MK, Nieoczym SD, Wlaz P. Clavulanic acid does not affect convulsions in acute seizure tests in mice. J Neural Transm 2012;119(1): 1-6.
  22. Beheshti Nasr SM, Moghimi A, M. Mohammad-Zadeh M, Shamsizadeh A and Noorbakhsh SM. The effect of minocycline on seizures induced by amygdala kindling in rats. Seizure 2013;22(8): 670-674.
  23. Erickson CA, Posey DJ, Stigler KA, Mullett J, Katschke AR,McDougle CJ. A retrospective study of memantine in children and adolescents with pervasive developmental disorders. Psychopharmacology (Berl) 2007;191(1): 141-147.
  24. Thabet F, Al Maghrabi M, Al Barraq A, Tabarki B. Cefepime-induced nonconvulsive status epilepticus: case report and review. Neurocrit Care 2009;10(3): 347-351.

Small Duplication of HPRT 1 Gene May Be Causative For Lesh-Nyhan Disease in Iranian Patients

Razieh BOROUJERDI, Mohsen SHARIATI, Hosein NADDAFNIA, Hojatolah REZAEI*

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 103-106
https://doi.org/10.22037/ijcn.v9i1.6080

How to Cite This Article: Boroujerdi R, Shariati M, Naddafnia H, Rezaei H. Small Duplication of HPRT 1 Gene May Be Causative For Lesh-
Nyhan Disease in Iranian Patients. Iran J Child Neurol. 2015 Winter;9(1):103-106.

Abstract

Deficiency of hypoxanthine-guanine phosphoribosyltransferase (HGPRT) is a rare inborn error of purine metabolism and is characterized by uric acid overproduction along with a variety of neurological manifestations that depend on a degree of the enzymatic deficiency. Inheritance of HPRT deficiency is X-linked recessive; thus, males are generally more affected and heterozygous females are carriers (usually asymptomatic). Human HPRT is encoded by a single structural gene on the long arm of the X chromosome at Xq26. More than 300 mutations in the HPRT1 gene have been detected. Diagnosis can be based on clinical and biochemical findings as well as enzymatic and molecular testing. Molecular diagnosis is the best way as it allows for faster and more accurate carrier and prenatal diagnosis. In this report, a new small duplication in the HPRT1 gene was found by sequencing, which has yet to be reported.

References

  1. Fu R, Jinnah HA. Genotype-Phenotype Correlations in Lesch-Nyhan Disease Moving Beyond The Gene. Journal of Biological Chemistry. 2012; 287(5):2997- 3008.
  2. Fontenelle LJ, Henderson JF. An enzymatic basis for the inability of erythrocytes to synthesize purine ribonucleotides de novo. Biochim Biophys Acta. 1969 Feb 18; 177(1):175-6. PubMed PMID: 5781193.
  3. Kelley WN, Wyngaardcn JB. Clinical syndromes associated with hypoxanthine guanine Phosphoribosyl transferase deficiency. In: J. B. Stanbury, J. B. Wyngaarden, D. S. Frederickson, J. L. Goldstein, M. S. Brown, editors. The Metabolic Basis of Inherited Disease. 5 ed. New York: McGraw Hill; 1983. p. 1115-43.
  4. Lesch M, Nyhan WL. A Familial Disorder of Uric Acid Metabolism and Central Nervous System Function. Am J Med. 1964 Apr; 36:561-70. PubMed PMID: 14142409.
  5. Christie R, Bay C, Kaufman IA, Bakay B, Borden M, Nyhan WL. Lesch-Nyhan disease: clinical experience with nineteen patients. Dev Med Child Neurol. 1982 Jun; 24(3):293-306. PubMed PMID: 7095300.
  6. Neychev VK, Jinnah HA. Sudden death in Lesch-Nyhan disease. Dev Med Child Neurol. 2006 Nov; 48(11):923- 6. PubMed PMID: 17044962. PubMed Central PMCID: 3507438.
  7. Torres RJ, Puig JG. Hypoxanthine - guanine phosphoribosyltransferase (HPRT) deficiency: Lesch- Nyhan syndrome. Orphanet J Rare Dis. 2007; 2:48.PubMed PMID: 18067674. PubMed Central PMCID: 2234399.
  8. Finette BA, Kendall H, Vacek PM. Mutational spectral analysis at the HPRT locus in healthy children. Mutat Res. 2002 Aug 29; 505(1-2):27-41. PubMed PMID: 12175903.
  9. Patel PI, Framson PE, Caskey CT, Chinault AC. Fine structure of the human hypoxanthine phosphoribosyltransferase gene. Mol Cell Biol. 1986 Feb; 6(2):393-403. PubMed PMID: 3023844. PubMed Central PMCID: 367528.
  10. Edwards A, Voss H, Rice P, Civitello A, Stegemann J, Schwager C, et al. Automated DNA Sequencing of the human HPRT locus. Genomics. 1990 Apr; 6(4):593-608. PubMed PMID: 2341149.
  11. Jinnah HA, De Gregorio L, Harris JC, Nyhan WL, O’Neill JP. The spectrum of inherited mutations causing HPRT deficiency: 75 new cases and a review of 196 previously reported cases. Mutat Res. 2000 Oct; 463(3):309-26. PubMed PMID: 11018746. Epub 2000/10/06. eng.
  12. Jinnah HA, Harris JC, Nyhan WL, O’Neill JP. The spectrum of mutations causing HPRT deficiency: an update. Nucleosides Nucleotides Nucleic Acids. 2004 Oct; 23(8-9):1153-60. PubMed PMID: 15571220. Epub 2004/12/02. eng.
  13. Bouwens-Rombouts AG, van den Boogaard MJ, Puig JG, Mateos FA, Hennekam RC, Tilanus MG. Identification of two new nucleotide mutations (HPRTUtrecht and HPRTMadrid) in exon 3 of the human hypoxanthine-guanine phosphoribosyltransferase (HPRT) gene. Human genetics. 1993 Jun; 91(5):451-4. PubMed PMID: 8314557.
  14. Lovett ST, Drapkin PT, Sutera VA, Jr., Gluckman- Peskind TJ. A sister-strand exchange mechanism for recA-independent deletion of repeated DNA sequences in Escherichia coli. Genetics. 1993 Nov; 135(3):631- 42. PubMed PMID: 8293969. PubMed Central PMCID: 1205708.
  15. Hartl L.D, Ruvolo M. Genetic Analysis of Genes and Genomes. Burlington: Jones & Bartlett Learning; 2012. p. 529.
  16. McKeran RO, Andrews TM, Howell A, Gibbs DA, Chinn S, Watts WE. The diagnosis of the carrier state for the Lesch--Nyhan syndrome. Q J Med. 1975 Apr; 44(174):189-205. PubMed PMID: 1178810.
  17. O’Neill JP. Mutation carrier testing in Lesch-Nyhan syndrome families: HPRT mutant frequency and mutation analysis with peripheral blood T lymphocytes. Genet Test. 2004 Spring; 8(1):51-64. PubMed PMID: 15140374.
  18. Torres RJ, Buno A, Mateos FA, Puig JG. Carrier state in HGPRT deficiency. A study in 14 Spanish families. Adv Exp Med Biol. 1998; 431:197-200. PubMed PMID: 9598058.
  19. Kleijer WJ, Van Den Berg P, Los FJ. Prenatal diagnosis of Lesch-Nyhan disease. Prenat Diagn. 2004 Aug; 24(8):658-9; author reply 9. PubMed PMID: 15305358.
  20. Graham GW, Aitken DA, Connor JM. Prenatal diagnosis by enzyme analysis in 15 pregnancies at risk for the Lesch- Nyhan syndrome. Prenat Diagn. 1996 Jul; 16(7):647-51. PubMed PMID: 8843475.
  21. Gibbs RA, Nguyen PN, McBride LJ, Koepf SM, Caskey CT. Identification of mutations leading to the Lesch- Nyhan syndrome by automated direct DNA sequencing of in vitro amplified cDNA. Proceedings of the National Academy of Sciences of the United States of America. 1989 Mar; 86(6):1919-PubMed PMID: 2928313. PubMed Central PMCID: 286816.
  22. Mak BS, Chi CS, Tsai CR, Lee WJ, Lin HY. New mutations of the HPRT gene in Lesch-Nyhan syndrome. Pediatric neurology. 2000 Oct; 23(4):332-5. PubMed PMID: 11068166.

Epstein-Barr Virus Encephalitis: A Case Report

Somayh HASHEMIAN, Farah ASHRAFZADEH*, Javad AKHONDIAN, Mehran BEIRAGHI TOOSI

Iranian Journal of Child Neurology, Vol. 9 No. 1 (2015), 22 January 2015, Page 107-111
https://doi.org/10.22037/ijcn.v9i1.5096

How to Cite This Article: Hashemian S, Ashrafzadeh F, Akhondian J, Beiraghi Toosi M. Epstein-Barr Virus Encephalitis: A Case Report. Iran J Child Neurol. 2015 Winter;9(1):107-110.

 

 Abstract

Many neurologic manifestations of Epstein-Barr virus (EBV) infection have been documented, including encephalitis, aseptic meningitis, transverse myelitis, and Guillain-Barre syndrome. These manifestations can occur alone or coincidentally with the clinical picture of infectious mononucleosis. EBV encephalitis is rare and is indicated as a wide range of clinical manifestations. We report a 10-year-old girl presented with fever, gait disturbance, and bizarre behavior for one week. The results of the physical examination were unremarkable. The diagnosis of EBV encephalitis was made by changes in titers of EBV specific antibodies and MRI findings. A cranial MRI demonstrated abnormal high signal intensities in the basal ganglia and the striatal body, especially in the putamen and caudate nucleus. EBV infection should be considered when lesions are localized to the basal ganglia.

References

  1. Fujimoto H, Asaoka K, Imiazumi T, Ayabe M, Shoji H, Kaji M. Epstein-Barr virus Infections of the Central Nervous System. Intern Med 2003; 42:33-40.
  2. Mathew AG, Parvez Y. Fulminant Epstein Barr virus encephalitis. Indian Pediatrics 2013; 50:418-419
  3. Kalita J, Maurya PK, Kumar B, Misra UK. Epstein Barr virus encephalitis: Clinical diversity and radiological similarity. Neurol India 2011; 59:605-7
  4. Baskin HJ, Hedlund G. Neuroimaging of Herpes Virus Infections in Children. Pediatr Radiol 2007; 37:949-63.
  5. Weinberg A, Li SH, Palmer M, Tyler K .Quantitative CSF PCR in Epstein-Barr Virus Infections of the Central Nervous System. Ann Neurol 2002; 52:543-8.
  6. Ono J, Shimizu K, Harada k, Mano T, Okada S. Characteristic MR Features of Encephalitis Caused by Epstein-Barr virus. Pediatr Radiol 1998; 28:569-70.
  7. Hausler M, Raamaekers T, Doenges M, Shweizer K ,Ritter K. Neurological Complications of Acute and Persistent Epstein-Barr Virus Infection in Pediatric Patients. Journal of Medical Virology 2002; 68:253-63.
  8. Young JY, Hyang LK. Transient Asymptomatic White Matter Lesions Following Epstein-Barr virus Encephalitis. Korean pediatric society 2011; 54:389- 93.
  9. Doja A, Bitnun A, Jones EL, Richardson S, Tellier R, Petric M, et al. Pediatric Epstein-Barr Virus-Associated Encephalitis:10-Year Review. Child Neurol 2006; 21:385-91.
  10. Kou K, Itoh M, Kawano Y. A Case Report of EB Virus- Induced Meningoencephalitis Associated with Brain MRI Abnormalities (basal ganglia). J Japan Peditr Sos 1994; 98:2052-9.
  11. Kunlong H, Hung-Tsai L, Minlan T. Epstein-Barr Virus Encephalitis in Children. Acta Pediatrica Taiwanica 2000; 3:140-6.