Skip to main content

Update on the genetic and epigenetic etiology of gestational diabetes mellitus: a review

Abstract

Background

Many studies have been conducted on the genetic and epigenetic etiology of gestational diabetes mellitus (GDM) in the last two decades because of the disease’s increasing prevalence and role in global diabetes mellitus (DM) explosion. An update on the genetic and epigenetic etiology of GDM then becomes imperative to better understand and stem the rising incidence of the disease. This review, therefore, articulated GDM candidate genes and their pathophysiology for the awareness of stakeholders.

Main body (genetic and epigenetic etiology, GDM)

The search discovered 83 GDM candidate genes, of which TCF7L2, MTNR1B, CDKAL1, IRS1, and KCNQ1 are the most prevalent. Certain polymorphisms of these genes can modulate beta-cell dysfunction, adiposity, obesity, and insulin resistance through several mechanisms. Environmental triggers such as diets, pollutants, and microbes may also cause epigenetic changes in these genes, resulting in a loss of insulin-boosting and glucose metabolism functions. Early detection and adequate management may resolve the condition after delivery; otherwise, it will progress to maternal type 2 diabetes mellitus (T2DM) and fetal configuration to future obesity and DM. This shows that GDM is a strong risk factor for T2DM and, in rare cases, type 1 diabetes mellitus (T1DM) and maturity-onset diabetes of the young (MODY). This further shows that GDM significantly contributes to the rising incidence and burden of DM worldwide and its prevention may reverse the trend.

Conclusion

Mutations and epigenetic changes in certain genes are strong risk factors for GDM. For affected individuals with such etiologies, medical practitioners should formulate drugs and treatment procedures that target these genes and their pathophysiology.

Background

Pregnant women develop insulin resistance at certain stages owing to increased placenta hormones, but most women overcome this condition by up-regulating insulin production through beta cell expansion [1]. Gestational diabetes mellitus (GDM) begins when a pregnant female does not make the extra insulin needed to normalize blood glucose during the second or third trimester of pregnancy [1]. Sometimes the glucose intolerance may be present before pregnancy, but not diagnosed [2, 3]. Uncontrolled GDM can cause high blood pressure, type 2 diabetes mellitus (T2DM), and increased risks of vascular diseases in pregnant women [4, 5]. Intrauterine exposure to high blood glucose may program the offspring to develop diabetes or obesity later in life [6]. It may also cause macrosomia, birth defects, preterm birth, and developmental delay [7,8,9].

GDM is the most common metabolic condition during pregnancy [10] with a global incidence rate between 1 and 28 % [11]. In 2017, GDM affected about 204 million women worldwide with a projection to increase to 308 million by 2045, mostly in developing countries [12]. Though most times the glucose intolerance normalizes soon after delivery, women with GDM have a high risk of developing T2DM later in life [13]. Compared to women with normal glucose tolerance, women with GDM are at least seven times more predisposed to T2DM [14]. Moreover, almost half of pregnant females with GDM will develop diabetes in a decade [14]. Offspring of women with GDM are also 8 times more prone to diabetes or pre-diabetes [15]. These show that GDM contributes immensely to the alarming incidence of diabetes worldwide [16]. Diabetes affected about 451 million people in 2017, of which 5 million died and USD 850 billion was spent on healthcare expenditure [17]. Considering the impact of GDM, the reduction of its prevalence and effective management of the affected will go a long way in stemming the incidence and burden of diabetes. However, to achieve a reduced prevalence of GDM, a proper understanding of its etiology is necessary. Fortunately, improved biological techniques in the last two decades have led to more understanding of the genetic and epigenetic etiology of the disease, thus an update becomes necessary. This review therefore articulated current findings on the genetic and epigenetic etiology of GDM.

Methods

Databases searched

An in-depth search of PubMed, Scopus, SpringerLink, Google Scholar, and ResearchGate databases was performed for relevant research articles on GDM.

Search terms

Some search terms used to retrieve articles are gestational diabetes mellitus, hyperglycemia, insulin resistance, obesity, glucose metabolism, beta-cell dysfunction, and gestational diabetes genes. Other search terms used include glucose insensitivity, epigenetics of diabetes mellitus, gestational diabetes testing and cost-effectiveness, and the prevalence of gestational diabetes.

Article inclusion criteria

Inclusion criteria include the following:

Research published in the English language.

Research that focused on GDM.

Studies that focused on the genetic and epigenetic etiology of GDM.

Articles that centered on GDM testing and cost-effectiveness.

Studies published between 2000 till date.

Article exclusion criteria

Exclusion criteria include the following:

Studies that are not available in English language.

Studies with only abstract available.

Research that described GDM, but with no clear genetic and epigenetic mechanisms

Studies published before the year 2000.

Results

Genetic etiology in GDM

The search found that mutations in some genes, or their variants, may interact with one another and environmental triggers to cause GDM (Fig. 1). The genetic etiology of GDM overlaps with T2DM, as most of the GDM candidate genes also predispose humans to T2DM. This explains the common pathophysiology of GDM and T2DM as both express beta-cell dysfunction and abnormal glucose metabolism [18]. Contrary to some reports, GDM also shares common pathophysiology with type 1 diabetes mellitus (T1DM) and maturity-onset diabetes of the young (MODY), but less than 10 % of GDM patients show these associations [18].

Fig. 1
figure 1

Genetic etiology of gestational diabetes mellitus

Though many genes reportedly showed an association with obesity, insulin resistance, and beta-cell dysfunction, the present study discovered only 83 genes with a clear GDM pathophysiology and are presented in Table 1.

Table 1 GDM predisposing genes showing locations and phathophysiology

Most frequent GDM candidate genes

The list of GDM candidate genes is inexhaustible as more genes are continually discovered; however, certain genes are most often linked with the disease. Table 2 shows the most frequent GDM candidate genes and their variants in various ethnic groups.

Table 2 Most prevalent gestational diabetes mellitus genes across countries, ethnicity, and race in the world

From Table 2, we used a pie chart (Fig. 2) to express the percentage occurrence of each gene based on ethnicity. TCF7L2 gene was the most frequent having present in 17 % of the ethnics, followed by MTNR1B with 15 %, CDKAL1 10 %, KCNQ1 10 %, and IRS1 10 %. Some other genes are not widespread, but are often found in certain ethnics or regions. These genes include ZRANB3, found in Africa [96]; ABCC8 found among Finnish [119]; Chemerin, found among Iranians [120]; and INS, found among the Greeks [121]. These genes can be used to develop a genetic testing guideline to predict the likelihood of GDM or determine its genetic and epigenetic etiology. This is important because there is no genetic testing procedure yet for GDM, partly because the condition is multifactorial in which several genes interact with environmental triggers to cause the disease. Thus, a mutation in a single gene may not explain full susceptibility to GDM and testing for all the candidate genes will be expensive and cumbersome. The low prevalence of GDM in the past also contributes to the lack of interest in developing a genetic testing guideline for the disease.

Fig. 2
figure 2

Percentage prevalence of most implicated GDM genes from data extracted

Epigenetic etiology in GDM

Epigenetics refers to the study of heritable changes in biological processes caused by modification of chemical tags on DNA such as methyl and ethyl groups [122]. These modifications are mediated by some mechanisms, including DNA methylation, histone modification, and microRNA expression [122]. Epigenetic mechanisms play important roles in several cellular activities, but certain environmental triggers can reprogram the epigenome, resulting in disease pathologies [123]. In particular, epigenetic mechanisms regulate several genes that maintain beta-cell morphology, proliferation, and functions, thus implies that epigenetic modification may disrupt insulin secretion and sensitivity, causing metabolic diseases, including GDM [124, 125]. Epigenetic changes are expressed in both somatic and gamete cells, and thus can be transmitted from generation to generation [126].

Studies have reported many instances of epigenetic modifications involving insulin synthesis and glucose metabolism in GDM. For instance, histone under-acetylation and over-methylation in the promoter region of the PDX1 gene reduce the insulin-boosting function of the gene [127]. Also, in a study that examined the methylation of the IL-10 gene among pregnant women, hypo-methylation of maternal blood cells and elevated plasma IL-10 levels were noticed in women with GDM [128]. In another study that compared the miRNA profiles of some diabetic pregnant rats with nondiabetic, repression of miR-338 and overexpression of miR-451 were associated with reduced β-cell mass in the diabetic [129]. In vitro upregulation of miR-451 and repression of miR-338 in the same study increased β-cell mass, leading to improved glucose metabolism [129]. In a study that investigated the miRNA expressions of maternal and fetal blood cells of some pregnant women, 29 miRNAs were upregulated in individuals with GDM [130]. Of these miRNAs, miRNA-340 was confirmed to downregulate the expression of the PAIP1 gene [130]. In vitro normalization of the miRNA-340 expression of the diabetic mothers increased insulin production [130].

Environmental triggers of genetic and epigenetic etiology of GDM

Both the genetic and epigenetic etiology of GDM are mediated by certain environmental triggers that change gene functions. The genetic triggers mutate the genes, while the epigenetic triggers affect the chemical tags on the DNA without affecting the nucleotide sequence (Fig. 3). Among the environmental triggers are pollution and microbial exposures, whose GDM modulatory roles have been established by several studies. In a study that monitored the effects of air pollution among pregnant Southern Californians, prepregnancy exposures to nitrogen dioxide (NO2), particulate matter (PM2.5 and PM10), and dioxin were related to GDM [131]. Nitrogen dioxide and particulate matter can cause oxidative stress, overexpression of proinflammatory cytokines, and endothelial dysfunction, resulting in increased insulin resistance [132]. Dioxin compounds can interact with peroxisome proliferator-activated receptor-γ (PPARG), disrupting insulin signaling pathways and resulting in insulin resistance and abnormal glucose metabolism [133]. Exposure to pathogenic microbial organisms may disrupt the gut microbiota and compromise the immune system, leading to metabolic disorders and GDM. Vu et al. [134] demonstrated in rabbits that a toxin produced by Staphylococcus aureus may interact with fat cells and the immune system, resulting in inflammation, insulin resistance and glucose intolerance [134]. In a study of the microbiota of some pregnant women, individuals with GDM showed gut microbiota imbalance containing majorly the phylum Actinobacteria and the genus Collinsella, Rothia and Desulfovibrio [135]. A balanced gut microbiota is necessary for optimum metabolism and the immune system. Aside from microbial infection, other environmental factors that may disrupt gut microbiota include certain diseases, oral microbiome, diets, and antibiotic use, among others [136].

Fig. 3
figure 3

Epigenetic etiology of gestational diabetes mellitus

Lifestyles such as short sleep, poor nutritional choices, advanced age, and physical inactivity are some environmental triggers that may predispose humans to GDM. Short hour sleep at night can increase body fat accumulation, reduce glucose metabolism, and predispose to T2DM and GDM. In a study that determines the frequency of GDM among sleep-deprived 668 Singaporeans, 131, representing 19 %, were diagnosed with GDM of which 27.3 % sleep less than 6 hours a night, while 16.8 % sleep between 7-8 hours [137]. Poor nutrition, such as energy-dense western diets may cause overweight and obesity, disrupting insulin signaling pathways and insulin sensitivity. Saturated fats can disrupt insulin signaling, induce inflammation and endothelial dysfunction, resulting in GDM. In a study that evaluated the effect of dietary patterns among pregnant Chinese women, diets containing high protein and low starch were associated with a reduced risk of GDM [138]. Energy-dense diets are deficient in betaine, which is a methyl donor for methylating important biological processes and as well a substrate of methionine metabolism [139]. Diets low in betaine may induce abnormal methylation of some genes involved in insulin synthesis and glucose metabolism. Advanced age may also predispose pregnant women to GDM because mitochondrial functions decline with age, leading to reduced metabolic activities and an increased body mass index [140]. Aging changes the epigenetic pattern, affecting the expression of some genes involved in glucose metabolism, particularly the COX7A1 gene in the respiratory chain [141]. In a study of 1688 women in northwest London who developed GDM, advanced maternal age was linked to GDM [142]. Studies that demonstrate the role of physical inactivity in the pathogenesis of GDM are scarce. However, a systematic and meta-analysis by Ming et al. [143] shows that physical activity during pregnancy can decrease the occurrence of GDM, suggesting that lack of exercise is a risk factor. Some mechanisms through which inactivity mediates diseases include mitochondrial dysfunction, changes in the composition of muscles, and insulin resistance, among others [144]. Physical inactivity influences the epigenome negatively, affecting several generations [145].

GDM testing, efficacy, and cost-effectiveness

Early detection of GDM is important to prevent its short- and long-term effects, especially the maternal progression to T2DM and fetal programming to DM later in life. Certain features such as BM1 above 30 kg/m2 as well as previous GDM, baby birth weight of 4.5 kg or above, and macrosomia suggest a need for a GDM test [146]. Pregnant women with a family history of DM and ethnic groups with a high prevalence of DM such as Asian, Black, African-Caribbean or Middle Eastern should also consider the test [146, 147]. As stated earlier, there is no genetic testing procedure yet for GDM, however, two tests, namely glucose challenge test (GCT) and oral glucose tolerance test (OGTT) are frequently conducted at 24-28 weeks of pregnancy to diagnose GDM. The two tests can be done in succession known as 2-step screening, or OGTT alone can be done called 1-step screening.

In the 2-step screening, the GCT (otherwise known as a glucose screening test) is done first and entails testing the blood glucose one hour after drinking a sweet substance without fasting. If the blood glucose is 140 mg/dL (7.8 mmol/L) or higher, then an OGTT is necessary [148]. The OGTT measures blood glucose after 8-hour fasting, after which a glucose substance (75 g) is taken and blood glucose re-measured after 1, 2 and 3 hours. High blood glucose levels at any two or more of the blood test times suggest GDM [148]. Though the pathophysiology of GDM are similar with T2DM, a GCT of 200 mg/dL or more could indicate T2DM [148].

Relatively recently, serum levels of C-reactive protein (CRP) as well as glycated hemoglobin (HbA1c) and random blood sugar (RBS) are used as screening tools for GDM in the first trimester. CRP concentrations of about 6 mg/L or higher in undiluted serum samples are considered positive for GDM [149]. According to the International Association of the Diabetes and Pregnancy Study Group (IADPSG), the cutoff level of HbA1c is 6.5 % and RBS is 11.1 mmol (200mg/dL) [150].

The cost-effectiveness of GDM testing is controversial because it depends on the region, race, screening tools, and methods. A systematic review by Fitria [151] reported that GDM testing and controlling is not effective in high-income countries. This could be due to the low prevalence of the disease in the region and the all-encompassing health care system. However, GDM testing could be worthwhile in countries with a high prevalence of GDM and nations with a poor healthcare system. For example, Marseille et al. [152] reported the cost-effectiveness of GDM testing using a devised model in Israel and India, which are known for high GDM incidence rates. The 2-step GTT is often recommended, however, a systematic review and meta-analysis by Saconne et al [153] showed no significant cost-effective difference between the two methods. The 2-step screening is also time-consuming and inconvenient, which may put off some patients [150, 154]. Glycated hemoglobin and RBS are simple GDM screening tools and are gaining acceptance worldwide, however, more awareness and understanding of the tools are necessary.

Conclusion

Several articles reviewed showed that mutation and epigenetic modifications in certain genes can predispose humans to GDM. Most of the GDM candidate genes identified have also been implicated in the pathogenesis of T2DM, and both diseases share a common pathophysiology. The two metabolic disorders expressed oxidative stress-induced beta-cell dysfunction and insulin resistance through adiposity and obesity. One major difference between GDM and T2DM is that it resolves most times after delivery, however, it may progress to T2DM if not checked. This shows that GDM is a strong risk factor for T2DM, thus, its detection and management may reduce the prevalence of DM worldwide. Of the GDM candidate genes identified, the variants of TCF7L2, MTNR1B, CDKAL1, IRS1 and KCNQ1 are the most widespread, while some others are confined to certain ethnic groups. A genetic testing procedure can be developed around these genes to predict the likelihood of GDM or determine its genetic and epigenetic etiology. This will go a long way in stemming the incidence of DM worldwide.

Availability of data and materials

Not applicable.

Abbreviations

CRP:

C-reactive protein

DM:

Diabetes mellitus

GCT:

Glucose challenge test

GDM:

Gestational diabetes mellitus

HbA1C:

Glycated hemoglobin

IADPSG:

International Association of the Diabetes and Pregnancy Study Group

MODY:

Maturity onset diabetes of the young

NO2 :

Nitrogen dioxide

OGTT:

Oral glucose tolerance test

PM2.5:

Particulate matter size 2.5

PM10:

Particulate matter size 10

RBS:

Random blood sugar

T1DM:

Type 1 diabetes mellitus

T2DM:

Type 2 diabetes mellitus

References

  1. National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Symptoms & Causes of Gestational Diabetes. https://www.niddk.nih.gov/health-information/diabetes/overview/what-is-diabetes/gestational/symptoms-causes

  2. Fernández-Morera JL, Rodríguez-Rodero S, Menéndez-Torre E et al (2010) The possible role of epigenetics in gestational diabetes: cause, consequence, or both. Obstet Gynecol Int. 2010:605163. https://doi.org/10.1155/2010/605163

    Article  PubMed  PubMed Central  Google Scholar 

  3. American Diabetes Association (ADA). Standards of medical care in diabetes- 2018”. Diabetes Care. 2018;41(Suppl 1):S13–S27, doi: https://doi.org/10.2337/dc18-S002

  4. Rayanagoudar G, Hashi AA, Zamora J et al (2016) Quantification of the type 2 diabetes risk in women with gestational diabetes: a systematic review and meta-analysis of 95,750 women. Diabetologia 59:1403–1411. https://doi.org/10.1007/s00125-016-3927-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Burlina S, Dalfrà MG, Lapolla A. Clinical and biochemical approach to predicting post-pregnancy metabolic decompensation. Diabetes Res Clin Pract, 2018;145:178–183. DOI:10.1016/j.diabres.2018.02.035. DOI: https://doi.org/10.1016/j.diabres.2018.02.035

    Article  CAS  PubMed  Google Scholar 

  6. Yessoufou A, Moutairou K. Maternal Diabetes in Pregnancy: Early and Long-Term Outcomes on the Offspring and the Concept of “Metabolic Memory. Experimental Diabetes Research, 2011; 2011: Article ID 218598. https://doi.org/10.1155/2011/218598.

    Article  Google Scholar 

  7. Metzger BE (2009) The HAPO Study Cooperative Research Group. Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study. Associations with neonatal anthropometrics. Diabetes 58:453–459. https://doi.org/10.2337/db08-1112

    Article  CAS  Google Scholar 

  8. Freinkel N (1980) Banting Lecture 1980: of pregnancy and progeny. Diabetes 29:1023–1035

    Article  CAS  PubMed  Google Scholar 

  9. Farahvar S, Walfisch A, Sheiner E (2019) Gestational diabetes risk factors and long-term consequences for both mother and offspring: a literature review. Expert Rev Endocrinol Metab 14(1):63–74. https://doi.org/10.1080/17446651.2018.1476135

    Article  CAS  PubMed  Google Scholar 

  10. Franzago M, Fraticelli F, Stuppia L, Vitacolonna E (2019) Nutrigenetics, epigenetics and gestational diabetes: consequences in mother and child. Eigenetics 14(3):215–235 https://doi.org/10.1080/15592294.2019.1582277

    Article  Google Scholar 

  11. Hod M, Kapur A, Sacks A et al (2015) The international Federation of Gynecology and Obstetrics (FIGO). Initiative on gestational diabetes mellitus: a pragmatic guide for diagnosis, management, and care. J Gynecol Obstet 131(Suppl 3):S173–S211

    Google Scholar 

  12. Diabetes G International Diabetes Federation – Home. Available online at www.idf.org/our-activities/care-prevention/gdm

  13. Zhao J1, Weiler HA. Long-term effects of gestational diabetes on offspring health are more pronounced in skeletal growth than body composition and glucose tolerance. Br J Nutr, 2010;104(11):1641-9. doi: 10.1017/S0007114510002631.

    Article  CAS  PubMed  Google Scholar 

  14. Damm P1, Houshmand-Oeregaard A, Kelstrup L, et al. Gestational diabetes mellitus and long-term consequences for mother and offspring: a view from Denmark. Diabetologia, 2016;59 (7):1396-1399. doi: https://doi.org/10.1007/s00125-016-3985-5.

    Article  CAS  PubMed  Google Scholar 

  15. Clausen TD, Mathiesen ER, Hansen T et al (2008) High prevalence of type 2 diabetes and pre-diabetes in adult offspring of women with gestational diabetes mellitus or type 1 diabetes: the role of intrauterine hyperglycemia. Diabetes Care 31:340–346. https://doi.org/10.2337/dc07-1596

    Article  PubMed  Google Scholar 

  16. Assche FAV, Holemans K, Aerts L (2011) Long-term consequences for offspring of diabetes during pregnancy. British Medical Bulletin 60(1):173–182 https://doi.org/10.1093/bmb/60.1.173

    Article  Google Scholar 

  17. International Diabetes Federation. Diabetes Atlas 8th ed., 2017. Available online at http://fmdiabetes.org/wp-content/uploads/2018/03/IDF-2017.pdf

  18. Kaaja R, Rönnemaa T (2008) Gestational Diabetes: Pathogenesis and Consequences to Mother and Offspring. Rev Diabet Stud. 5(4):194–202. https://doi.org/10.1900/RDS.2008.5.194

    Article  PubMed  Google Scholar 

  19. Shu L, Sauter NS, Schulthess FT, Matveyenko AV, Oberholzer. Transcription factor 7-like 2 regulates beta-cell survival and function in human pancreatic islets. Diabetes, 2008; 57: 645-653.

  20. MacDonald PE, Ha XF, Wang J, Smukler SR, Sun AM, Gaisano HY (2001) Members of the Kv1 and Kv2 voltage-dependent K(+) channel families regulate insulin secretion. Mol Endocrinol 15(8):1423–1435. https://doi.org/10.1210/mend.15.8.0685

    Article  CAS  PubMed  Google Scholar 

  21. Huerta-Chagoya A, Vázquez-Cárdenas P, Moreno-Macías H, Tapia-Maruri L, Rodríguez-Guillén R, López-Vite E (2015) Genetic determinants for gestational diabetes mellitus and related metabolic traits in Mexican women. PloS one 10(5):e0126408. https://doi.org/10.1371/journal.pone.0126408

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Prokopenko I, Langenberg C, Florez JC et al (2009) Variants in MTNR1B influence fasting glucose levels. Nature genetics 41(1):77–81. 19060907

    Article  CAS  PubMed  Google Scholar 

  23. Shang M, Wen Z (2018) Increased placental IGF-1/mTOR activity in macrosomia born to women with gestational diabetes. Diabetes Research and Clinical Practice 146:211–219 https://doi.org/10.1016/j.diabres.2018.10.017

    Article  CAS  PubMed  Google Scholar 

  24. Casellas A, Mallol C, Salavert A, Jimenez V, Garcia M, Agudo J (2015) Insulin-like Growth Factor 2 Overexpression Induces β-Cell Dysfunction and Increases Beta-cell Susceptibility to Damage. The Journal of Biological Chemistry 290:16772–16785. https://doi.org/10.1074/jbc.M115.642041

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ramirez VI, Miller E, Meireles CL, Gelfond J, Krummel DA, Powell TL (2014) Adiponectin and IGFBP-1 in the development of gestational diabetes in obese mothers. BMJ Open Diabetes Research and Care 2:e000010. https://doi.org/10.1136/%20bmjdrc-2013-000010

    Article  PubMed  PubMed Central  Google Scholar 

  26. DeMambro VE, Clemmons DR, Horton LG, Bouxsein ML, Wood TL, Beamer WG (2008) Gender-specific changes in bone turnover and skeletal architecture in IGFBP2-null mice. Endocrinology 149:2051–2061. https://doi.org/10.1210/en.2007-1068

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chon SJ, Kim SY, Cho NR, Min DL, Hwang YJ, Mamura M (2013) Association of variants in PPARγ², IGF2BP2, and KCNQ1 with a susceptibility to gestational diabetes mellitus in a Korean population. Yonsei Medical Journal 54(2):352–357. https://doi.org/10.3349/ymj.2013.54.2.352

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Schwanstecher C, Meyer U, Schwanstecher M (2002) K(IR)6.2 polymorphism predisposes to type 2 diabetes by inducing overactivity of pancreatic beta-cell ATP-sensitive K(+) channels. Diabetes 51:875–879. https://doi.org/10.2337/diabetes.51.3.875

    Article  CAS  PubMed  Google Scholar 

  29. Ober C, Xiang KS, Thisted RA, Indovina KA, Wason CJ, Dooley S (1989) Increased risk for gestational diabetes mellitus associated with insulin receptor and insulin-like growth factor II restriction fragment length polymorphisms. Genet Epidemiol 6:559–569

    Article  CAS  PubMed  Google Scholar 

  30. Ceasrine AM, Lin EE, Lumelsky DN, Iyer R, Kuruvilla R (2018) Adrb2 controls glucose homeostasis by developmental regulation of pancreatic islet vasculature. ELife 7:e39689. https://doi.org/10.7554/eLife.39689

    Article  PubMed  PubMed Central  Google Scholar 

  31. Guan L, Cui X, Zhou H (2018) Meta-analysis of the association between the Trp64Arg polymorphism of the beta-3 adrenergic receptor and susceptibility to gestational diabetes mellitus. J Obstet Gynaecol 38(2):172–176. https://doi.org/10.1080/01443615.2017.1331339

    Article  CAS  PubMed  Google Scholar 

  32. Feng Y, Jiang CD, Chang AM et al (2019) Interactions among insulin resistance, inflammation factors, obesity-related gene polymorphisms, environmental risk factors, and diet in the development of gestational diabetes mellitus. J Matern Fetal Neonatal Med. 32(2):339–347. https://doi.org/10.1080/14767058.2018.1446207

    Article  CAS  PubMed  Google Scholar 

  33. Kapoor RR, Flanagan SE, James CT, McKiernan J, Thomas AM, Harmer SC (2011) Hyperinsulinaemic hypoglycaemia and diabetes mellitus due to dominant ABCC8/KCNJ11 mutations. Diabetologia 54(10):2575–2583. https://doi.org/10.1007/s00125-011-2207-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Castro-Martínez AG, Sánchez-Corona J, Vázquez-Vargas AP, García-Zapién AG, López-Quintero A, Villalpando-Velazco HJ. Association analysis of calpain 10 gene variants/haplotypes with gestational diabetes mellitus among Mexican women. Cell Mol Biol (Noisy-le-grand). 2018; 28;64(3):81-86. doi: https://doi.org/10.14715/cmb/2018.64.3.13.

    Article  PubMed  Google Scholar 

  35. Megia A, Gallart L (2004) Fernández-Real J-M, Vendrell J, Simón I, Gutierrez C. Mannose-Binding Lectin Gene Polymorphisms Are Associated with Gestational Diabetes Mellitus. The Journal of Clinical Endocrinology & Metabolism 89(10):5081–5087 https://doi.org/10.1210/jc.2004-0211

    Article  CAS  Google Scholar 

  36. Catalano PM (2014) Trying to understand gestational diabetes. Diabet. Med 31:273–281. https://doi.org/10.1111/dme.12381

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yang Q, Graham TE, Mody N et al (2005) Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature 436:356–362

    Article  CAS  PubMed  Google Scholar 

  38. Shimin Hu, Shujuan Ma, Xun Li, et al. Relationships of SLC2A4, RBP4, PCK1, and PI3K Gene Polymorphisms with Gestational Diabetes Mellitus in a Chinese Population. BioMed Research International, 2018; 2019:Article ID 7398063. https://doi.org/10.1155/2019/7398063.

    CAS  Google Scholar 

  39. Shimin H, Junxia Y, Yiping Y, Guilian Y, Hui Z, Xun L (2019) Association of polymorphisms in STRA6 gene with gestational diabetes mellitus in a Chinese Han population. Medicine 98(11):e14885. https://doi.org/10.1097/MD.0000000000014885

    Article  CAS  Google Scholar 

  40. Zhu B, Huang K, Yan S, et al. VDR Variants rather than Early Pregnancy Vitamin D Concentrations Are Associated with the Risk of Gestational Diabetes: The Ma’anshan Birth Cohort (MABC) Study. J. Diabetes Res, 2019; 2019: Article ID 8313901. https://doi.org/10.1155/2019/8313901.

    Google Scholar 

  41. Apaydın M, Beyse S, Eyerci N, Pinarli FA, Ulubay M, Kizilgul M. The VDR gene FokI polymorphism is associated with gestational diabetes mellitus in Turkish women. BMC Medical Genetics, 2019; 20: Article number: 82. DOI https://doi.org/10.1186/s12881-019-0820-0

  42. Wang K, Chen Q, Feng Y, et al. Single Nucleotide Polymorphisms in CDKAL1 Gene Are Associated with Risk of Gestational Diabetes Mellitus in Chinese Population. J Diabetes Res, 2019; 2019: Article ID 3618103. https://doi.org/10.1155/2019/3618103.

    CAS  Google Scholar 

  43. Okruszko A, Kinalski M, Kuźmicki M, Mirończuk K, Wawrusiewicz-Kurylonek N, Kretowski KIA (2007) Glucokinase gene mutations in gestational diabetes in Polish population. Prediction of diabetes mellitus development after delivery. Przegl Lek 64(6):401–405

    PubMed  Google Scholar 

  44. Mei J, Liao S, Liu Y, Tan Y, Wang H, Liang Y (2015) Association of variants in CDKN2A/2B and CDKAL1 genes with gestational insulin sensitivity and disposition in pregnant Han Chinese women. J Diabetes Investig 6(3):295–301. https://doi.org/10.1111/jdi.12315

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang Y, Nie M, Li W et al (2011) Association of Six Single Nucleotide Polymorphisms with Gestational Diabetes Mellitus in a Chinese Population. PLoS ONE 6(11):e26953 https://doi.org/10.1371/journal.pone.0026953

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Cho YM, Kim TH, Lim S, Choi SH, Shin HD, Lee HK (2009) Type 2 diabetes-associated genetic variants discovered in the recent genome-wide association studies are related to gestational diabetes mellitus in the Korean population. Diabetol 52:253–261. https://doi.org/10.1007/s00125-008-1196-4

    Article  CAS  Google Scholar 

  47. Seman NA, Mohamud WN, Östenson CG, et al. Increased DNA methylation of the SLC30A8 gene promoter is associated with type 2 diabetes in a Malay population. Clin. Epigenetics, 2015; 7: 30. https://doi.org/10.1186/%20s13148-015-0049-5.

  48. Marcello MA, Calixto AR, de Almeida JF, et al. Polymorphism in LEP and LEPR May Modify Leptin Levels and Represent Risk Factors for Thyroid Cancer. Int. J. Endocrinol. 2015: 173218. https://doi.org/10.1155/2015/173218.

    Article  CAS  Google Scholar 

  49. Lin R, Ju H, Yuan Z, Zeng L, Sun Y, Su Z (2017) Association of maternal and fetal LEPR common variants with maternal glycemic traits during pregnancy. Sci Rep 7(1):3112. https://doi.org/10.1038/s41598-017-03518-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Watanabe RM, Xiang AH, Allayee H, et al. Variation in the P2-promoter region of hepatocyte nuclear factor-4a (HNF4A) is associated with ß-cell function in Mexican American families of a proband with gestational diabetes. Diabetes, 2005; 54 (supplement 1): A28.

  51. Ridderstråle M, Groop L (2009) Genetic dissection of type 2 diabetes. Molecular and Cellular Endocrinology 297(1-2):10–17

    Article  PubMed  CAS  Google Scholar 

  52. Ategbo JM, Grissa O, Yessoufou A, Hichami A, Dramane KL, Moutairou K (2006) Modulation of adipokines and cytokines in gestational diabetes and macrosomia. J Clin Endocrinol Metab. 91:4137–4143

    Article  CAS  PubMed  Google Scholar 

  53. Toppings NB, McMillan JM, Au P, Suchowersky O, Donovan LE. Wolfram Syndrome: A Case Report and Review of Clinical Manifestations, Genetics Pathophysiology, and Potential Therapies. Case Reports in Endocrinol, 2018; 2018: Article ID 9412676 doi:https://doi.org/10.1155/2018/9412676.

    Article  Google Scholar 

  54. Vana DR, Adapa D, Prasad VSS, Choudhury A, Ahuja G. Diabetes mellitus types: Key genetic determinants and risk assessment. Genet Mol Res, 2019; 18 (2): gmr16039952.

  55. Kwak SH, Park BL, Kim H, German MS, Go MJ, Jung HS. Association of Variations in TPH1 and HTR2B with Gestational Weight Gain and Measures of Obesity. Obesity, 2012; 20 (1). https://doi.org/10.1038/oby.2011.253

    Article  PubMed  CAS  Google Scholar 

  56. Oh CM, Park S, Kim H (2016) Serotonin as a New Therapeutic Target for Diabetes Mellitus and Obesity. Diabetes Metab J 40(2):89–98. https://doi.org/10.4093/dmj.2016.40.2.89

    Article  PubMed  PubMed Central  Google Scholar 

  57. Beysel S, Eyerci N, Ulubay M, Caliskan M, Kizilgul M, Hafızoğlu M. Maternal genetic contribution to pre-pregnancy obesity, gestational weight gain, and gestational diabetes mellitus. Diabetol Metabol Synd, 2019; 11: Article number: 37. https://doi.org/10.1186/s13098-019-0434-x

  58. Pollin TI, Jablonski KA, McAteer JB et al (2011) Triglyceride response to an intensive lifestyle intervention is enhanced in carriers of the GCKR Pro446Leu polymorphism. J Clin Endocrinol Metab 96:E1142–E1147. https://doi.org/10.1210/jc.2010-2324

    Article  PubMed  PubMed Central  Google Scholar 

  59. Grimsby J, Coffey JW, Dvorozniak MT, Magram J, Li G, Matschinsky FM. Characterization of glucokinase regulatory protein-deficient mice. J Biol Chem, 2000; 275 :7826 –7831. doi: https://doi.org/10.1074/jbc.275.11.7826

    Article  CAS  PubMed  Google Scholar 

  60. Zheng L, Li C, Qi WH et al (2017) Expression of macrophage migration inhibitory factor gene in placenta tissue and its correlation with gestational diabetes mellitus. Zhonghua yi xue za zhi 97:3388–3391

    CAS  PubMed  Google Scholar 

  61. Kawai VK, Levinson RT, Adefurin A, Kurnik D, Collier SP, Conway D (2017) Variation in the α2A-adrenergic receptor gene and risk of gestational diabetes. Pharmacog 18(15):1381–1386. https://doi.org/10.2217/pgs-2017-0079

    Article  CAS  Google Scholar 

  62. Leitner M, Fragner L, Danner S, Holeschofsky N, Leitner K, Tischler S (2017) Combined metabolomic analysis of plasma and urine reveals AHBA, Tryptophan and Serotonin metabolism as potential risk factors in Gestational Diabetes Mellitus (GDM). Front Mol Biosci 4:84. https://doi.org/10.3389/fmolb.2017.00084

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Beysel S, Eyerci N, Ulubay M, Caliskan M, Kizilgul M, Hafızoğlu M. Maternal genetic contribution to pre-pregnancy obesity, gestational weight gain, and gestational diabetes mellitus. Diabetol Metab Synd, 2019; 11: Article number: 37. https://doi.org/10.1186/s13098-019-0434-x

  64. Mellado-Gil JM, Fuente-Martín E, Lorenzo PI, Cobo-Vuilleumier N, López-Noriega L, Martín-Montalvo, A. The type 2 diabetes-associated HMG20A gene is mandatory for islet beta cell functional maturity. Cell Death & Disease, 2018; 9: Article number: 279.

  65. Hodson DJ, Mitchell RK, Marselli L, Pullen TJ, Gimeno Brias S, Semplici F (2014) ADCY5 couples glucose to insulin secretion in human islets. Diabetes 63(9):3009–3021. https://doi.org/10.2337/db13-1607

    Article  PubMed  PubMed Central  Google Scholar 

  66. Mojtaba E, Mahdi K, Mehdi KJR, Amir S (2011) Serum interleukin-1 beta plays an important role in insulin secretion in type II diabetic. Inter J Bio 1(3):93–99

    Google Scholar 

  67. Meier JJ, Ritzel RA, Maedler K, Gurlo T, Butler PC (2006) Increased vulnerability of newly forming beta cells to cytokine-induced cell death. Diabetolog 49:83–89. https://doi.org/10.1007/s00125-005-0069-3

    Article  CAS  Google Scholar 

  68. Kang J, Liu C-H, Lee C-N, Li H-Y, Yang C-W, Huang S-C (2019) Novel Interleukin-10 Gene Polymorphism Is Linked to Gestational Diabetes in Taiwanese Population. Front. Genet 10:89. https://doi.org/10.3389/fgene.2019.00089

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Martin-Montalvo A, López-Noriega L, Jiménez-Moreno C, Herranz A, Lorenzo PI, Cobo-Vuilleumier N (2019) Transient PAX8 Expression in Islets During Pregnancy Correlates With β-Cell Survival, Revealing a Novel Candidate Gene in Gestational Diabetes Mellitus. Diabetes 68(1):109–118 https://doi.org/10.2337/db18-0285

    Article  CAS  PubMed  Google Scholar 

  70. Takhshid MA, Haem Z, Aboualizadeh F (2015) The association of circulating adiponectin and + 45 T/G polymorphism of adiponectin gene with gestational diabetes mellitus in Iranian population. J Diabetes Metab Disord 14:30. https://doi.org/10.1186/s40200-015-0156-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Hasanvand Z, Sadeghi A, Rezvanfar MR, Goodarzi MT, Rahmannezhad G, Mashayekh FJ (2018) Association between chemerin rs17173608 and rs4721 gene polymorphisms and gestational diabetes mellitus in Iranian pregnant women. Gene 649:87–92 https://doi.org/10.1016/j.gene.2018.01.061

    Article  CAS  PubMed  Google Scholar 

  72. Ademoglu E, Berberoglu Z, Dellal FD, Ariel MK, Kose A, Candan Z. Higher Levels of Circulating Chemerin in Obese Women with Gestational Diabetes Mellitus. Acta Endocrinologica (Buc), 2015; XI (1): 32-37. doi: https://doi.org/10.4183/aeb.2015.32.

    Article  Google Scholar 

  73. Steppan CM, Bailey ST, Bhat S et al (2001) The hormone resistin links obesity to diabetes. Nature 409:307–312

    Article  CAS  PubMed  Google Scholar 

  74. Aslan M, Celik O, Celik N et al (2012) Cord blood nesfatin-1 and apelin-36 levels in gestational diabetes mellitus. Endocrine 41:424–429

    Article  CAS  PubMed  Google Scholar 

  75. Franz M, Polterauer M, Springer S, Kuessel L, Haslinger P, Worda C (2018) Maternal and neonatal omentin-1 levels in gestational diabetes. Arch gynecol obst 297(4):885–889. https://doi.org/10.1007/s00404-018-4652-5

    Article  CAS  Google Scholar 

  76. Ferreira AF, Rezende JC, Vaikousi E et al (2011) Maternal serum visfatin at 11-13 weeks of gestation in gestational diabetes mellitus. Clin Chem 57:609–613

    Article  CAS  PubMed  Google Scholar 

  77. Mellado-Gil JM, Fuente-Martín E, Lorenzo PI, Cobo-Vuilleumier N, López-Noriega L, Martín-Montalvo A. The type 2 diabetes-associated HMG20A gene is mandatory for islet beta cell functional maturity. Cell Death & Disease, 2018; 9:Article number: 279.

  78. Ding M, Chavarro J, Olsen S, Lin Y, Ley SH, Bao W. Genetic variants of gestational diabetes mellitus: a study of 112 SNPs among 8722 women in two independent populations. Diabetolog, 2018; 61 (8): 1758–1768. https://doi.org/10.1007/s00125-018-4637-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Khan IA, Shaik NA, Pasupuleti N, Chav S, Jahan P, Hasan Q (2015) Screening of mitochondrial mutations and insertion–deletion polymorphism in gestational diabetes mellitus in the Asian Indian population. Saudi J Bio Sci 22(3):243–248 https://doi.org/10.1016/j.sjbs.2014.11.001

    Article  CAS  Google Scholar 

  80. Banerjee RR, Cyphert HA, Walker EM, Chakravarthy H, Peiris H, Gu X (2016) Gestational Diabetes Mellitus From Inactivation of Prolactin Receptor and MafB in Islet β-Cells. Diabetes 65(8):2331–2341. https://doi.org/10.2337/db15-1527

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Zha W, Ho HTB, Hu T, Hebert MF, Wang MJ. Serotonin transporter deficiency drives estrogen-dependent obesity and glucose intolerance. Sci Rep, 2017; 7: Article number: 1137. DOI https://doi.org/10.1038/s41598-017-01291-5.

  82. Leipold H, Knoefler M, Gruber C, Klein K, Haslinger P, Worda C (2006) Plasminogen activator inhibitor 1 gene polymorphism and gestational diabetes mellitus. Obstet Gynecol 107(3):651–656. https://doi.org/10.1097/01.AOG.0000199953.27961.f9

    Article  PubMed  Google Scholar 

  83. Stuebe AM, Wise A, Nguyen T, Herring A, North KE, Siega-Riz AM (2014) Maternal genotype and gestational diabetes. Am J Perinatol. 31(1):69–76. https://doi.org/10.1055/s-0033-1334451

    Article  PubMed  Google Scholar 

  84. Chagnon MJ, Elchebly M, Uetani N, Dombrowski L, Cheng A, Mooney RA (2006) Altered glucose homeostasis in mice lacking the receptor protein tyrosine phosphatase sigma. Can J Physiol Pharmacol 84:755–763. https://doi.org/10.1139/y06-020

    Article  CAS  PubMed  Google Scholar 

  85. Alamolhoda SH, Yazdkhasti M, Namdari M, Zakariayi SJ, Mirabi P (2019) Association between C-reactive protein and gestational diabetes: a prospective study. J Obstet Gynaecol 9:1–5. https://doi.org/10.1080/01443615.2019.1631767

    Article  CAS  Google Scholar 

  86. Zhang YH, Van Hove JL, McCabe ER, Dipple KM (2015) Gestational Diabetes Associated with a Novel Mutation (378-379insTT) in the Glycerol Kinase Gene. Mol Genet Metab Rep 4:42–45. https://doi.org/10.1016/j.ymgmr.2015.06.004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Xu T, Shi Y, Liu J, Liu Y, Zhu A, Xie C (2017) The rs10229583 polymorphism near paired box gene 4 is associated with gestational diabetes mellitus in Chinese women. J Inter Med Res 46(1):116–121. https://doi.org/10.1177/0300060517714934

    Article  CAS  Google Scholar 

  88. Gong M, Yu Y, Liang L, Vuralli D, Froehler S, Kuehnen P (2019) HDAC4 mutations cause diabetes and induce β-cell FoxO1 nuclear exclusion. Mol Genet Gen med 7(5):e602. https://doi.org/10.1002/mgg3.602

    Article  CAS  Google Scholar 

  89. Farhan S, Handisurya A, Todoric J, Tura A, Pacini G, Wagner O (2012) Fetuin-A Characteristics during and after Pregnancy: Result from a Case Control Pilot Study. Inter J endocrinol 896736. https://doi.org/10.1155/2012/896736

    Article  CAS  Google Scholar 

  90. Kralisch S, Hoffmann A, Lössner U (2017) Kratzsch J3, Blüher M, Stumvoll M. Regulation of the novel adipokines/ hepatokines fetuin A and fetuin B in gestational diabetes mellitus. Metab 68:88–94. https://doi.org/10.1016/j.metabol.2016.11.017

    Article  CAS  Google Scholar 

  91. Bonakdaran S, Khorasani ZM, Jafarzadeh F (2017) Increased Serum Level of Fgf21 in Gestational Diabetes Mellitus. Acta Endocrinol 13(3):278–281. https://doi.org/10.4183/aeb.2017.278

    Article  CAS  Google Scholar 

  92. Xie K, Chen T, Zhang Y, Wen J, Cui X, You L. Association of rs10830962 polymorphism with gestational diabetes mellitus risk in a Chinese population. Sci Rep, 2019; 9:Article number: 5357. DOI https://doi.org/10.1038/s41598-019-41605-3

  93. Huerta-Chagoya A, Vázquez-Cárdenas P, Moreno-Macías H et al (2015) Genetic Determinants for Gestational Diabetes Mellitus and Related Metabolic Traits in Mexican Women. PLoS ONE 10(5):e0126408 https://doi.org/10.1371/journal.pone.0126408

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Wu L, Cui L, Tam W et al (2016) Genetic variants associated with gestational diabetes mellitus: a meta-analysis and subgroup analysis. Sci Rep 6:30539. https://doi.org/10.1038/srep30539

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Shaat N, Lernmark A, Karlsson E et al (2007) A variant in the transcription factor 7-like 2 (TCF7L2) gene is associated with an increased risk of gestational diabetes mellitus. Diabetologia 50:972–979

    Article  CAS  PubMed  Google Scholar 

  96. Adeyemo AA, Zaghloul NA, Chen G et al (2019) ZRANB3 is an African-specific type 2 diabetes locus associated with beta-cell mass and insulin response. Nat Commun 10:3195. https://doi.org/10.1038/s41467-019-10967-7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Lin P-C, Lin W-T, Yeh Y-H, Wung S-F (2016) Transcription Factor 7-Like 2 (TCF7L2) rs7903146 Polymorphism as a Risk Factor for Gestational Diabetes Mellitus: A Meta-Analysis. PLoS One 11(4):e0153044 https://doi.org/10.1371/journal.pone.0153044

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Lauenborg J, Grarup N, Damm P et al (2009) Common Type 2 Diabetes Risk Gene Variants Associate with Gestational Diabetes. J Clin Endocrinol & Metab 94(1):145–150 https://doi.org/10.1210/jc.2008-1336

    Article  CAS  Google Scholar 

  99. Kawai VK, Levinson RT, Adefurin A et al (2017) A genetic risk score that includes common type 2 diabetes risk variants is associated with gestational diabetes. Clin Endocrinol 87(2):149–155. https://doi.org/10.1111/cen.13356

    Article  CAS  Google Scholar 

  100. Kwak SH, Kim TH, Cho YM, Choi SH, Jang HC, Park KS (2010) Polymorphisms in KCNQ1 are associated with gestational diabetes in a Korean population. Horm Res Paediatr. 74:333–338

    Article  CAS  PubMed  Google Scholar 

  101. Fatima SS, Chaudhry B, Khan TA, Farooq S. KCNQ1 rs2237895 polymorphism is associated with Gestational Diabetes in Pakistani Women. Pak J Med Sci. 2016 Nov-Dec;32(6):1380-1385. doi: https://doi.org/10.12669/pjms.326.11052.

  102. Kwak SH, Jang HC, Park KS (2012) Finding Genetic Risk Factors of Gestational Diabetes. Genomics Inform. 10(4):239–243. https://doi.org/10.5808/GI.2012.10.4.239

    Article  PubMed  PubMed Central  Google Scholar 

  103. Ding M (2018) Chavarro J1, Olsen S, Lin Y, Ley SH1, Bao W, et al. Genetic Variants of Gestational Diabetes Mellitus: a Study of 112 SNPs Among 8,722 Women in Two Independent Populations. Diabetologia. 61(8):1758–1768. https://doi.org/10.1007/s00125-018-4637-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Alharbi KK, Al-Sulaiman AM, Bin Shedaid MK, et al. MTNR1B genetic polymorphisms as risk factors for gestational diabetes mellitus: a case-control study in a single tertiary care center. Ann Saudi Med. 2019; 39(5): 309-318. doi: https://doi.org/10.5144/0256-4947.2019.309.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Mansoori Y, Daraei A, Naghizadeh MM, Salehi R (2015) Significance of a common variant in the CDKAL1 gene with susceptibility to type 2 diabetes mellitus in Iranian population. Adv Biomed Res 4:45. https://doi.org/10.4103/2277-9175.151256

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Shaat N, Ekelund M, Lernmark A et al (2005) Association of the E23K polymorphism in the KCNJ11 gene with gestational diabetes mellitus. Diabetolog 48:2544–2551

    Article  CAS  Google Scholar 

  107. Fallucca F, Dalfra MG, Sciullo E et al (2006) Polymorphisms of insulin receptor substrate 1 and beta3-adrenergic receptor genes in gestational diabetes and normal pregnancy. Metab 55:1451–1456

    Article  CAS  Google Scholar 

  108. Alharbi KK, Khan IA, Abotalib Z, Al-Hakeem MM. Insulin Receptor Substrate-1 (IRS-1) Gly927Arg: Correlation with Gestational Diabetes Mellitus in Saudi Women. BioMed Res Intern, 2014, Article ID 146495. https://doi.org/10.1155/2014/146495.

    Article  CAS  Google Scholar 

  109. Rosta K, Al-Aissa Z, Hadarits O et al (2017) Association study with 77 SNPs confirms the robust role for the rs10830963/G of MTNR1B variant and identifies two novel associations in gestational diabetes mellitus development. PLoS One 12:e0169781

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Cho YM, Kim TH, Lim S et al (2009) Type 2 diabetes-associated genetic variants discovered in the recent genome-wide association studies are related to gestational diabetes mellitus in the Korean population. Diabetolog 52(2):253–261

    Article  CAS  Google Scholar 

  111. Wang Y, Nie M, Li W et al (2011) Association of six single nucleotide polymorphisms with gestational diabetes mellitus in a Chinese population. PloS One 6(11):e26953

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Lauenborg J, Grarup N, Damm P et al (2009) Common type 2 diabetes risk gene variants associate with gestational diabetes. J Clin Endocrinol Metab 94(1):145–150

    Article  CAS  PubMed  Google Scholar 

  113. Stuebe A, Wise A, Nguyen T, Herring A, North K, Siega-Riz A (2013) Maternal Genotype and Gestational Diabetes. Am. J. Perinatol. 31:069–076

    Article  Google Scholar 

  114. Jamalpour S, Zain SM, Mosavat M, Mohamed Z, Omar S.Z. A case-control study and meta-analysis confirm glucokinase regulatory gene rs780094 is a risk factor for gestational diabetes mellitus. Gene 2018, 650, 34–40.

    Article  CAS  PubMed  Google Scholar 

  115. Anghebem-Oliveira MI, Webber S, Alberton D et al (2017) The GCKR Gene Polymorphism rs780094 is a Risk Factor for Gestational Diabetes in a Brazilian Population. J. Clin. Lab. Anal 31:e22035

    Article  CAS  Google Scholar 

  116. Bonakdaran S, Khorasani ZM, Jafarzadeh F (2017) Increased Serum Level of Fgf21 in Gestational Diabetes Mellitus. Acta Endocrinol (Buchar). 13(3):278–281. https://doi.org/10.4183/aeb.2017.278

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Chen C, Cheung BM, Tso AW et al (2011) High plasma level of fibroblast growth factor 21 is an Independent predictor of type 2 diabetes: a 5.4-year population-based prospective study in Chinese subjects. Diabetes Care. 34(9):2113–2115. https://doi.org/10.2337/dc11-0294

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Nitert D, Barrett HL, Kubala MH et al (2014) Increased Placental Expression of Fibroblast Growth Factor 21 in Gestational Diabetes Mellitus. J Clin Endocrinol & Metab 99(4):E591–E598 https://doi.org/10.1210/jc.2013-2581

    Article  CAS  Google Scholar 

  119. Rissanen J, Markkanen A, Karkkainen P et al (2000) Sulfonylurea receptor 1 gene variants are associated with gestational diabetes and type 2 diabetes but not with altered secretion of insulin. Diabetes Care 23:70–73

    Article  CAS  PubMed  Google Scholar 

  120. Hasanvand Z, Sadeghi A, Rezvanfar MR, Goodarzi MT, Rahmannezhad G, Mashayekhi FJ (2018) Association between chemerin rs17173608 and rs4721 gene polymorphisms and gestational diabetes mellitus in Iranian pregnant women. Gene. 649:87–92. https://doi.org/10.1016/j.gene.2018.01.061

    Article  CAS  PubMed  Google Scholar 

  121. Litou H, Anastasiou E, Thalassinou L et al (2007) Increased prevalence of VNTR III of the insulin gene in women with gestational diabetes mellitus (GDM). Diabetes Res Clin Pract 76:223–228

    Article  CAS  PubMed  Google Scholar 

  122. Handy DE, Castro R, Loscalzo J (2011) Epigenetic Modifications: Basic Mechanisms and Role in Cardiovascular Disease. Circulation. 123(19):2145–2156. https://doi.org/10.1161/CIRCULATIONAHA.110.956839

    Article  PubMed  PubMed Central  Google Scholar 

  123. Feil R, Fraga MF (2011) Epigenetics and the environment: Emerging patterns and implications. Nat Rev Genet 13:97–109. https://doi.org/10.1038/nrg3142

    Article  CAS  Google Scholar 

  124. Ordovas JM, Smith CE (2010) Epigenetics and cardiovascular disease. Nat Rev Cardiol 7:510–519. https://doi.org/10.1038/nrcardio.2010.104

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Stankov K, Benc D, Draskovic D (2013) Genetic and epigenetic factors in etiology of diabetes mellitus type 1. Pediatr 132:1112–1122

    Article  Google Scholar 

  126. Xie Z, Chang C, Zhou Z (2014) Molecular mechanisms in autoimmune type 1 diabetes: a critical review. Clin Rev Allergy Immunol 47:174–192

    Article  CAS  PubMed  Google Scholar 

  127. Kimmins S, Sassone-Corsi P (2005) Chromatin remodelling and epigenetic features of germ cells. Nature 434(7033):583–589

    Article  CAS  PubMed  Google Scholar 

  128. Kang J, Lee C-N, Li H-Y, Hsu K-H, Wang S-H, Lin S-Y (2018) Association of Interleukin-10 Methylation Levels With Gestational Diabetes in a Taiwanese Population. Front Genet 9:222. https://doi.org/10.3389/fgene.2018.00222

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Jacovetti C, Abderrahmani A, Parnaud G, Jonas JC, Peyot ML, Cornu M (2012) MicroRNAs contribute to compensatory β cell expansion during pregnancy and obesity. J Clin Invest 122(10):3541–3551. https://doi.org/10.1172/JCI64151

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Stirm L, Huypens P, Sass S, Batra R, Fritsche L, Brucker S. Maternal whole blood cell miRNA-340 is elevated in gestational diabetes and inversely regulated by glucose and insulin. Sci Rep, 2018; 8:1366-1378. | DOI:https://doi.org/10.1038/s41598-018-19200-9.

  131. Jo H, Eckel SP, Chen J-C, Cockburn M, Martinez MP, Chow T (2019) Associations of gestational diabetes mellitus with residential air pollution exposure in a large Southern California pregnancy cohort. Environ Inter 130:104933–104939 https://doi.org/10.1016/j.envint.2019.104933

    Article  CAS  Google Scholar 

  132. Lodovici M, Bigagli E. Oxidative stress and air pollution exposure. J Toxicol, 2011; (2011): Article 487074

  133. Saldana TM, Basso O, Hoppin JA, Baird DD, Knott C, Blair A (2007) Pesticide Exposure and Self-Reported Gestational Diabetes Mellitus in the Agricultural Health Study. Diabetes Care 30(3):529–534. https://doi.org/10.2337/dc06-1832

    Article  PubMed  Google Scholar 

  134. Vu BG, Stach CS, Kulhankova K, Salgado-Pabón W, Klingelhutz AJ, Schlievert PM. Chronic Superantigen Exposure Induces Systemic Inflammation, Elevated Bloodstream Endotoxin, and Abnormal Glucose Tolerance in Rabbits: Possible Role in Diabetes. mBio, 2015; 6 (2): e02554-14 DOI: https://doi.org/10.1128/mBio.02554-14.

  135. Crusell MKW, Hansen TH, Nielsen T, Allin KH, Rühlemann MC, Damm P (2018) Gestational diabetes is associated with change in the gut microbiota composition in third trimester of pregnancy and postpartum. Microbiome 6:89 https://doi.org/10.1186/s40168-018-0472-x

    Article  PubMed  PubMed Central  Google Scholar 

  136. Mokkala K, Röytiö H, Munukka E, Pietilä S, Ekblad U, Rönnemaa T (2016) Gut Microbiota Richness and Composition and Dietary Intake of Overweight Pregnant Women Are Related to Serum Zonulin Concentration, a Marker for Intestinal Permeability. J Nutr 146(9):1694–1700. https://doi.org/10.3945/jn.116.235358

    Article  CAS  PubMed  Google Scholar 

  137. Duke-NUS Medical School. Association between insufficient sleep, gestational diabetes mellitus discovered. ScienceDaily. www.sciencedaily.com/releases/2017/01/170109092634.htm (accessed October 26, 2019).

  138. Mak JKL, Pham NM, Lee AH, Tang L, Pan X-F, Binns CW. Dietary patterns during pregnancy and risk of gestational diabetes: a prospective cohort study in Western China. Nutr J, 2018; 17:Article number: 107. DOI https://doi.org/10.1186/s12937-018-0413-3

  139. Cai D, Liu H, Hu Y, Jiang Y, Zhao R Gestational Betaine, Liver Metabolism, and Epigenetics. In: Patel V., Preedy V. (eds) Handbook of Nutrition, Diet, and Epigenetics. Springer, Cham. 2017. DOI https://doi.org/10.1007/978-3-319-31143-2_82-1

    Google Scholar 

  140. Sivitz W. MItochondrial Dysfunction in Obesity and Diabetes. US Endocrinol, 2010; 6 (1): 20-27 DOI: https://doi.org/10.17925/USE.2010.06.1.20.

    Article  Google Scholar 

  141. R¨onn T, Poulsen P, Hansson O, et al. Age influences DNA methylation and gene expression of COX7A1 in human skeletal muscle. Diabetolog, 2008; 51:1159{1168. DOI: https://doi.org/10.1007/s00125-008-1018-8.

    Article  PubMed  CAS  Google Scholar 

  142. Makgoba M, Savvidou M, Steer P (2012) An analysis of the interrelationship between maternal age, body mass index and racial origin in the development of gestational diabetes mellitus. BJOG 119:276–282. https://doi.org/10.1111/j.1471-0528.2011.03156.x

    Article  CAS  PubMed  Google Scholar 

  143. Ming W, Ding W, Zhang CJP, Zhong L, Long Y, Li Z. The effect of exercise during pregnancy on gestational diabetes mellitus in normal-weight women: a systematic review and meta-analysis. BMC Pregnancy and Childbirth, 2018; 18: Article number: 440. DOI https://doi.org/10.1186/s12884-018-2068-7.

  144. Eriksson KF, Lindg¨arde F. Poor physical fitness, and impaired early insulin response but late hyperinsulinaemia, as predictors of NIDDM in middleaged Swedish men. Diabetolog, 1996; 39:573-579.

  145. Grazioli E, Dimauro I, Mercatelli N et al (2017) Physical activity in the prevention of human diseases: role of epigenetic modifications. BIC Genomics 18(Suppl 8):802. https://doi.org/10.1186/s12864-017-4193-5

    Article  CAS  Google Scholar 

  146. World Health Organization. Diagnostic criteria and classification of hyperglycaemia first detected in pregnancy. Geneva:; 2013. http://www.who.int/diabetes/publications/Hyperglycaemia_In_Pregnancy/en/

  147. United Kingdom National Health Service. Overview: Gestational Diabetes. Updated 2019. Available at https://www.nhs.uk/conditions/gestational-diabetes/.

  148. United States National Institute of Health. Tests & Diagnosis for Gestational Diabetes. Updated 2017. Available at https://www.niddk.nih.gov/health-information/diabetes/overview/what-is-diabetes/gestational/tests-diagnosis

  149. Farghaly TA, Helmy NA, Abbas AM, Ahmed AGM C-reactive protein as a screening test for gestational diabetes mellitus in first-trimester of pregnancy: a prospective cohort study. Int J Reprod Contracept Obstet Gynecol, 2018;7:4798-803. DOI https://doi.org/10.18203/2320-1770.ijrcog20184922

    Article  Google Scholar 

  150. Schaible B, Calhoun BC, Bush S, Ramser B, Seybold DJ, Broce M. Hemoglobin A1c as a screening strategy for gestational diabetes. Med Dent Res, 2018: 1 (1): 1-4. DOI: https://doi.org/10.15761/MDR.1000103

  151. Fitria N, van Asselt ADI, Postma MJ (2019) Cost-effectiveness of controlling gestational diabetes mellitus: a systematic review. Eur J Health Eco 20(3):407–417. https://doi.org/10.1007/s10198-018-1006-y

    Article  Google Scholar 

  152. Marseille E, Lohse N, Jiwani A, Hod M, Seshiah V, Yajnik CS. The cost-effectiveness of gestational diabetes screening including prevention of type 2 diabetes: application of a new model in India and Israel. J Maternal-Fetal & Neonat Med, 2013; 26 (8). https://doi.org/10.3109/14767058.2013.765845.

    Article  Google Scholar 

  153. Saccone G, Caissutti C, Khalifeh MDA, Scifres C, Simhan HN (2017) One Step versus Two Step approach for gestational diabetes screening: systematic review and meta-analysis of the randomized trials. Journal of Maternal-Fetal and Neonat Med 32(8):1–211. https://doi.org/10.1080/14767058.2017.1408068

    Article  Google Scholar 

  154. Amreen S, Suneel A, Shetty A, Vasudeva A, Kumar P Use of glycosylated HbA1c and random blood sugar as a screening tool for gestational diabetes mellitus in first trimester. Int J Reprod Contracept Obstet Gynecol, 2018;7:524-8. DOI https://doi.org/10.18203/2320-1770.ijrcog20180166

    Article  Google Scholar 

Download references

Acknowledgements

None.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

TY conceptualized, did the literature search, drafted the manuscript, and did correspondence. TS designed the article and did the literature search. YA did the article selection and proofreading. AU did the article selection and proofreading. All authors have read and approved the manuscript.

Corresponding author

Correspondence to Tajudeen O. Yahaya.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yahaya, T.O., Salisu, T., Abdulrahman, Y.B. et al. Update on the genetic and epigenetic etiology of gestational diabetes mellitus: a review. Egypt J Med Hum Genet 21, 13 (2020). https://doi.org/10.1186/s43042-020-00054-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43042-020-00054-8

Keywords