Mutation spectrum of 7 exons of ATP7B gene in

Iranian patients with Wilson disease

Narges Zali, Seyed Reza Mohebbi, Sahar Esteghamati, Farzad Firouzi, Seyed Mohammad Kazem Hosseini Asl, Mohsen Chiani, Faramarz Derakhshan, Amir Houshang Mohammad Alizadeh, Seyed Ali Malek Hosseini, Mohammad Reza Zali

Research Centre for Gastroenterology and Liver Diseases, Shahid Beheshti Medical University, Tehran, Iran

 

ABSTRACT

Aim: To investigate the ATP7B gene in ethnically different Iranian patients with Wilson disease.

Background: Copper transporting beta polypeptide, ATP7B or WND gene is predominantly expressed in liver and has been identified as the defective gene. WND gene is located on Chr13q14.3 and consists of 21 expressed exons.

Patients and methods: Diagnosis of WD was verified according to the biochemical characterizations including serum ceroluplasmin (<0.2g/l), 24-hour urine copper after challenging by D-penicillamine (>25mmol/24h), liver copper (>250µg/g dry weight), Kayser-Fleischer rings, Cu-ATPase activity, and liver and brain imaging features. A total of 70 patients aged 5-40 years, were included. Patients were classified in three different categories; hepatic, neurologic and combined based on their clinical presentations. Bi-directional sequencing was performed using the ABI 3130xl, Genetic analyzer (Applied-Biosystem).

Results: Totally, 12 different mutations have been found in 18 patients with Wilson's disease. Of these, T788I, 2803-2805 ACG>-TG, 2532delA and H1069N were novel mutations. Meanwhile, we have found 4 polymorphisms.

Conclusion: Mutations are highly distributed among the different exons of ATP7B gene in Iranian patients with WD.

Keywords: ATP7B gene, Wilson disease, Iran.

(Gastroenterology and Hepatology from bed to bench 2008;1(1):3-9).

 

 


INTRODUCTION


[1]

Wilson's disease (WD), an autosomal recessive disorder with incidence of 1 in 30,000 (1 in 5000 to 1 in 30,000), is characterized by excessive copper accumulation in liver and brain. Abnormal accumulation in liver can induce asymptomatic rise of enzymes or major clinical illnesses such as, chronic active hepatitis, liver cirrhosis, and fulminant acute hepatic failure with or without intravenous hemolysis (1-3). Excessive copper in the brain induces neurological dysfunction such as bradykinesia, rigidity, tremor, chorea and dystonia. KF ring (Kayser Fleischer) is an important eye sign  almost universally present in Wilson patients with neurological manifestations. Apart from extarpyramidal syndrome, other neurological presentations are cerebellar ataxia, progressive myoclonic epilepsy, cognitive deterioration and proximal muscle weakness (4-7). Biochemically, Wilson is characterized by low plasma ceruloplasmin and copper, elevated liver Cu, and increased excretion of 24-hour urinary Cu (8,9).

Copper transporting beta polypeptide, ATP7B or WND gene (OMIM#277900), is predominantly expressed in liver, and has been identified as the defective gene. WND gene is located on Chr13q14.3 and consists of 21 expressed exons (10,11). It spans 80 kb of genomic sequence encoding a protein with several membrane-spanning domains, an ATPase consensus sequence, a hinge domain, a phosphorylation site, and at least 2 putative copper-binding sites. This protein functions as a monomer, exporting copper out of the cells, such as the efflux of hepatic copper into the bile. Alternate transcriptional splice variants, encoding different isoforms with distinct cellular localizations, have been characterized (12).

To date, more than 250 diseases causing mutations have been reported in ATP7B gene that are widely distributed among its different exons, however, of which 25% are located in its ATP-binding domain. Furthermore, these mutations are due to splice-site, small insertions/deletions, non-sense and missense mutations in a frequency of 7.4%, 27.0%, 7.4% and 58.2%, respectively (13). It should be noted, some of these mutations are population specific and much prevalent in a particular population such as, either H1069Q in European or R778L in Asian population (14).

With respect to the numerous reported mutations in ATP7B gene, as well as lack of the information regarding the WND mutations in Iranian population, we decided to investigate the ATP7B gene in ethnically different Iranian patients with Wilson disease. In the present study we analyzed some exons of ATP7B gene by sequencing method to elucidate the ATP7B mutation spectrum in patients with Wilson disease. 

 

PATIENTS and METHODS

We included all WD patients referring to Research Center for Gastroenterology and Liver Diseases (RCGLD) of Shahid Beheshti Medical University in Tehran, Iran.

Diagnosis of WD was verified according to the biochemical characterizations including serum ceroluplasmin (<0.2g/l), 24-hour urine copper after challenging by D-penicillamine (>25mmol/24h), liver copper (>250µg/g dry weight), Kayser-Fleischer rings, Cu-ATPase activity, and liver and brain imaging features. Patients who met two of the aforementioned features entered the study. Meanwhile all patients were checked for abnormal liver enzymes, including alkaline phosphatase (ALK.P), aspartate aminotransferase (AST), alanine aminotransferase (ALT), and total bilirubin, direct bilirubin, albumin, international normalized ratio (INR), and prothrombin time (PT). A total of 70 patients aged 5-40 years, were included. Patients were classified in three different categories, hepatic (58 cases), neurologic (5 cases) and combined (7 cases) based on their clinical presentations. Hepatic patients had liver involvement. Neurological patients had neurological symptoms such as tremor, clumsiness, dysarthria, and ataxia in addition to dystonic or parkinsonian syndrome. Mixed-type patients were diagnosed based on liver involvement and neurological features.

The study was reviewed by the local ethics committee and appropriate informed consent was obtained from all patients and their families.

Table 1. Primers, their sequence, annealing temperature and extension time

Exon primers

5'→3'

Tm (°C)

extension time (sec)

Exon6- F

Exon6- R

AAACCCACAAAGTCTACTGAGGCAC

GTTGGGCCCAGGTAGAGGAAG

65

30

Exon8-9-F

Exon8-9- R

TGTCGCTCATTGAACTCTCCTCC

CTTTCGTAGCTGGATTGAGAGTGG

67

40

Exon10-11-12-F

Exon10-11-12- R

AGTGGCCATGTGAGTGATAAG

TAAAACACAACCACCATATAGCC

59

40

Exon13- F

Exon13- R

ATGGCAGAGCAGTGTGGAATAC

GTGGCTCTCAGGCTTTTCTCTC

63

40

Exon14- F

Exon14- R

GGTGTCTTGTTTCCTGTCTGAG

TAGGAGAGAAGGACATGGTGAG

61

30

F: Forward, R: Reverse

Polymerase Chain Reaction and Sequencing

DNA was extracted from whole blood using proteinase K-phenol-chloroform method. Eight exons (6,8,9,10,11,12,13,14) of ATP7B gene were amplified by PCR in a 25µl reaction (H2O 19.4µl, dNTP 10mMol 0.5µl, MgCl2 50mMol 0.75µl, 0.3µl of 12.5pMol primers (as listed in table 1), and Taq DNA polymerase 1unit). Amplification was performed in a three step reaction for 40 cycles (for exons 6 and 14) and 45 cycles for the other exons, 95°C 30s, 72°C 30s, 95°C 5min, and 72°C 10min as initial denaturation and final extension.

Bi-directional sequencing was performed using the ABI 3130xl, Genetic analyzer (Applied-Biosystem, USA) in accordance with supplier's instructions. Forward and reverse sequences were aligned and compared with reference sequence using computer software Bioedit and clustalX.

To assess the possibility that the novel mutations detected in this study might be polymorphisms that do not alter the function of the gene, we tested 50 control chromosomes in our population. We have classified sequence changes in the WD gene as polymorphisms if they do not modify the amino acid sequence of the protein product or result in nonconservative changes in nonessential residues of the protein or are detected in normal chromosomes of the same population (control group) or in chromosomes with defined disease causing mutation.

Mutations are named according to the guidelines from http://www.HGMD.org/, using the reference sequence with the GenBank accession number NM_00053.1. The nucleotide +1 is the A of the ATG translation initiation codon. The ATG translation initiation codon is also the first codon.

 

RESULTS

We screened 70 WD patients from 54 unrelated families for nucleotide variants in seven exons of the ATP7B gene. The study population included 40 males and 30 females with their age ranged 5-40 years.

Of 70 patients, 18 (25.7%) had at least one mutation, while 52 patients failed to show any mutation in the studied exons. Altogether, 12 different mutations including 9 missense and 3 deletion (one of which is nonsense) have been found. According to the literature and the “Wilson disease mutation database” (http://www.uofa-medical-genetics.org/wilson/ index.html), the following 4 mutations are being reported for the first time: T778I, 2532delA, 2803-2805ACG>-TG, and H1069N (table 2).

Totally, 15 (21.4%) patients were homozygous, one (1.4%) was compound heterozygous with different mutations identified on both alleles and 2 (2.9%) were compound heterozygous for whom the second mutation was not identified. The most common mutation is H1069Q point mutation, presents in 4 of 70 patients (5.7%), all of whom are homozygous. Table 2 summarizes the various identified genotypes and the corresponding age at disease onset, presence of KF ring plus the involved organ liver, brain or both.

Interestingly, D642H mutation has been detected in one family which leads to both hepatic and mixed Wilson's disease presentations. In the heterozygous form of this mutation, our patients were two sisters with hepatic WD, and their brother who was homozygous for D642H and presented with rather neurological WD.

A novel substitution in exon 9 involving the transmembrane domain of the protein, Thr788Ilu, was identified in 2 homozygous patients who were brothers. The change of a hydrophilic to a non-polar hydrophobic amino acid can change its function. Surprisingly, the elder brother (28 years) had only neurologic manifestation, while the younger (23 years) presented with acute hepatic failure and was subjected for liver transplantation. Both were revealed to have KF ring. This mutation was not found in any of the normal chromosomes we tested.

Table 2. Characteristics of 27 Iranian patients with Wilson’s disease with ATP7B mutations

Mutation

Nucleotide

change

Type

Exon

Domain

Manifestation

KFR

(+/-)

Age at onset of symptoms (yrs)

Homozygotes

 

 

 

 

 

 

 

 

1639delAT

1639delAT

 

Deletion

6

Between CuBD and TM

H

+

9

D642H

D642H

1924G>C

 

Missense

6

Cu6/TM1

H/N

-

16

T778I

T778I

2363C>T

Missense

9

TM4

N

+

28

T778I

T778I

2363C>T

Missense

9

TM4

H

+

23

P840L

P840L

2519C>T

Missense

10

TD

H

?

25

2532delA

2532delA

 

Deletion

10

TD

H

+

9

V890M

V890M

2668G>A

Missense

11

TD/TM5

H

-

19

H1069Q

H1069Q

3207C>A

Missense

14

ATP loop

A874V

A874V

2621C>T

Missense

11

TD

H

-

12

A874V

A874V

2621C>T

Missense

11

TD

H

-

14

2803-2805 ACG>-TG

2803-2805 ACG>-TG

 

Nonsense

11

TD

H

+

21

H1069N

H1069N

3205C>A

Missense

14

ATP loop

H/N

+

19

H1069Q

H1069Q

3207C>A

Missense

14

ATP loop

H

+

17

H1069Q

H1069Q

3207C>A

Missense

14

ATP loop

H

+

18

H1069Q

H1069Q

3207C>A

Missense

14

ATP loop

H

+

18

Compound heterozygote

R778L

R969Q

2333G>T/2906G>A

Missense/ Missense

8/13

TM4/TM6

H

?

22

Compound heterozygote (second mutation not yet identified)

D642H

?

1924G>C

 

Missense

6

Cu6/TM1

H

-

14

D642H

?

1924G>C

 

Missense

6

Cu6/TM1

H

-

21

 

H: Hepatic, N: Neurologic, CuBD: Copper biding domain, TM: Transmembrane Domain, TD: Transduction Domain

 

 

Table 3. Comparison of the observed polymorphisms in

WD patients and controls

SNP

exons

Frequency  in cases

Frequency in controls

A2495G (K831R)

10

0.31

0.38

G2855A (K952R)

12

0.26

0.31

G3009A (A1003A)

13

0.1

0.15

G2973A (T991T)

13

0.05

0.08

 

 

  Exon 14 has been identified as a hot spot for mutations. We detected two disease causing mutations in this exon, one of which was novel. Totally, 4 patients revealed to have H1069Q, of whom one has another disease causing mutation (V890M) in exon 11. Surprisingly, all of these patients presented with hepatic involvement. H1069N mutation was detected in a 19-year old girl, presented with acute hepatic failure and neurological manifestations.

One of the novel deletions found in this study involved one nucleotide in exon 10 (2532delA). It occurred in a 9-year old boy presented with hepatic failure.

2803-2805ACG>-TG mutation was detected in a member of a relatively large family. Homozygous patient had hepatic WD and we could not detect any neurological signs in the affected patient.

In addition to 12 different mutations, 4 different sequence variations with no clinical significance were identified (table 3). These variants have been defined in previous studies as a polymorphism detected in a normal population or not altering the amino acid composition in the protein.

  

DISCUSSION

Wilson's disease is an autosomal recessive disorder, which is generally described by recurrence fulminant hepatitis and chronic liver cirrhosis (15). In most of the cases, neurological symptoms, including depression, Parkinson symptom, pseudosclerosis, distonia and akinetic rigid syndrome will be appeared in the third decade of life (16-18).

This study was our preliminary attempt to describe the mutation spectrum of seven exons of ATP7B gene among Iranian population. Besides, the data presented herein point out the prevalence of the mutations and their associated phenotype. Our study is being continued to evaluate all other exons of ATP7B gene.

We found 12 different mutations, including 4 novel disease causing mutations, in 18 individuals out of 70 referred patients. In the rest of the patients we could not detect any mutation in these exons, however, they may have mutations in other exons. Also we identified several amino acid changes or non-change mutation that were defined as the single nucleotide polymorphisms (SNP) due to their high prevalence in either healthy populations or patients. We used them as the polymorphic markers to confirm our results and perform the segregation analysis of ATP7B mutation screening in the patients.

Exon 14 has been introduced as a hot spot for mutations; we detected three different mutations in this exon, of which 2 were novel. H1069Q, located in exon14, is the most frequent mutation in ATP7B gene responsible for WD in US, and central European population (19,20). We observed this mutation in four patients all of whom had only hepatic presentation and aged 5 to 40 years. Moreover, we found a novel causative mutation, H1069N. We screened 50 unrelated individuals for those mutations and none of the tested subjects were heterozygote or homozygote. H1069N mutation has been occurred in a conserved region. Alteration happens in non-polar amino acid “histidin” to hydrophilic “asparagines” within the highly conserved SEHPL domain that is highly deleterious (21-23).

2803-2805ACG>-TG mutation was detected in a relatively large family. Homozygous patients had hepatic WD and we could not detect any neurological signs in these patients. However, with a quick glimpse to all observed phenotype presented by frameshift mutations in our patients, we can confirm the role of recognized frameshift mutations, 1639delC and 2803-2805ACG>-TG in presentation of hepatic signs of WD without any neurological symptoms.

Mutations in exon 6 of WND gene, including 1639delAT and D642H, result to WD in either heterozygous or homozygous state. We can explain this debate through the previously described predominant effect of causative mutations in exon6 of ATP7B gene. D642H was observed in 3 siblings of one family. Two sisters with heterozygous mutation had hepatic WD, and their brother who was homozygous for the so-called mutation had mixed phenotype including liver involvement in addition to neurological symptoms.

R778L is one of the most prevalent mutations in Eastern Asian countries, as reported in Chinese, Korean, Japanese, and Hungarian and leads to liver involvement in WD patients. We found this mutation in the compound heterozygous state with R969Q in one patient who had hepatic failure. It represents the mixed ethnic background of Iranian population.

Needless to say, analysis of other ATP7B exons should be carried out in order to draw a better conclusion.

In conclusion, mutations are highly distributed among the studied exons of ATP7B gene in Iranian patients with WD. It should be noted that most of the WD patients were categorized in the group with hepatic involvement, while neurological signs have been observed in 17.1% of all patients with WD.

REFERENCES

1.      Scheinberg IH, Sternlieb I. Wilson disease and idiopathic copper toxicosis. Am J Clin Nutr 1996;63(5):842S-5S.

2.      Wilson FP. A preliminary study of the biochemical relations of various lipoid substances in the liver. Biochem J 1912;6(1):100-5.

3.      Eisenbach C, Sieg O, Stremmel W, et al. Diagnostic criteria for acute liver failure due to Wilson disease. World J Gastroenterol 2007;13(11):1711-4.

4.      Das SK, Ray K. Wilson's disease: an update. Nat Clin Pract Neurol 2006;2(9):482-93.

5.      Das M, Misra UK, Kalita J. A study of clinical, MRI and multimodality evoked potentials in neurologic Wilson disease. Eur J Neurol 2007;14(5):498-504.

6.      Ferenci P, Czlonkowska A, Merle U, et al. Late-onset Wilson's disease. Gastroenterology 2007;132(4):1294-8.

7.      Nagao K. What's your diagnosis? Wilson disease (hepatolenticular degeneration) with Keyser-Fleischer ring. Pediatr Ophthalmol Strabismus 2007;44(2):84, 111.

8.      Tanner MS. Disorders of copper metabolism. In: Kelly DA, editor. Diseases of the liver and biliary system in children. Oxford: Blackwell Science. 1999;p: 167–85.

9.      Twomey PJ, Viljoen A, House IM, et al. Copper: ceruloplasmin ratio. J Clin Pathol 2007;60(4):441-2.

10.   Bull PC, Barwell JA, Hannah HT, et al. Isolation of new probes in the region of the Wilson disease locus, 13q14.2-->q14.3. Cytogenet Cell Genet 1993;64(1):12-7.

11.   Bull PC, Cox DW. Long range restriction mapping of 13q14.3 focused on the Wilson disease region.
Genomics 1993;16(3):593-8.

12.   Bull PC, Thomas GR, Rommens JM, et al. The Wilson disease gene is a putative copper transporting P-type ATPase similar to the Menkes gene. Nat Genet 1993;5(4):327-37.

13.   Hsi G, Cox DW. A comparison of the mutation spectra of Menkes disease and Wilson disease. Hum Genet 2004;114(2):165-72.  

14.   Kim EK, Yoo OJ, Song KY, et al. Identification of three novel mutations and a high frequency of the Arg778Leu mutation in Korean patients with Wilson disease. Hum Mutat 1998;11(4):275-8.

15.   Cox, Roberts EA. Wilson disease. In: Feldman M, Friedman LS, Sleisenger MH, editors. Sleisenger and Forstran’s gastrointestinal and liver disease: pathophysiology/diagnosis/management. 7th edition. Saunders, Philadelphia, 2002;p:1269–77.

16.   Ferenci P, Czlonkowska A, Merle U, et al. Late-onset Wilson's disease. Gastroenterology 2007;132(4):1294-8.

17.   Czlonkowska A, Tarnacka B, Moller JC, et al. Unified Wilson's disease rating scale; a proposal for the neurological scoring of Wilson's disease patients. Neurol Neurochir Pol 2007;41(1):1-12.

18.   Takeshita Y, Shimizu N, Yamaguchi Y, et al. Two families with Wilson disease in which siblings showed different phenotypes. J Hum Genet 2002;47:543-47.

19.   Panagiotakaki E, Tzetis M, Manolaki N, et al. Genotype-phenotype correlations for a wide spectrum of mutations in the Wilson disease gene (ATP7B). Am J Med Genet 2004;131A:168-73.

20.   Payne AS, Kelly EJ, Gitlin JD. Functional expression of the Wilson disease protein reveals mislocalization and impaired copper-dependent trafficking of the common H1069Q mutation. Proc Nat Acad Sci 1998;95:10854-59.

21.   Gromadzka G, Schmidt HH, Genschel J, et al. H1069Q mutation in ATP7B and biochemical parameters of copper metabolism and clinical manifestation of Wilson's disease. Mov Disord 2006;21(2):245-8.

22.   Tsivkovskii R, Efremov RG, Lutsenko S. The role of the invariant His-1069 in folding and function of the Wilson's disease protein, the human copper-transporting ATPase ATP7B. J Biol Chem 2003;278(15):13302-8.

23.  Morgan CT, Tsivkovskii R, Kosinsky YA, et al. The distinct functional properties of the nucleotide-binding domain of ATP7B, the human copper-transporting ATPase: analysis of the Wilson disease mutations E1064A, H1069Q, R1151H, and C1104F.J Biol Chem 2004;279(35):36363-71.

 


Receivedspan>: 15 October 2007   Accepted: 10 December 2007

Reprint or Correspondence: Mohammad Reza Zali, MD. Research Center for Gastroenterology and Liver Disease, 7th floor, Taleghani Hospital, Evin, Tehran, Iran.

E-mail: article@rigld.ir