Submit or Track your Manuscript LOG-IN

Interaction between MTHFR Polymorphisms and Maternal Age Increases the Risk of Congenital Heart Defects in Down Syndrome

PJZ_51_3_865-870

 

 

Interaction between MTHFR Polymorphisms and Maternal Age Increases the Risk of Congenital Heart Defects in Down Syndrome

S. Justin Carlus1,*, Atiyeh M Abdallah1,2, Abdulhadi H. Al-Mazroea1, Mazen Khalid Al-Harbi3 and Khalid M Al-Harbi1

1Cardiogenetic Unit, Department of Pediatrics, College of Medicine, Taibah University, Al-Madinah, Kingdom of Saudi Arabia

2West Midlands Regional Genetics Laboratory, Birmingham Women’s and Children’s Hospital, Birmingham, United Kingdom

3Al-Rayan Colleges, Al-Madinah Kingdom of Saudi Arabia

ABSTRACT

Congenital heart disease (CHD) is responsible for one-third of all congenital anomalies in newborns and is the most frequent cause of infant deaths. Several cohort studies show that down syndrome (DS) and CHD are associated, and maternal hyperhomocysteinemia is an independent risk factor for CHD. In Saudi Arabia CHD represents one of the most important health problems. Here we examined the association between methylenetetrahydrofolate reductase (MTHFR) gene polymorphisms and CHD and DS co-inheritance in patients from Al Madinah, Saudi Arabia. MTHFR rs1801133 and rs1801131 polymorphisms were genotyped in 99 CHD patients with or without DS and 126 ethnically matched controls by allelic discrimination. Of 99 patients with CHD, 26 had DS. MTHFR rs1801133 and rs1801131genotypes and alleles were not significantly different between controls and CHD patients. Further, in CHD individuals, these genotypes failed to show any significant association with DS. However, maternal age increased the risk of CHD in DS (OR=5.32; 95% CIs 1.43-19.82; p=0.013). Mantel-Haenszel analysis showed that MTHFR polymorphisms confounded the effect of maternal age CHD in DS. MTHFR polymorphisms appear to be risk factors for CHD in DS.


Article Information

Received 12 January 2019

Revised 03 February 2019

Accepted 07 February 2019

Available online 18 March 2019

Authors’ Contribution

SJC and KMAH presented the concept of the study. SJC and AMA designed methodology. AHAM and MKAH did data curation. SJC wrote the manuscript. KMAH reviewed and edited the manuscript.

Key words

Congenital heart disease, Down syndrome, Methylenetetrahydrofolate reductase, Polymorphism, Maternal age, Saudi Arabia.

DOI: http://dx.doi.org/10.17582/journal.pjz/2019.51.3.865.870

* Corresponding author: justincarlus@gmail.com

0030-9923/2019/0003-0865 $ 9.00/0

Copyright 2019 Zoological Society of Pakistan



Introduction

Congenital heart disease (CHD) comprises one-third of all congenital anomalies in newborns and is the most frequent cause of infant deaths. Long-term morbidity and mortality from CHD pose a substantial challenge to healthcare systems globally (Dolk et al., 2011). The exact etiology of CHD remains uncertain, the disease is heterogeneous, but it is believed to be multifactorial: environmental factors contribute (Gladki et al., 2015; Gorini et al., 2014; Huhta and Linask, 2013); and twin studies suggest familial aggregation of CHD (Kuo et al., 2017). Specific recurrence patterns and phenotypes also suggest that genetically-determined developmental mechanisms exert a strong influence in CHD etiology (Ellesøe et al., 2018).

Several cohort studies have established a relationship between Down syndrome (DS) and CHD (Ferencz et al., 1989; Kidd et al., 1993), with 4%-10% of all CHD cases associated with DS and 40%-60% of DS patients having CHD (de Rubens-Figueroa et al., 2003). Several lines of evidence indicate that maternal hyperhomocysteinemia is an independent risk factor for CHD (Malik et al., 2017), and recent population studies in humans have indicated that folic acid supplementation before and/or during pregnancy can decrease CHD risk (Czeizel et al., 2015; Leirgul et al., 2015; Li et al., 2013). This evidence prompted several follow-on studies examining the role of folic acid in CHD risk (Parnell and Correa, 2017; Xu et al., 2016; Feng et al., 2015).

Folates serve as cofactors for nucleotide synthesis and co-substrates of DNA methyltransferases (Stover and Field, 2011). DNA methylation plays a key role in embryonic development, and abnormally methylated genes have been detected in fetuses with both isolated and syndromic heart malformations (Serra-Juhe et al., 2015). Methylenetetrahydrofolate reductase (MTHFR; EC 1.5.1.20) is a key enzyme in folate and homocysteine (Hcy) metabolism (Bhaskar et al., 2011). The gene encoding human MTHFR is known to have two important functional polymorphisms, C677T/A222V/rs1801133 and A1298C/Glu429Ala/rs1801131, which generate a thermolabile form of MTHFR that subsequently increases total plasma Hcy and decreases serum folate levels (Goyette et al., 1994). A recent computational modeling study demonstrated that the rs1801133 polymorphism causes conformational changes in the tertiary structure of MTHFR by reducing its FAD-binding ability (Abhinand et al., 2016). However, results from studies examining the relationship between MTHFR rs1801133 and rs1801131 polymorphisms and CHD risk have been inconsistent (Zhang et al., 2018; Asim et al., 2017; Yu et al., 2017; Guo et al., 2017). We recently highlighted the high burden of DS and CHD in the Al Madinah region of Saudi Arabia (Abdulhadi et al., 2016). Here we investigated associations between MTHFR polymorphisms (rs1801133 and rs1801131) and the risk of CHD and DS co-occurrence in a Saudi population.

 

Patients and Methods

Ethical statement

The Institutional Ethics Committees of Maternity and Children Hospital and Centre for Genetics and Inherited Diseases (CGID), Taibah University, Al Madinah, Kingdom of Saudi Arabia granted ethical approval. As many of the subjects in the CHD group were minors (<18 years old), written informed consent was obtained from the parents or legal guardians of the children enrolled in this study as necessary.

Study population

Ninety-nine CHD patients with (n=26) or without (n=73) DS attended the Pediatric Cardiology Clinic at the Maternity and Children Hospital, Al Madinah, Kingdom of Saudi Arabia were enrolled in this study. Participants with mosaic, translocation of chromosome 21, non-cardiac congenital anomalies were excluded from this study. The main inclusion criteria for DS was cases had full trisomy 21 confirmed by karyotype and/or Fluorescent in situ Hybridization (FISH) for chromosome 21. CHD and CHD in DS samples were collected after checking the family history, physical and clinical examination, electrocardiograms (ECG), chest x-rays, 2D-echocardiography, Karyotypes or FISH data from the patient’s case sheet. One hundred and twenty-six healthy subjects without CHD or a family history of CHD were recruited as controls. All participants were of Saudi Arabian ethnicity.

Genotyping

Two ml of whole peripheral blood was collected from each participant after obtaining informed consent. DNA was extracted using the QIAamp DNA Mini Kit (Qiagen, Hilden, Germany). Two MTHFR SNPs [rs1801133: C___1202883_20 and rs1801131: C____850486_20] were genotyped using primers and probes purchased from Applied Biosystems, Foster City, CA. Reactions were carried out in a final volume of 5 μL (2.5 μL TaqMan PCR Master Mix, 0.125 μL TaqMan Genotyping Assay Mix, 1.375 μL nuclease free water, and 1 μL of 10 ng/μL DNA) in a 96-well optical microplate (Applied Biosystems). At least two no-template controls without any DNA were included on each plate. The PCR conditions were initial denaturation at 95°C for 10 min followed by 40 denaturation cycles (95°C for 15 s) and annealing/extension (60°C for 1 min). PCR amplification was performed using Applied Biosystems’ StepOnePlus Real-Time PCR system, and fluorescence was also measured using its

 

Table I.- The distribution of MTHFR genotypes in control and CHD subjects.

SNP

Genotype

Control (n=126)

CHD (n=99)

OR (95% CI)

p-value

rs1801133

CC

88 (69.8)

69 (69.7)

Reference

1.0*

CT

33 (26.2)

26 (26.3)

0.90 (0.48-1.75)

TT

5 (4.0)

4 (4.0)

0.85 (0.21-3.46)

CT+TT Vs. CC

38 (30.2)

30 (30.3)

0.93 (0.52-1.67)

0.810

TT Vs. CC+CT

5 (4.0)

4 (4.0)

1.01 (0.26-3.97)

0.990

MAF

17.1

17.2

HWp

0.402

0.445

rs1801131

AA

57 (45.2)

51 (51.5)

Reference

0.618*

AC

52 (41.3)

35 (35.4)

0.73 (0.40-1.33)

CC

17 (13.5)

13 (13.1)

0.82 (0.34-1.94)

AC+CC Vs. AA

69 (54.7)

48 (48.5)

1.29 (0.75-2.20)

0.360

CC Vs. AA+AC

17 (13.5)

13 (13.1)

1.03 (0.44-2.12)

0.920

MAF

34.1

30.8

HWp

0.357

0.089

OR, odds ratio; CI, confidence interval; MAF, minor allele frequency; aHWp, Hardy-Weinberg p value.

 

Table II.- The distribution of MTHFR genotypes and other risk factors among CHD patients and their association with Down syndrome.

Gene

Genotype

CHD

Univariate

OR (95% CI)

p-value

Without DS (n=73)

With DS (n=26)

rs1801133

CC

52

17

Reference

CT

18

8

2.38 (0.58-9.66)

TT

3

1

1.03 (0.08-13.24)

0.831

rs1801131

AA

38

13

Reference

AC

27

8

1.43 (0.39-5.19)

CC

8

5

4.64 (0.86-25.06)

0.542

Sex

Female

35

13

Reference

Male

38

13

1.12 (0.42-3.54)

0.707

Paternal age

< 40 y

51

9

Reference

≥ 40 y

22

17

2.45 (0.66-9.19)

0.182

Maternal age

< 35 y

61

11

Reference

≥ 35 y

12

15

5.32 (1.43-19.82)

0.013

OR, odds ratio; CI, confidence interval.

 

Sequence Detection Software (SDS) v. 2.3. A genotype call rate over 99% was considered for the analysis. As a quality control measure, 10% of samples selected at random were included for duplicate genotyping.

Statistical analyses

Allele frequencies of both polymorphisms in CHD and control groups were calculated by the gene counting method. The genotype distribution for each polymorphism was evaluated for Hardy–Weinberg equilibrium. The strength of the association between MTHFR gene polymorphisms and CHD was evaluated using the χ2 test and odds ratios (OR) with 95% confidence intervals (CI). The influence of different genotypes on the relationship between DS and maternal age and their interaction was examined using the Mantel-Haenszel stratified analysis. All statistical analyses were performed in SPSS v. 14.0 (IBM Statistics, Chicago, IL).

 

Results

Of 99 patients with CHD, 26 (26.3%) had DS. Among DS patients, atrioventricular septal defect (AVSD) was the most common cardiac defect followed by ventricular septal defect (VSD) and atrial septal defect (ASD). The distributions of MTHFR rs1801133 and rs1801131 genotypes and alleles in CHD and control groups are shown in Table I. The MTHFR rs1801133 (p=0.402) and rs1801131 (p=0.357) genotype distributions in the control group followed Hardy­Weinberg equilibrium (p=0.359).

Genotypes and alleles were not statistically different between controls and individuals with CHD (Table I). MTHFR rs1801133 and rs1801131 polymorphisms were not associated with CHD risk (Table I) and, in CHD individuals, MTHFR rs1801133 and rs1801131 genotypes failed to show any significant association with DS (Table II).

Univariate analysis showed that increased maternal age (≥35 years) contributed to CHD risk in DS patients (OR: 5.32, 95% CI: 1.43-19.82; p=0.013), while increased paternal age (≥40 years) had no effect on CHD in DS patients (OR: 2.45, 95% CI: 0.66-9.19; p=0.182) (Table II). Significant heterogeneity in the effect of maternal age on DS was observed among different genotypes of MTHFR rs1801133 and rs1801131 polymorphisms. The M-H combined OR for maternal age and rs1801133 (p<0.001) was 6.33 and for maternal age and rs1801131 was 7.42 (p<0.001) (Table III).

 

Table III.- Association between CHD with or without down syndrome and maternal age stratified by MTHFR genotypes.

Gene

Genotype

OR (95% CI)

p-Value*

Maternal age

rs1801133

CC

11.78 (3.29-42.14)

<0.001

CT

3.50 (0.59-20.68)

TT

-

M-H combined

6.33 (2.37-16.93)

rs1801131

AA

9.96 (2.37 -41.86)

<0.001

AC

5.75 (1.00-32.95)

CC

4.47 (0.30-73.38)

M-H combined

7.42 (2.67-20.64)

CHD, congenital heart disease; M-H, Mantel-Haenszel.*Homogeneity test p value.

 

Discussion

The pathogenesis of CHD is complex and remains poorly understood due to its multifactorial etiology. Individuals with DS show several common cardiac malformations such as VSDs, atrioventricular canals, and the tetralogy of Fallot (Tandon and Edwards, 1973). As folate serves as a DNA methyltransferase co-substrate, reduced folate causes hypomethylation and consequently abnormal segregation of chromosomes or chromosome nondisjunction (Fenech, 2011; James et al., 1999; Blom and Smulders, 2011). One common factor in these folate abnormalities is MTHFR rs1801133 polymorphisms, with decreased enzyme activity causing impaired re-methylation of Hcy to methionine and subsequent hyperhomocysteinemia (Yigit et al., 2013).

However, the association between maternal genetic polymorphisms in folate metabolism genes and DS risk is still controversial (Yang et al., 2013; Brandalize et al., 2009, 2010). Significantly elevated Hcy levels in mothers of children with CHD suggest that maternal hyperhomocysteinemia is an independent risk factor for CHD (Malik et al., 2017; Lu et al., 2011; Verkleij-Hagoort et al., 2007). Due to the high prevalence of CHDs in neonates born with DS and DS survivors in Al Madinah, CHDs are considered as an important health problem in Saudi Arabia, a problem further exacerbated by widespread consanguinity in this region (Abdulhadi et al., 2016). Thus, maternal supplementation with folic acid is likely to be associated with a reduced risk of CHD in DS. Given this perspective and to inform management, we considered the association between MTHFR polymorphisms and CHD in DS susceptibility in a Saudi population.

We did not identify any significant association between MTHFR polymorphisms and CHD risk. Both polymorphisms had a confounding effect on the relationship between CHD in DS and maternal age. The presence of the maternal MTHFR rs1801133 “T” allele was associated with an increased risk of DS in a Brazilian population (Brandalize et al., 2009), while another study of a Croatian population reported no association between maternal MTHFR polymorphisms and CHD in DS (Bozovic et al., 2011; Elsayed et al., 2013). MTHFR rs1801131 was over-transmitted in patients with CHD in DS ASDs (Locke et al., 2010). A meta-analysis showed that MTHFR rs1801131 polymorphisms reduced the risk of CHD in patients without DS (Yu et al., 2017), while another suggested that the maternal MTHFR rs1801133 “T” allele was a risk factor for DS-affected pregnancies (Rai et al., 2014). Finally, a recent study showed that the MTHFR gene was twice as likely to be promoter methylated in mothers of children with DS with CHD than mothers of children with DS without CHD (Asim et al., 2017).

The following strengths and limitations must be taken into consideration when interpreting our results. Significant strengths include the inclusion of subjects with CHD diagnosed by echocardiography and clinical cardiac examination and DS by karyotype or FISH for chromosome 21. However, the study also has several limitations: plasma Hcy levels were not determined and correlated with DS-related CHD or MTHFR variants, and maternal MTHFR gene polymorphisms were not tested. Further, the study is relatively small and retrospective, which may impact on the validity of the results.

 

Conclusion

In conclusion, polymorphisms in MTHFR may interact with other confounding variables and contribute to the increased risk of CHD in DS.

 

Acknowledgements

This project was funded by King Abdulaziz City for Science and Technology (KACST), award numbers (AT-34-369).

 

Statement of conflict of interests

The authors have no conflicts of interest.

 

References

Abdulhadi, H.A., Sahar, A.F.H., Lama, M.E.S., Justin, C., Yousef, A. and Khalid, M.A., 2016. Congenital heart disease in Saudi Arabia: the role of molecular genetics with a focus on down syndrome. Aust. J. Basic appl. Sci., 10: 98-109.

Abhinand, P.A., Shaikh, F., Bhakat, S., Radadiya, A., Bhaskar, L.V., Shah, A. and Ragunath, P.K., 2016. Insights on the structural perturbations in human MTHFR Ala222Val mutant by protein modeling and molecular dynamics. J. Biomol. Struct. Dynam., 34: 892-905. https://doi.org/10.1080/07391102.2015.1057866

Asim, A., Agarwal, S., Panigrahi, I., Saiyed, N. and Bakshi, S., 2017. MTHFR promoter hypermethylation may lead to congenital heart defects in down syndrome. Intractable Rare Dis. Res., 6: 295-298. https://doi.org/10.5582/irdr.2017.01068

Bhaskar, L.V., Murthy, J. and Venkatesh Babu, G., 2011. Polymorphisms in genes involved in folate metabolism and orofacial clefts. Arch. Oral Biol., 56: 723-737. https://doi.org/10.1016/j.archoralbio.2011.01.007

Blom, H.J. and Smulders, Y., 2011. Overview of homocysteine and folate metabolism. With special references to cardiovascular disease and neural tube defects. J. Inherit. Metab. Dis., 34: 75-81. https://doi.org/10.1007/s10545-010-9177-4

Bozovic, I.B., Vranekovic, J., Cizmarevic, N.S., Mahulja-Stamenkovic, V., Prpic, I. and Brajenovic-Milic, B., 2011. MTHFR C677T and A1298C polymorphisms as a risk factor for congenital heart defects in down syndrome. Pediat. Int. Off. J. Japan Pediat. Soc., 53: 546-550. https://doi.org/10.1111/j.1442-200X.2010.03310.x

Brandalize, A.P., Bandinelli, E., dos Santos, P.A., Roisenberg, I. and Schuler-Faccini, L., 2009. Evaluation of C677T and A1298C polymorphisms of the MTHFR gene as maternal risk factors for down syndrome and congenital heart defects. Am. J. med. Genet. A, 149A: 2080-2087. https://doi.org/10.1002/ajmg.a.32989

Brandalize, A.P., Bandinelli, E., dos Santos, P.A. and Schuler-Faccini, L., 2010. Maternal gene polymorphisms involved in folate metabolism as risk factors for down syndrome offspring in Southern Brazil. Dis. Mark., 29: 95-101. https://doi.org/10.1155/2010/250324

Czeizel, A.E., Vereczkey, A. and Szabo, I., 2015. Folic acid in pregnant women associated with reduced prevalence of severe congenital heart defects in their children: A national population-based case-control study. Eur. J. Obstet. Gynecol.Reprod. Biol., 193: 34-39. https://doi.org/10.1016/j.ejogrb.2015.06.024

de Rubens-Figueroa, J., del Pozzo-Magana, B., Pablos-Hach, J.L., Calderon-Jimenez, C. and Castrejon-Urbina, R., 2003. Heart malformations in children with Down syndrome. Rev. Esp. Cardiol., 56: 894-899.

Dolk, H., Loane, M. and Garne, E., 2011. Congenital heart defects in Europe: Prevalence and perinatal mortality, 2000 to 2005. Circulation, 123: 841-849. https://doi.org/10.1161/CIRCULATIONAHA.110.958405

Ellesøe, S.G., Workman, C.T., Bouvagnet, P., Loffredo, C.A., McBride, K.L., Hinton, R.B., van Engelen, K., Gertsen, E.C., Mulder, B.J.M., Postma, A.V., Anderson, R.H., Hjortdal, V.E., Brunak, S. and Larsen, L.A., 2018. Familial co-occurrence of congenital heart defects follows distinct patterns. Eur. Heart J., 39: 1015-1022. https://doi.org/10.1093/eurheartj/ehx314

Elsayed, G.M., Elsayed, S.M. and Ezz-Elarab, S.S., 2013. Maternal MTHFR C677T genotype and septal defects in offspring with Down syndrome: A pilot study. Egyptian J. med. Hum. Genet., 15: 39-44. https://doi.org/10.1016/j.ejmhg.2013.09.003

Fenech, M., 2011. Micronuclei and their association with sperm abnormalities, infertility, pregnancy loss, pre-eclampsia and intra-uterine growth restriction in humans. Mutagenesis, 26: 63-67. https://doi.org/10.1093/mutage/geq084

Feng, Y., Wang, S., Chen, R., Tong, X., Wu, Z. and Mo, X., 2015. Maternal folic acid supplementation and the risk of congenital heart defects in offspring: A meta-analysis of epidemiological observational studies. Scient. Rep., 5: 8506. https://doi.org/10.1038/srep08506

Ferencz, C., Neill, C.A., Boughman, J.A., Rubin, J.D., Brenner, J.I. and Perry, L.W., 1989. Congenital cardiovascular malformations associated with chromosome abnormalities: An epidemiologic study. J. Pediatr., 114: 79-86. https://doi.org/10.1016/S0022-3476(89)80605-5

Gladki, M.M., Skladzien, T. and Skalski, J.H., 2015. The impact of environmental factors on the occurrence of congenital heart disease in the form of hypoplastic left heart syndrome. Kardiochir Torakochirur. Pol., 12: 204-207.

Gorini, F., Chiappa, E., Gargani, L. and Picano, E., 2014. Potential effects of environmental chemical contamination in congenital heart disease. Pediatr. Cardiol., 35: 559-568. https://doi.org/10.1007/s00246-014-0870-1

Goyette, P., Sumner, J.S., Milos, R., Duncan, A.M., Rosenblatt, D.S., Matthews, R.G. and Rozen, R., 1994. Human methylenetetrahydrofolate reductase: Isolation of cDNA, mapping and mutation identification. Nat. Genet., 7: 195-200. https://doi.org/10.1038/ng0694-195

Guo, Q.N., Wang, H.D., Tie, L.Z., Li, T., Xiao, H., Long, J.G. and Liao, S.X., 2017. Parental genetic variants, MTHFR 677C>T and MTRR 66A>G, associated differently with fetal congenital heart defect. Biomed. Res. Int., 2017: 3043476. https://doi.org/10.1155/2017/3043476

Huhta, J. and Linask, K.K., 2013. Environmental origins of congenital heart disease: The heart-placenta connection. Semin. Fetal Neonatal. Med., 18: 245-250. https://doi.org/10.1016/j.siny.2013.05.003

James, S.J., Pogribna, M., Pogribny, I.P., Melnyk, S., Hine, R.J., Gibson, J.B., Yi, P., Tafoya, D.L., Swenson, D.H., Wilson, V.L. and Gaylor, D.W., 1999. Abnormal folate metabolism and mutation in the methylenetetrahydrofolate reductase gene may be maternal risk factors for down syndrome. Am. J. clin. Nutr., 70: 495-501. https://doi.org/10.1093/ajcn/70.4.495

Kidd, S.A., Lancaster, P.A. and McCredie, R.M., 1993. The incidence of congenital heart defects in the first year of life. J. Paediat. Child Hlth., 29: 344-349. https://doi.org/10.1111/j.1440-1754.1993.tb00531.x

Kuo, C.F., Lin, Y.S., Chang, S.H., Chou, I.J., Luo, S.F., See, L.C., Yu, K.H., Huang, L.S. and Chu, P.H., 2017. Familial aggregation and heritability of congenital heart defects. Circulation J., 82: 232-238. https://doi.org/10.1253/circj.CJ-17-0250

Leirgul, E., Gildestad, T., Nilsen, R.M., Fomina, T., Brodwall, K., Greve, G., Vollset, S.E., Holmstrom, H., Tell, G.S. and Oyen, N., 2015. Periconceptional folic acid supplementation and infant risk of congenital heart defects in Norway 1999-2009. Paediat. Perinat. Epidemiol., 29: 391-400. https://doi.org/10.1111/ppe.12212

Li, X., Li, S., Mu, D., Liu, Z., Li, Y., Lin, Y., Chen, X., You, F., Li, N., Deng, K., Deng, Y., Wang, Y. and Zhu, J., 2013. The association between periconceptional folic acid supplementation and congenital heart defects: A case-control study in China. Prevent. Med., 56: 385-389. https://doi.org/10.1016/j.ypmed.2013.02.019

Locke, A.E., Dooley, K.J., Tinker, S.W., Cheong, S.Y., Feingold, E., Allen, E.G., Freeman, S.B., Torfs, C.P., Cua, C.L., Epstein, M.P., Wu, M.C., Lin, X., Capone, G., Sherman, S.L. and Bean, L.J., 2010. Variation in folate pathway genes contributes to risk of congenital heart defects among individuals with down syndrome. Genet. Epidemiol., 34: 613-623. https://doi.org/10.1002/gepi.20518

Lu, Y., Wang, H. and Wang, X., 2011. Relationship of hyperhomocysteinemia in pregnant rats and congenital heart defects in the newborn rats. Zhong Nan Da Xue Xue Bao Yi Xue Ban, 36: 68-73.

Malik, R.A., Lone, M.R., Ahmed, A., Koul, K.A. and Malla, R.R., 2017. Maternal hyperhomocysteinemia and congenital heart defects: A prospective case control study in Indian population. Indian Heart J., 69: 17-19. https://doi.org/10.1016/j.ihj.2016.07.014

Parnell, A.S. and Correa, A., 2017. Analyses of trends in prevalence of congenital heart defects and folic acid supplementation. J. Thorac. Dis., 9: 495-500. https://doi.org/10.21037/jtd.2017.03.16

Rai, V., Yadav, U., Kumar, P., Yadav, S.K. and Mishra, O.P., 2014. Maternal methylenetetrahydrofolate reductase C677T polymorphism and down syndrome risk: A meta-analysis from 34 studies. PLoS One, 9: e108552. https://doi.org/10.1371/journal.pone.0108552

Serra-Juhe, C., Cusco, I., Homs, A., Flores, R., Toran, N. and Perez-Jurado, L.A., 2015. DNA methylation abnormalities in congenital heart disease. Epigenetics, 10: 167-177. https://doi.org/10.1080/15592294.2014.998536

Stover, P.J. and Field, M.S., 2011. Trafficking of intracellular folates. Adv. Nutr., 2: 325-331. https://doi.org/10.3945/an.111.000596

Tandon, R. and Edwards, J.E., 1973. Cardiac malformations associated with down’s syndrome. Circulation, 47: 1349-1355. https://doi.org/10.1161/01.CIR.47.6.1349

Verkleij-Hagoort, A., Bliek, J., Sayed-Tabatabaei, F., Ursem, N., Steegers, E. and Steegers-Theunissen, R., 2007. Hyperhomocysteinemia and MTHFR polymorphisms in association with orofacial clefts and congenital heart defects: a meta-analysis. Am. J. med. Genet. A, 143A: 952-960. https://doi.org/10.1002/ajmg.a.31684

Xu, A., Cao, X., Lu, Y., Li, H., Zhu, Q., Chen, X., Jiang, H. and Li, X., 2016. A meta-analysis of the relationship between maternal folic acid supplementation and the risk of congenital heart defects. Int. Heart J., 57: 725-728. https://doi.org/10.1536/ihj.16-054

Yang, M., Gong, T., Lin, X., Qi, L., Guo, Y., Cao, Z., Shen, M. and Du, Y., 2013. Maternal gene polymorphisms involved in folate metabolism and the risk of having a down syndrome offspring: A meta-analysis. Mutagenesis, 28: 661-671. https://doi.org/10.1093/mutage/get045

Yigit, S., Karakus, N. and Inanir, A., 2013. Association of MTHFR gene C677T mutation with diabetic peripheral neuropathy and diabetic retinopathy. Mol. Vis., 19: 1626-1630.

Yu, D., Zhuang, Z., Wen, Z., Zang, X. and Mo, X., 2017. MTHFR A1298C polymorphisms reduce the risk of congenital heart defects: A meta-analysis from 16 case-control studies. Italian J. Pediat., 43: 108. https://doi.org/10.1186/s13052-017-0425-1

Zhang, R., Huo, C., Wang, X., Dang, B., Mu, Y. and Wang, Y., 2018. Two common MTHFR gene polymorphisms (C677T and A1298C) and fetal congenital heart disease risk: An Updated meta-analysis with trial sequential analysis. Cell Physiol. Biochem., 45: 2483-2496. https://doi.org/10.1159/000488267

To share on other social networks, click on any share button. What are these?

Pakistan Journal of Zoology

April

Pakistan J. Zool., Vol. 56, Iss. 2, pp. 503-1000

Featuring

Click here for more

Subscribe Today

Receive free updates on new articles, opportunities and benefits


Subscribe Unsubscribe