Skip to main content
Erschienen in: BMC Pregnancy and Childbirth 1/2020

Open Access 01.12.2020 | Research article

Polymorphisms of TGF-β1 and TGF-β3 in Chinese women with gestational diabetes mellitus

verfasst von: Yinglei Xu, Chunlian Wei, Cuijiao Wu, Mengmeng Han, Jingli Wang, Huabin Hou, Lu Zhang, Shiguo Liu, Ying Chen

Erschienen in: BMC Pregnancy and Childbirth | Ausgabe 1/2020

Abstract

Background

Gestational diabetes mellitus (GDM) is a pregnancy-specific carbohydrate intolerance Which can cause a large number of perinatal and postpartum complications. The members of Transforming growth factor-β (TGF-β) superfamily play key roles in the homeostasis of pancreatic β-cell and may involve in the development of GDM. This study aimed to explore the association between the polymorphisms of TGF-β1, TGF-β3 and the risk to GDM in Chinese women.

Methods

This study included 919 GDM patients (464 with preeclampsia and 455 without preeclampsia) and 1177 healthy pregnant women. TaqMan allelic discrimination real-Time PCR was used to genotype the TGF-β1 (rs4803455) and TGF-β3 (rs2284792 and rs3917201), The Hardy-Weinberg equilibrium (HWE) was evaluated by chi-square test.

Results

An increased frequency of TGF-β3 rs2284792 AA and AG genotype carriers was founded in GDM patients (AA vs. AG + GG: χ2 = 6.314, P = 0.012, OR = 1.270, 95%CI 1.054–1.530; AG vs. GG + AA: χ2 = 8.545, P = 0.003, OR = 0.773, 95%CI 0.650–0.919). But there were no significant differences in the distribution of TGF-β1 rs4803455 and TGF-β3 rs3917201 between GDM and healthy women. In addition, no significant differences were found in allele and genotype frequencies among GDM patients with preeclampsia (PE).

Conclusions

The AA and AG genotype of TGF-β3 rs2284792 polymorphism may be significantly associated with increased risk of GDM in Chinese population.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12884-020-03459-w.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
TGF-β
Transforming growth factor-β
GDM
Gestational diabetes mellitus
HWE
Hardy-Weinberg equilibrium
PE
Preeclampsia
SNPs
Signal Nucleotide Polymorphisms
OGTT
Oral glucose tolerance test
ORs
Odds ratios
Cis
confidence intervals

Background

GDM is the most common maternal metabolic disturbance that is defined as glucose intolerance of variable severity with onset or first detection during pregnancy [1, 2]. The prevalence of GDM varies from 1 to 22% of all pregnancies depending on different populations and diagnostic criteria [35]. GDM not only increases the risk of maternal and fetal perinatal complications, but also has long-term adverse consequences for offspring [6, 7]. The most familiar complication following GDM is PE which shares common clinical risk factors with GDM such as obesity, advanced maternal age and diabetes [8]. GDM is characterized by increased insulin resistance and defective insulin secretion which is due to the inability of pancreatic β cells [2]. However, the etiology is complex due to disordered metabolism and intrauterine environment during pregnancy. Extensive efforts have been made to explore the pathogenesis and to find new targets for prediction of GDM [2, 9, 10].
The TGF-β superfamily, including TGF-β isoforms, activins, inhibins and bone morphogenetic proteins (BMPs), is involved in a myriad of biological processes such as cell proliferation, differentiation and death [11]. In addition, TGF-β signaling has been indicated to play key roles in the development of GDM and GDM risk factors. BMPs disfunction will impair insulin signal and glucose homeostasis in the setting of diabetes [12]. Activins can promote the proliferation of pancreatic β-cell and secretion of insulin [13]. TGF-β isoforms are known to stimulate adipocyte proliferation, insulin resistance and subclinical inflammation [14].
In recent years, the role of genetic factors in the pathogenesis of GDM has been increasingly investigated. The major genetic studies of GDM are candidate gene studies, which have revealed that some single nucleotide polymorphisms (SNPs) in cytokine genes are associated with susceptibility to GDM [15, 16]. SNPs within the coding and signal sequences can affect gene transcriptional activity, and then change the production of proteins [17]. Several studies have reported that altered cytokines expression are related to the severity and progression of the GDM [18, 19]. Therefore, the cytokine genes with positive SNP loci may be a pregnancy biomarker for screening GDM.
TGF-β1 and TGF-β3 belong to TGF-β isoforms and have differential expression in the human endometrium and placenta [11]. Both of them contribute to normal homeostasis of pancreas and insulin action [20]. The enhanced expression of TGF-β1 induced by hyperglycemia was detected in individuals with GDM [21, 22]. Although there is no direct relation between TGF-β3 and GDM, TGF-β3 participates in many GDM complications such as PE and pregnancy-induced hypertension [23]. Three tag SNPs (rs4803455, rs2284792, and rs3917201), located in introns of TGF-β1 and TGF-β3 locus respectively, can affect the transcriptional activity and change the expression of proteins [2426]. Therefore, we supposed that these three SNPs might be target SNPs, and try to investigate the relationship between polymorphisms of TGF-β1, TGF-β3 and the risk of GDM.

Methods

Subjects

This study was conducted based on 919 pregnant women with GDM and 1177 healthy pregnant women with normal glucose tolerance, recruited from the clinical pregnancy registries at the Affiliated Hospital of Qingdao University, People’s Hospital of Liaocheng City and People’s Hospital of Linyi City. Informed consent was issued and signed by all subjects and all investigations were approved by the ethics committee of the Affiliated Hospital of Qingdao University.
All the participants underwent a 75 g oral glucose tolerance test (OGTT) at 24–28 weeks’ gestation. The diagnosis of GDM was based on the International Association of Diabetes and Pregnancy Study Groups (IADPSG) criteria when one of the following plasma glucose values in the OGTT was met or exceeded, fasting plasma glucose 92 mg/dl (5.1 mmol/l), 1 h plasma glucose 180 mg/dl (10.0 mmol/l) and 2 h plasma glucose 153 mg/dl (8.5 mmol/l). Plasma glucose during OGTT of the follow-up study was measured by enzymatic hexokinase photometric assay. Exclusion criteria included heart diseases, chronic hypertension, diabetes mellitus, thyroid diseases, kidney disorders, abnormal liver function, twin or multiple pregnancies, as well as in-vitro fertilization in the present gestation. Women were recruited in the study group when first diagnosed as GDM at 24–28 weeks’ gestation. Then, they were taken blood for testing before diet control and insulin therapy. Besides, 919 GDM patients were categorized into 455 without PE and 464 with PE which was determined on the base of the questionnaire, clinical features, and data. A newly onset of hypertension (≥140/90 mmHg) with proteinuria C of 300 mg or higher in 24-h after 20 weeks of gestation was diagnosed as PE.

Methods

Genomic DNA was extracted from peripheral venous blood by alkaline lysis method and collected by centrifugal column in the Qiagen blood DNA extraction kit (Qiagen, Hilden, Germany). TaqMan allelic discrimination real-time PCR (Life Technologies, Grand Island, NY, USA) was used to genotype the polymorphisms of rs4803455 in TGF-β1, rs2284792 and rs3917201 in TGF-β3. The TaqMan probes and primers were designed by Applied Bio-systems or Life Technologies (New York. USA). TGF-β1 and TGF-β3 were amplified using the following primers: 5′-GCTGCAAACATTCTGGGGTTT-3′ for TGF-β1 rs4803455, 5′-GGGTGGGACCAGGGAATCT-3′ for TGF-β3 rs2284792 and 5′-CGCCTCAAGAAGCAGAAGGAT-3′ for TGF-β3 rs3917201. Reaction volume was 25 μl: 1.25 μL 20 × SNP Genotyping Assay, 12.5 μL 2 × PCR Master Mix, and 11.25 μL DNA and DNase-free water. 1000™ Thermal cycler and CFX96™ Real-time system (Bio-Rad, California, USA) were carried out to amplifications as following conditions: 95 °C for 3 min, followed by 45 cycles at 95 °C for 15 s and 60 °C for 1 min. The fluorescent signals from VIC/FAM-labeled probes were detected for each cycle. Discrimination of genotypes was conducted with BioRad CFX manager 3.0 software.

Statistical analysis

Statistical software package IBM SPSS 22.0 (SPSS Inc., Chicago, IL, USA) was used to manipulate all data. Student’s t-test was utilized to compare the demographic and clinical characteristics of cases and controls. An analysis of variance (ANOVA) was used to conduct the genotype-phenotype analysis. A chi-square test was performed to assess the HWE in the controls. Allelic and genotypic distributions were enrolled in the comparison by using Pearson’s χ2 test which was substituted with Fisher’s exact test when expected values were below 5. P <  0.05 (two-sided) was considered to represent statistically significance. Odds ratios (ORs) and 95% confidence intervals (CIs) were used to reveal the relative risk degree. A P-value < 0.05 (two-sided) was taken as statistical significance for all statistical analyses.

Results

Demographic and clinical characteristics of GDM and controls

Subjects were categorized into 919 GDM patients and 1177 controls. Demographic and clinical data of different groups were summarized in the supplemental table.
Both groups had similar age distribution, times of gravidity, and number of abortions. The mean age of cases and controls was 30.71 ± 4.18 and 30.75 ± 4.21 years old. However, in GDM group, weeks of admission and delivery intended to be earlier (P <  0.001) and the weight gain of newborns was heavier than in the control group as expected (P <  0.001).

TGF-β1 and TGF-β3 polymorphism analysis

The subjects of the control group enrolled in this study were in accordance with HWE for these SNPs and displayed a group representative at the significance level of P>0.05.
The distributions of the genotypes and alleles in GDM cases and controls were reported in Table 1. We observed a statistically significant difference between GDM and healthy women in the frequencies of TGF-β3 rs2284792 (χ2 = 9.064, P = 0.011). However, no statistical differences were detected either in TGF-β1 rs4803455 or in TGF-β3 rs3917201 between two groups in terms of genotypic frequencies. As shown in Table 1, the allelic frequencies of rs2284792 between two groups were not obviously different (χ2 = 1.592, P = 0.207, OR = 1.082, 95%CI 0.957–1.224). When categorized into three models (AA vs AG + GG, GG vs AG + AA and AG vs GG + AA), there was a significant difference between these two groups (For AA vs AG + GG model, χ2 = 6.314, P = 0.012, OR 1.270, 95%CI 1.054–1.530; For AG vs GG + AA model, χ2 = 8.545, P = 0.003, OR = 0.773, 95%CI 0.650–0.919). Consistently, allelic frequencies of TGF-β1 rs4803455 or TGF-β3 rs3917201 were statistically insignificant.
Table 1
The comparison of genotypic and allelic frequencies of all SNPs between GDM (all cases) and controls
 
Cases
Controls
χ2
p-value
OR
95%CI
rs4803455
 Genotypes
  AA
142
173
1.206
0.574
  
  AC
412
556
    
  CC
365
448
    
 Alleles
  A
696
902
0.089
0.766
1.019
0.899–1.156
  C
1142
1452
    
rs2284792
 Genotypes
  AA
268
404
9.064
0.011*
  
  AG
487
548
    
  GG
164
225
    
  AA
268
404
    
  AG + GG
651
773
6.314
0.012*
1.270
1.054–1.530
  GG
164
225
    
  AG + AA
755
952
0.511
0.458
1.088
0.871–1.360
  AG
487
548
    
  GG + AA
432
629
8.545
0.003*
0.773
0.650–0.919
 Alleles
  A
1023
1356
1.592
0.207
1.082
0.957–1.224
  G
815
998
    
rs3917201
 Genotypes
  GG
220
294
0.303
0.895
  
  AG
468
592
    
  AA
231
291
    
 Alleles
  A
908
1180
0.218
0.641
1.029
0.911–1.163
  G
930
1174
    
*p < 0.05 is considered statistically significant, OR ODDs ratio, CI Confidence interval

TGF-β1 and TGF-β3 polymorphism analysis between GDM patients with and without PE

To further study the association between variants of the three SNPs and complications, samples were categorized into GDM cases with and without PE. The distributions of the genotypes and alleles in GDM patients with and without PE are shown in Tables 2 and 3.
Table 2
The comparison of genotypic and allelic frequencies of all SNPs between GDM without PE and controls
 
Cases
Controls
χ2
p-value
OR
95%CI
rs4803455
 Genotypes
  AA
70
173
0.631
0.730
  
  AC
205
556
    
  CC
180
448
    
 Alleles
  A
345
902
0.046
0.831
1.017
0.869–1.191
  C
565
1452
    
rs2284792
 Genotypes
  AA
122
404
9.774
0.008*
  
  AG
247
548
    
  GG
86
225
    
  AA
122
404
    
  AG + GG
333
773
8.476
0.004*
1.472
1.122–1.813
  GG
86
225
    
  AG + AA
369
952
0.010
0.921
1.014
0.769–1.336
  AG
247
548
    
  GG + AA
208
629
7.842
0.005*
0.734
0.590–0.912
 Alleles
  A
491
1356
3.555
0.059
1.159
0.994–1.352
  G
419
998
    
rs3917201
  Genotypes
  GG
108
294
0.571
0.752
  
  AG
227
592
    
  AA
120
291
    
Alleles
  A
443
1180
0.549
0.459
1.060
0.909–1.235
  G
467
1174
    
*p < 0.05 is considered statistically significant, OR ODDs ratio, CI Confidence interval
Table 3
The comparison of genotypic and allelic frequencies of all SNPs between GDM with PE and controls
 
Cases
Controls
χ2
p-value
OR
95%CI
rs4803455
 Genotypes
  AA
72
173
1.266
0.531
  
  AC
207
556
    
  CC
185
448
    
  Alleles
  A
351
902
    
  C
577
1452
    
rs2284792
 Genotypes
  AA
146
404
3.619
0.164
  
  AG
240
548
    
  GG
78
225
    
 Alleles
  A
532
1356
0.021
0.885
1.011
0.867–1.179
  G
396
998
    
rs3917201
 Genotypes
  GG
112
294
0.359
0.836
  
  AG
241
592
    
  AA
111
291
    
  Alleles
  A
463
1180
0.015
0.903
1.009
0.867–1.175
  G
465
1174
    
*p < 0.05 is considered statistically significant, OR ODDs ratio, CI Confidence interval
In GDM cases without PE group, the statistical difference between cases and controls in genotypic distributions of TGF-β3 rs2284792 was observed (χ2 = 9.774, P = 0.008). Also, the same was found for allelic frequencies in AA vs AG + GG (χ2 = 8.476, P = 0.004 OR = 1.427, 95%CI 1.122–1.813) and AG vs GG + AA (χ2 = 7.842, P = 0.005 OR = 0.734, 95%CI 0.590–0.912). In contrast to TGF-β3 rs2284792, no obvious difference was found in either the genotypic distributions or allelic frequencies of TGF-β1 rs4803455 and TGF-β3 rs3917201 among GDM only cases.
In GDM cases with PE group, however, no obvious difference was found in either the genotypic distributions or allelic frequencies of three SNPs (for rs4803455, χ2 = 1.266, P = 0.531 by genotype, χ2 = 0.069, P = 0.793, OR = 1.021, 95%CI 0.873–1.194 by allele; when for rs2284792, χ2 = 3.619, P = 0.164 by genotype, χ2 = 0.021, P = 0.885, OR = 1.011, 95%CI 0.867–1.1791by allele; and for rs3917201, χ2 = 0.359, P = 0.836 by genotype, χ2 = 0.015, P = 0.903, OR = 1.009, 95%CI 0.867–1.175 by allele).

Analysis of genotype-phenotype relationship

Analysis of the relationship between the genotypes of TGF-β3 rs2284792 and demographic characteristics among total GDM patients was shown in Table 4. However, no statistical differences were found for the genotype-phenotype relationship of rs2284792.
Table 4
Associations between genotypes of rs2284792 and characteristics among total GDM patients
Rs2284792(A/G)
AA
AG
GG
AA vs. AG
AA vs GG
AG vs GG
AA vs AG + GG
GG vs AG + AA
AG vs GG + AA
(n)
(n)
(n)
Pa
pb
pc
pd
pe
pf
Cases
268
487
164
      
Demographic characteristics (Mean ± S)
 Fasting blood glucose (mmol/l)
5.90 ± 2.13
5.75 ± 2.22
5.76 ± 2.33
0.087
0.097
0.960
0.059
0.407
0.360
 Systolic blood pressure (mmHg)
137.65 ± 22.24
137.41 ± 24.86
137.53 ± 22.06
0.166
0.425
0.468
0.224
0.997
0.231
 Diastolic blood pressure (mmHg)
77.09 ± 12.36
77.17 ± 11.86
77.01
0.519
0.560
0.137
0.972
0.186
0.177
 WBC (× 109/L)
10.51 ± 2.99
10.62 ± 3.05
10.62 ± 3.05
0.122
0.144
0.921
0.099
0.514
0.436
 RBC (× 1012/L)
4.41 ± 1.73
4.37 ± 1.17
4.51 ± 1.91
0.588
0.233
0.234
0.997
0.085
0.185
 Hb (g/L)
116.43 ± 17.40
116.47 ± 14.12
116.32 ± 14.31
0.823
0.451
0.308
0.753
0.343
0.560
 neutrophil (×109/L)
8.40 ± 2.54
8.24 ± 2.43
8.44 ± 2.81
0.157
0.779
0.068
0.557
0.167
0.056
 PLT (×109/L)
227.30 ± 58.26
226.72 ± 67.55
226.69 ± 58.86
0.102
0.149
0.953
0.079
0.562
0.252
 PT (s)
10.64 ± 1.59
10.68 ± 1.60
10.71 ± 1.83
0.436
0.283
0.572
0.330
0.402
0.808
 APTT (s)
30.57 ± 3.93
30.52 ± 3.91
30.63 ± 3.38
0.441
0.446
0.134
0.720
0.192
0.300
 ALT (IU/L)
27.66 ± 21.05
26.95 ± 16.47
27.14 ± 19.28
0.065
0.356
0.663
0.075
0.920
0.132
 AST (IU/L)
29.82 ± 18.52
29.53 ± 16.51
29.57 ± 19.35
0.384
0.599
0.930
0.389
0.871
0.510
 Creatinine (umol/L)
58.58 ± 18.75
58.49 ± 19.15
58.49 ± 17.09
0.716
0.788
0.986
0.704
0.931
0.782
 Body mass before pregnancy (kg)
59.73 ± 3.43
63.58 ± 0.95
63.04 ± 0.74
0.146
0.159
0.647
0.116
0.959
0.422
 Body mass increase during pregnancy (kg)
17.53 ± 1.25
17.57 ± 0.62
16.24 ± 0.47
0.978
0.318
0.086
0.614
0.072
0.123
 BMI before pregnancy (kg/m2)
23.97 ± 0.79
24.30 ± 0.34
24.23 ± 0.40
0.698
0.804
0.896
0.754
0.974
0.837
 BMI at birth (kg/m2)
30.7 ± 0.92
30.91 ± 0.42
30.43 ± 0.46
0.841
0.827
0.456
0.964
0.463
0.472
Pa value between AA and AG; Pb value between AA and GG; Pc value between AG and GG; Pd value between AA and AG + GG; Pe value between GG and AG + AA; Pf value between AG and GG + AA
p < 0.05 is considered statistically significant. WBC White Blood Cell, RBC Red Blood Cell, Hb Hemoglobin, PLT Platelet, PT prothrombin time, APTT activated partial thromboplastin time, ALT glutamic pyruvic transaminase, AST glutamic oxaloacetic transaminase

Discussion

In this study, the associations between TGF-β1, TGF-β3 polymorphisms and GDM were examined in a Chinese population. Among women with GDM, we firstly found an effective association between the tag SNP TGF-β3 rs2284792 and GDM risk. Besides, we confirmed that the A allele and the AA and AG genotypes were susceptible, while the G allele/GG genotype may be protective factors. However, there were no statistically significant differences in the distribution of TGF-β1 rs4803455 and TGF-β3 rs3917201 genotypes between GDM and healthy women.
Previous genetic study of GDM is to find candidate genes that was based on biological plausibility [27]. Recently, genome-wide association analysis studies were performed to identify some susceptibility genes associated with GDM [4]. The genetic variants of candidate genes have been revealed to contribute to the risk of GDM. For example, rs12255372 variant in Transcription factor 7-like 2 was indicated to interact with adiposity to alter β-cell function in 132 Mexican-American families with GDM [28]. The homozygosity for G972R polymorphism in Insulin receptor substrate-1 might indicate an increased risk for GDM in Saudi women [29]. There was also was significantly associated with genotypes and alleles of the CC chemokine ligand 2 rs1024611 and rs4586 polymorphisms [18] and GDM. Interestingly, many GDM associated candidate genes can express cytokines implicated in the inflammatory conditions during pregnancy [30].
GDM is characterized by varying degrees of hyperglycemia due to the inability of pancreatic β-cells to adequately respond to the increased insulin requirements during the second and third trimester [31, 32]. The etiology of GDM may be explained by many factors including cytokines, hormones, lifestyle as well as genetic disposition [33]. TGF-β isoforms are multifunctional factors that regulate embryonic development, immunity, and epithelial homeostasis [34]. Genetic polymorphisms of TGF-β isoforms were linked with an increased likelihood of having GDM and complications such as PE and diabetic nephropathy [15, 35]. With such attributes, we chose TGF-β1 and TGF-β3 as target genes to uncover the genetic disposition of GDM.
TGF-β1 is reported to be a key cytokine in insulin resistance and obesity. Over-expression of TGF-β1 can lead to decreased β-cell mass and insulin secretion [36]. TGF-β1 rs4803455 polymorphism is an A/C single-nucleotide variation on chromosome 19q13.2 and can alter the expression of insulin receptor substrate 2 associated with insulin resistant in GDM, but not depending on its expression in the pathway [37]. Moreover, a previous study suggested that TGF-β1 rs4803455 showed the effectiveness to capture the associations with cancer risk [38]. However, our data revealed that TGF-β1 rs4803455 was not a significant risk factor of GDM in the Chinese Population. The difference between these studies could be attributed to the discordance of population genetic background. However, the finite sample size in these studies is another limiting factor to have a coincident conclusion.
This is the first study to show the relationship between the genetic polymorphism of TGF-β3 gene and GDM. Candidate SNPs previously described were chosen based on their location within the gene, and a tag SNP (rs2284792: A > G) selected with SNP picker using data from the Caucasian population was located within the introns of TGF-β3 [39]. Our studies revealed an effective association between the tag SNP rs2284792 and GDM risk. Besides, we confirmed that the A allele and the A allele-containing genotypes (AA and AG) were susceptible, while the G allele/GG genotype may be protective factors. TGF-βs in mammals exhibit many overlapping biological activities and appear interchangeable. TGF-β3 knock-in ameliorate inflammation due to TGF-β1 deficiency while promoting glucose tolerance [40]. Reduced TGF-β3 expression can cause hypertrophy and induce glucose intolerance [41]. Therefore, altered generation made by polymorphic variants in TGF-β3 may affect glucose homeostasis, thus leading to GDM.
GDM is a transient presentation of long-standing metabolic malfunction and may be expected to have an association with PE [42]. The pathophysiology of PE is characterized by endothelial dysfunction which may be induced by down-regulation of TGF-β signaling. TGF-β isoforms were predisposed to have obvious susceptible associations with PE and were supposed as a biomarker for assessment of PE severity [43, 44]. TGF-β1 codon 10 T/C was observed to have a higher frequency of T>C allele in Type 2 Diabetes Mellitus patients with hypertension [45]. A fetal TGF-β3 variant (rs11466414) is associated with PE in a predominantly Hispanic population [44]. In consideration of comparable clinical characteristics, we hypothesized that the variants of TGF-β isoforms may relate to the development of both disease conditions. Then, we analyzed TGF-β1 (rs4803455) and TGF-β3 (rs2284792 and rs3917201) polymorphisms among GDM cases with PE. However, no obvious difference was found in either the genotypic distributions or allelic frequencies among above three SNPs. The complexity of several pathogenic pathways including metabolic, immune, and endothelial dysfunction can account for the invalid assumption. Insulin resistance which is highly prevalent in patients with GDM can only partially explain the development of PE [42]. To sum up, TGF-β3 rs2284792 may be the independent effective genetic locus for GDM alone.

Conclusions

This study indicated that the AA and AG genotype rs2284792 polymorphism of TGF-β3 was associated with the increased risk of GDM. However, some evident shortcomings are the limited sample size and the different ethnic origins. Furthermore, some environmental factors, such as behavioral and pharmacological interventions, will be considered in our future studies. All these studies highlight the need of long-term cohort studies of women with GDM for ultimately improving pregnancy outcomes.

Acknowledgments

Not applicable.
Informed consent was issued and signed by all subjects and all investigations were approved by the ethics committee of the Affiliated Hospital of Qingdao University.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Lowe WL Jr, Scholtens DM, Sandler V, Hayes MG. Genetics of Gestational Diabetes Mellitus and Maternal Metabolism. Curr Diabetes Rep. 2016;16(2):15.CrossRef Lowe WL Jr, Scholtens DM, Sandler V, Hayes MG. Genetics of Gestational Diabetes Mellitus and Maternal Metabolism. Curr Diabetes Rep. 2016;16(2):15.CrossRef
2.
Zurück zum Zitat Johns EC, Denison FC, Norman JE, Reynolds RM. Gestational diabetes mellitus: mechanisms, treatment, and complications. Trends Endocrinol Metab. 2018;29(11):743–54.PubMedCrossRef Johns EC, Denison FC, Norman JE, Reynolds RM. Gestational diabetes mellitus: mechanisms, treatment, and complications. Trends Endocrinol Metab. 2018;29(11):743–54.PubMedCrossRef
3.
Zurück zum Zitat Chiefari E, Arcidiacono B, Foti D, Brunetti A. Gestational diabetes mellitus: an updated overview. J Endocrinol Investig. 2017;40:899–909.CrossRef Chiefari E, Arcidiacono B, Foti D, Brunetti A. Gestational diabetes mellitus: an updated overview. J Endocrinol Investig. 2017;40:899–909.CrossRef
4.
Zurück zum Zitat Zhang C, Bao W, Rong Y, Yang H, Bowers K, Yeung E, Kiely M. Genetic variants and the risk of gestational diabetes mellitus: a systematic review. Hum Reprod Update. 2013;19(4):376–90.PubMedPubMedCentralCrossRef Zhang C, Bao W, Rong Y, Yang H, Bowers K, Yeung E, Kiely M. Genetic variants and the risk of gestational diabetes mellitus: a systematic review. Hum Reprod Update. 2013;19(4):376–90.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Enninga EAL, Egan AM, Alrahmani L, Leontovich AA, Ruano R, Sarras MP Jr. Frequency of Gestational Diabetes Mellitus Reappearance or Absence during the Second Pregnancy of Women Treated at Mayo Clinic between 2013 and 2018. J Diabetes Res. 2019;2019:9583927.PubMedPubMedCentralCrossRef Enninga EAL, Egan AM, Alrahmani L, Leontovich AA, Ruano R, Sarras MP Jr. Frequency of Gestational Diabetes Mellitus Reappearance or Absence during the Second Pregnancy of Women Treated at Mayo Clinic between 2013 and 2018. J Diabetes Res. 2019;2019:9583927.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Buchanan TA, Xiang AH, Page KA. Gestational diabetes mellitus: risks and management during and after pregnancy. Nat Rev Endocrinol. 2012;8(11):639–49.PubMedPubMedCentralCrossRef Buchanan TA, Xiang AH, Page KA. Gestational diabetes mellitus: risks and management during and after pregnancy. Nat Rev Endocrinol. 2012;8(11):639–49.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Agha-Jaffar R, Oliver N, Johnston D, Robinson S. Gestational diabetes mellitus: does an effective prevention strategy exist? Nat Rev Endocrinol. 2016;12(9):533–46.PubMedCrossRef Agha-Jaffar R, Oliver N, Johnston D, Robinson S. Gestational diabetes mellitus: does an effective prevention strategy exist? Nat Rev Endocrinol. 2016;12(9):533–46.PubMedCrossRef
8.
Zurück zum Zitat Weissgerber TL, Mudd LM. Preeclampsia and diabetes. Curr Diabetes Rep. 2015;15(3):9.CrossRef Weissgerber TL, Mudd LM. Preeclampsia and diabetes. Curr Diabetes Rep. 2015;15(3):9.CrossRef
9.
Zurück zum Zitat Syngelaki A, Kotecha R, Pastides A, Wright A, Nicolaides KH. First-trimester biochemical markers of placentation in screening for gestational diabetes mellitus. Metabolism. 2015;64(11):1485–9.PubMedCrossRef Syngelaki A, Kotecha R, Pastides A, Wright A, Nicolaides KH. First-trimester biochemical markers of placentation in screening for gestational diabetes mellitus. Metabolism. 2015;64(11):1485–9.PubMedCrossRef
10.
Zurück zum Zitat Alyas S, Roohi N, Ashraf S, Ilyas S, Ilyas A. Early pregnancy biochemical markers of placentation for screening of gestational diabetes mellitus (GDM). Diabetes Metab Syndr. 2019;13(4):2353–6.PubMedCrossRef Alyas S, Roohi N, Ashraf S, Ilyas S, Ilyas A. Early pregnancy biochemical markers of placentation for screening of gestational diabetes mellitus (GDM). Diabetes Metab Syndr. 2019;13(4):2353–6.PubMedCrossRef
11.
Zurück zum Zitat Jones RL, Stoikos C, Findlay JK. L.a. Salamonsen.TGF-beta superfamily expression and actions in the endometrium and placenta. Reproduction. 2006;132(2):217–32.PubMedCrossRef Jones RL, Stoikos C, Findlay JK. L.a. Salamonsen.TGF-beta superfamily expression and actions in the endometrium and placenta. Reproduction. 2006;132(2):217–32.PubMedCrossRef
12.
Zurück zum Zitat Perera N, Ritchie RH, Tate M. The Role of Bone Morphogenetic Proteins in Diabetic Complications. ACS Pharmacol Transl Sci. 2020;3(1):11–20.PubMedCrossRef Perera N, Ritchie RH, Tate M. The Role of Bone Morphogenetic Proteins in Diabetic Complications. ACS Pharmacol Transl Sci. 2020;3(1):11–20.PubMedCrossRef
13.
Zurück zum Zitat Thadhani R, Powe CE, Tjoa ML, Khankin E, Ye J, Ecker J, Schneyer A, Karumanchi SA. First-trimester follistatin-like-3 levels in pregnancies complicated by subsequent gestational diabetes mellitus. Diabetes Care. 2010;33(3):664–9.PubMedCrossRef Thadhani R, Powe CE, Tjoa ML, Khankin E, Ye J, Ecker J, Schneyer A, Karumanchi SA. First-trimester follistatin-like-3 levels in pregnancies complicated by subsequent gestational diabetes mellitus. Diabetes Care. 2010;33(3):664–9.PubMedCrossRef
14.
Zurück zum Zitat Yadav H, Quijano C, Kamaraju AK, Gavrilova O, Malek R, Chen W, Zerfas P, Zhigang D, Wright EC, Stuelten C, et al. Protection from obesity and diabetes by blockade of TGF-beta/Smad3 signaling. Cell Metab. 2011;14(1):67–79.PubMedPubMedCentralCrossRef Yadav H, Quijano C, Kamaraju AK, Gavrilova O, Malek R, Chen W, Zerfas P, Zhigang D, Wright EC, Stuelten C, et al. Protection from obesity and diabetes by blockade of TGF-beta/Smad3 signaling. Cell Metab. 2011;14(1):67–79.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat El-Sherbini SM, Shahen SM, Mosaad YM, Abdelgawad MS, Talaat RM. Gene polymorphism of transforming growth factor-beta1 in Egyptian patients with type 2 diabetes and diabetic nephropathy. Acta Biochim Biophys Sin Shanghai. 2013;45(4):330–8.PubMedCrossRef El-Sherbini SM, Shahen SM, Mosaad YM, Abdelgawad MS, Talaat RM. Gene polymorphism of transforming growth factor-beta1 in Egyptian patients with type 2 diabetes and diabetic nephropathy. Acta Biochim Biophys Sin Shanghai. 2013;45(4):330–8.PubMedCrossRef
16.
Zurück zum Zitat Cao M, Zhang L, Chen T, Shi A, Xie K, Li Z, Xu J, Chen Z, Ji C, Wen J. Genetic Susceptibility to Gestational Diabetes Mellitus in a Chinese Population. Front Endocrinol (Lausanne). 2020;11:247.CrossRef Cao M, Zhang L, Chen T, Shi A, Xie K, Li Z, Xu J, Chen Z, Ji C, Wen J. Genetic Susceptibility to Gestational Diabetes Mellitus in a Chinese Population. Front Endocrinol (Lausanne). 2020;11:247.CrossRef
18.
Zurück zum Zitat Teler J, Tarnowski M, Safranow K, Maciejewska A, Sawczuk M, Dziedziejko V, Sluczanowska-Glabowska S, Pawlik A. CCL2, CCL5, IL4 and IL15 gene polymorphisms in women with gestational diabetes mellitus. Horm Metab Res. 2017;49(1):10–5.PubMed Teler J, Tarnowski M, Safranow K, Maciejewska A, Sawczuk M, Dziedziejko V, Sluczanowska-Glabowska S, Pawlik A. CCL2, CCL5, IL4 and IL15 gene polymorphisms in women with gestational diabetes mellitus. Horm Metab Res. 2017;49(1):10–5.PubMed
19.
Zurück zum Zitat Gomes CP, Torloni MR, Gueuvoghlanian-Silva BY, Alexandre SM, Mattar R, Daher S. Cytokine levels in gestational diabetes mellitus: a systematic review of the literature. Am J Reprod Immunol. 2013;69(6):545–57.PubMed Gomes CP, Torloni MR, Gueuvoghlanian-Silva BY, Alexandre SM, Mattar R, Daher S. Cytokine levels in gestational diabetes mellitus: a systematic review of the literature. Am J Reprod Immunol. 2013;69(6):545–57.PubMed
20.
Zurück zum Zitat Gao Y, Zhang R, Dai S, Zhang X, Li X, Bai C. Role of TGF-beta/Smad pathway in the transcription of pancreas-specific genes during Beta cell differentiation. Front Cell Dev Biol. 2019;7:351.PubMedPubMedCentralCrossRef Gao Y, Zhang R, Dai S, Zhang X, Li X, Bai C. Role of TGF-beta/Smad pathway in the transcription of pancreas-specific genes during Beta cell differentiation. Front Cell Dev Biol. 2019;7:351.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Yener S, Demir T, Akinci B, Bayraktar F, Kebapcilar L, Ozcan MA, Biberoglu S, Yesil S. Transforming growth factor-beta 1 levels in women with prior history of gestational diabetes mellitus. Diabetes Res Clin Pract. 2007;76(2):193–8.PubMedCrossRef Yener S, Demir T, Akinci B, Bayraktar F, Kebapcilar L, Ozcan MA, Biberoglu S, Yesil S. Transforming growth factor-beta 1 levels in women with prior history of gestational diabetes mellitus. Diabetes Res Clin Pract. 2007;76(2):193–8.PubMedCrossRef
22.
Zurück zum Zitat Marcantoni E, Dovizio M, O'Gaora P, Di Francesco L, Bendaya I, Schiavone S, Trenti A, Guillem-Llobat P, Zambon A, Nardelli GB, et al. Dysregulation of gene expression in human fetal endothelial cells from gestational diabetes in response to TGF-beta1. Prostaglandins Other Lipid Mediat. 2015;120:103–14.PubMedCrossRef Marcantoni E, Dovizio M, O'Gaora P, Di Francesco L, Bendaya I, Schiavone S, Trenti A, Guillem-Llobat P, Zambon A, Nardelli GB, et al. Dysregulation of gene expression in human fetal endothelial cells from gestational diabetes in response to TGF-beta1. Prostaglandins Other Lipid Mediat. 2015;120:103–14.PubMedCrossRef
23.
Zurück zum Zitat Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim YM, Bdolah Y, Lim KH, Yuan HT, Libermann TA, et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med. 2006;12(6):642–9.PubMedCrossRef Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim YM, Bdolah Y, Lim KH, Yuan HT, Libermann TA, et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med. 2006;12(6):642–9.PubMedCrossRef
24.
Zurück zum Zitat Slattery ML, Herrick JS, Lundgreen A, Wolff RK. Genetic variation in the TGF-beta signaling pathway and colon and rectal cancer risk. Cancer Epidemiol Biomark Prev. 2011;20(1):57–69.CrossRef Slattery ML, Herrick JS, Lundgreen A, Wolff RK. Genetic variation in the TGF-beta signaling pathway and colon and rectal cancer risk. Cancer Epidemiol Biomark Prev. 2011;20(1):57–69.CrossRef
25.
Zurück zum Zitat Li X, Tan H, Chen M, Zhou S. Transforming growth factor beta 1 related gene polymorphisms in gestational hypertension and preeclampsia: a case-control candidate gene association study. Pregnancy Hypertens. 2018;12:155–60.PubMedCrossRef Li X, Tan H, Chen M, Zhou S. Transforming growth factor beta 1 related gene polymorphisms in gestational hypertension and preeclampsia: a case-control candidate gene association study. Pregnancy Hypertens. 2018;12:155–60.PubMedCrossRef
26.
Zurück zum Zitat Slattery ML, Trivellas A, Pellatt AJ, Mullany LE, Stevens JR, Wolff RK, Herrick JS. Genetic variants in the TGFbeta-signaling pathway influence expression of miRNAs in colon and rectal normal mucosa and tumor tissue. Oncotarget. 2017;8(10):16765–83.PubMedPubMedCentralCrossRef Slattery ML, Trivellas A, Pellatt AJ, Mullany LE, Stevens JR, Wolff RK, Herrick JS. Genetic variants in the TGFbeta-signaling pathway influence expression of miRNAs in colon and rectal normal mucosa and tumor tissue. Oncotarget. 2017;8(10):16765–83.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Yuen L, Wong VW, Simmons D. Ethnic disparities in gestational diabetes. Curr Diab Rep. 2018;18(9):68.PubMedCrossRef Yuen L, Wong VW, Simmons D. Ethnic disparities in gestational diabetes. Curr Diab Rep. 2018;18(9):68.PubMedCrossRef
28.
Zurück zum Zitat Watanabe RM, Allayee H, Xiang AH, Trigo E, Hartiala J, Lawrence JM, Buchanan TA. Transcription factor 7-like 2 (TCF7L2) is associated with gestational diabetes mellitus and interacts with adiposity to alter insulin secretion in Mexican Americans. Diabetes. 2007;56(5):1481–5.PubMedCrossRef Watanabe RM, Allayee H, Xiang AH, Trigo E, Hartiala J, Lawrence JM, Buchanan TA. Transcription factor 7-like 2 (TCF7L2) is associated with gestational diabetes mellitus and interacts with adiposity to alter insulin secretion in Mexican Americans. Diabetes. 2007;56(5):1481–5.PubMedCrossRef
29.
Zurück zum Zitat 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 Int. 2014;2014:146495.PubMedPubMedCentralCrossRef 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 Int. 2014;2014:146495.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Wedekind L, Belkacemi L. Altered cytokine network in gestational diabetes mellitus affects maternal insulin and placental-fetal development. J Diabetes Complicat. 2016;30(7):1393–400.CrossRef Wedekind L, Belkacemi L. Altered cytokine network in gestational diabetes mellitus affects maternal insulin and placental-fetal development. J Diabetes Complicat. 2016;30(7):1393–400.CrossRef
32.
Zurück zum Zitat Kampmann U, Madsen LR, Skajaa GO, Iversen DS, Moeller N, Ovesen P. Gestational diabetes: A clinical update. World J Diabetes. 2015;6(8):1065–72.PubMedPubMedCentralCrossRef Kampmann U, Madsen LR, Skajaa GO, Iversen DS, Moeller N, Ovesen P. Gestational diabetes: A clinical update. World J Diabetes. 2015;6(8):1065–72.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Fernandez-Real JM, Pickup JC. Innate immunity, insulin resistance and type 2 diabetes. Diabetologia. 2012;55(2):273–8.PubMedCrossRef Fernandez-Real JM, Pickup JC. Innate immunity, insulin resistance and type 2 diabetes. Diabetologia. 2012;55(2):273–8.PubMedCrossRef
34.
Zurück zum Zitat Yamanaka Y, Friess H, Buchler M, Beger HG, Gold LI, Korc M. Synthesis and expression of transforming growth factor beta-1, beta-2, and beta-3 in the endocrine and exocrine pancreas. Diabetes. 1993;42(5):746–56.PubMedCrossRef Yamanaka Y, Friess H, Buchler M, Beger HG, Gold LI, Korc M. Synthesis and expression of transforming growth factor beta-1, beta-2, and beta-3 in the endocrine and exocrine pancreas. Diabetes. 1993;42(5):746–56.PubMedCrossRef
35.
Zurück zum Zitat Conti E, Zezza L, Ralli E, Caserta D, Musumeci MB, Moscarini M, Autore C, Volpe M. Growth factors in preeclampsia: a vascular disease model. A failed vasodilation and angiogenic challenge from pregnancy onwards? Cytokine Growth Factor Rev. 2013;24(5):411–25.PubMedCrossRef Conti E, Zezza L, Ralli E, Caserta D, Musumeci MB, Moscarini M, Autore C, Volpe M. Growth factors in preeclampsia: a vascular disease model. A failed vasodilation and angiogenic challenge from pregnancy onwards? Cytokine Growth Factor Rev. 2013;24(5):411–25.PubMedCrossRef
36.
Zurück zum Zitat Yadav H, Devalaraja S, Chung ST, Rane SG. TGF-beta1/Smad3 pathway targets PP2A-AMPK-FoxO1 signaling to regulate hepatic gluconeogenesis. J Biol Chem. 2017;292(8):3420–32.PubMedPubMedCentralCrossRef Yadav H, Devalaraja S, Chung ST, Rane SG. TGF-beta1/Smad3 pathway targets PP2A-AMPK-FoxO1 signaling to regulate hepatic gluconeogenesis. J Biol Chem. 2017;292(8):3420–32.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Slattery ML, Pellatt DF, Wolff RK, Lundgreen A. Genes, environment and gene expression in colon tissue: a pathway approach to determining functionality. Int J Mol Epidemiol Genet. 2016;7(1):45–57.PubMedPubMedCentral Slattery ML, Pellatt DF, Wolff RK, Lundgreen A. Genes, environment and gene expression in colon tissue: a pathway approach to determining functionality. Int J Mol Epidemiol Genet. 2016;7(1):45–57.PubMedPubMedCentral
38.
Zurück zum Zitat Ayala de Miguel P, Enguix-Riego MV, Cacicedo J, Delgado BD, Perez M, Praena-Fernandez JM, Quintana Cortes L, Borrega Garcia P, Del Campo ER, Lopez Guerra JL. Prognostic value of the TGFbeta1 rs4803455 single nucleotide polymorphism in small cell lung cancer. Tumori. 2020:300891620946841. https://doi.org/10.1177/0300891620946841. Ayala de Miguel P, Enguix-Riego MV, Cacicedo J, Delgado BD, Perez M, Praena-Fernandez JM, Quintana Cortes L, Borrega Garcia P, Del Campo ER, Lopez Guerra JL. Prognostic value of the TGFbeta1 rs4803455 single nucleotide polymorphism in small cell lung cancer. Tumori. 2020:300891620946841. https://​doi.​org/​10.​1177/​0300891620946841​.
39.
Zurück zum Zitat Drozdzik M, Kaczmarek M, Malinowski D, Bros U, Kazienko A, Kurzawa R, Kurzawski M. TGFbeta3 (TGFB3) polymorphism is associated with male infertility. Sci Rep. 2015;5:17151.PubMedPubMedCentralCrossRef Drozdzik M, Kaczmarek M, Malinowski D, Bros U, Kazienko A, Kurzawa R, Kurzawski M. TGFbeta3 (TGFB3) polymorphism is associated with male infertility. Sci Rep. 2015;5:17151.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Hall BE, Wankhade UD, Konkel JE, Cherukuri K, Nagineni CN, Flanders KC, Arany PR, Chen W, Rane SG, Kulkarni AB. Transforming growth factor-beta3 (TGF-beta3) knock-in ameliorates inflammation due to TGF-beta1 deficiency while promoting glucose tolerance. J Biol Chem. 2013;288(44):32074–92.PubMedPubMedCentralCrossRef Hall BE, Wankhade UD, Konkel JE, Cherukuri K, Nagineni CN, Flanders KC, Arany PR, Chen W, Rane SG, Kulkarni AB. Transforming growth factor-beta3 (TGF-beta3) knock-in ameliorates inflammation due to TGF-beta1 deficiency while promoting glucose tolerance. J Biol Chem. 2013;288(44):32074–92.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Petrus P, Mejhert N, Corrales P, Lecoutre S, Li Q, Maldonado E, Kulyte A, Lopez Y, Campbell M, Acosta JR, et al. Transforming growth factor-beta3 regulates adipocyte number in subcutaneous white adipose tissue. Cell Rep. 2018;25(3):551–60 e5.PubMedCrossRef Petrus P, Mejhert N, Corrales P, Lecoutre S, Li Q, Maldonado E, Kulyte A, Lopez Y, Campbell M, Acosta JR, et al. Transforming growth factor-beta3 regulates adipocyte number in subcutaneous white adipose tissue. Cell Rep. 2018;25(3):551–60 e5.PubMedCrossRef
42.
Zurück zum Zitat Carpenter MW. Gestational diabetes, pregnancy hypertension, and late vascular disease. Diabetes Care. 2007;30(Suppl 2):S246–50.PubMedCrossRef Carpenter MW. Gestational diabetes, pregnancy hypertension, and late vascular disease. Diabetes Care. 2007;30(Suppl 2):S246–50.PubMedCrossRef
43.
Zurück zum Zitat Lim JH, Kim SY, Park SY, Lee MH, Yang JH, Kim MY, Chung JH, Lee SW, Ryu HM. Soluble endoglin and transforming growth factor-beta1 in women who subsequently developed preeclampsia. Prenat Diagn. 2009;29(5):471–6.PubMedCrossRef Lim JH, Kim SY, Park SY, Lee MH, Yang JH, Kim MY, Chung JH, Lee SW, Ryu HM. Soluble endoglin and transforming growth factor-beta1 in women who subsequently developed preeclampsia. Prenat Diagn. 2009;29(5):471–6.PubMedCrossRef
44.
Zurück zum Zitat Wilson ML, Desmond DH, Goodwin TM, Miller DA, Ingles SA. Maternal and fetal variants in the TGF-beta3 gene and risk of pregnancy-induced hypertension in a predominantly Latino population. Am J Obstet Gynecol. 2009;201(3):295 e1–5.CrossRef Wilson ML, Desmond DH, Goodwin TM, Miller DA, Ingles SA. Maternal and fetal variants in the TGF-beta3 gene and risk of pregnancy-induced hypertension in a predominantly Latino population. Am J Obstet Gynecol. 2009;201(3):295 e1–5.CrossRef
45.
Zurück zum Zitat Ramirez A, Hernandez M, Suarez-Sanchez R, Ortega C, Peralta J, Gomez J, Valladares A, Cruz M, Vazquez-Moreno MA. F. Suarez-Sanchez.Type 2 diabetes-associated polymorphisms correlate with SIRT1 and TGF-beta1 gene expression. Ann Hum Genet. 2020;84(2):185–94.PubMedCrossRef Ramirez A, Hernandez M, Suarez-Sanchez R, Ortega C, Peralta J, Gomez J, Valladares A, Cruz M, Vazquez-Moreno MA. F. Suarez-Sanchez.Type 2 diabetes-associated polymorphisms correlate with SIRT1 and TGF-beta1 gene expression. Ann Hum Genet. 2020;84(2):185–94.PubMedCrossRef
Metadaten
Titel
Polymorphisms of TGF-β1 and TGF-β3 in Chinese women with gestational diabetes mellitus
verfasst von
Yinglei Xu
Chunlian Wei
Cuijiao Wu
Mengmeng Han
Jingli Wang
Huabin Hou
Lu Zhang
Shiguo Liu
Ying Chen
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Pregnancy and Childbirth / Ausgabe 1/2020
Elektronische ISSN: 1471-2393
DOI
https://doi.org/10.1186/s12884-020-03459-w

Weitere Artikel der Ausgabe 1/2020

BMC Pregnancy and Childbirth 1/2020 Zur Ausgabe

Hirsutismus bei PCOS: Laser- und Lichttherapien helfen

26.04.2024 Hirsutismus Nachrichten

Laser- und Lichtbehandlungen können bei Frauen mit polyzystischem Ovarialsyndrom (PCOS) den übermäßigen Haarwuchs verringern und das Wohlbefinden verbessern – bei alleiniger Anwendung oder in Kombination mit Medikamenten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Weniger postpartale Depressionen nach Esketamin-Einmalgabe

Bislang gibt es kein Medikament zur Prävention von Wochenbettdepressionen. Das Injektionsanästhetikum Esketamin könnte womöglich diese Lücke füllen.

Bei RSV-Impfung vor 60. Lebensjahr über Off-Label-Gebrauch aufklären!

22.04.2024 DGIM 2024 Kongressbericht

Durch die Häufung nach der COVID-19-Pandemie sind Infektionen mit dem Respiratorischen Synzytial-Virus (RSV) in den Fokus gerückt. Fachgesellschaften empfehlen eine Impfung inzwischen nicht nur für Säuglinge und Kleinkinder.

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.