Skip to main content
Erschienen in: Journal of Endocrinological Investigation 8/2023

01.08.2023 | Review

Sex differences in glycolipidic disorders after exposure to maternal hyperglycemia during early development

verfasst von: S.-s. Luo, H. Zhu, H-f. Huang, G.-l. Ding

Erschienen in: Journal of Endocrinological Investigation | Ausgabe 8/2023

Einloggen, um Zugang zu erhalten

Abstract

Purpose

The aim of this review was to summarize sex differences in glycolipid metabolic phenotypes of human and animal models after exposure to maternal hyperglycemia and overview the underlying mechanisms, providing a new perspective on the maternal hyperglycemia-triggered risk of glycolipidic disorders in offspring.

Methods

A comprehensive literature search within PubMed was performed. Selected publications related to studies on offspring exposed to maternal hyperglycemia investigating the sex differences of glycolipid metabolism were reviewed.

Results

Maternal hyperglycemia increases the risk of glycolipid metabolic disorders in offspring, such as obesity, glucose intolerance and diabetes. Whether with or without intervention, metabolic phenotypes have been shown to exhibit sex differences between male and female offspring in response to maternal hyperglycemia, which may be related to gonadal hormones, organic intrinsic differences, placenta, and epigenetic modifications.

Conclusion

Sex may play a role in the different incidences and pathogenesis of abnormal glycolipid metabolism. More studies investigating both sexes are needed to understand how and why environmental conditions in early life affect long-term health between male and female individuals.
Literatur
1.
Zurück zum Zitat Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, Stein C, Basit A, Chan JCN, Mbanya JC, Pavkov ME, Ramachandaran A, Wild SH, James S, Herman WH, Zhang P, Bommer C, Kuo S, Boyko EJ, Magliano DJ (2022) IDF diabetes atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract 183:109119PubMedCrossRef Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, Stein C, Basit A, Chan JCN, Mbanya JC, Pavkov ME, Ramachandaran A, Wild SH, James S, Herman WH, Zhang P, Bommer C, Kuo S, Boyko EJ, Magliano DJ (2022) IDF diabetes atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract 183:109119PubMedCrossRef
2.
Zurück zum Zitat Lowe WL, Scholtens DM, Kuang A, Linder B, Lawrence JM, Lebenthal Y, McCance D, Hamilton J, Nodzenski M, Talbot O, Brickman WJ, Clayton P, Ma RC, Tam WH, Dyer AR, Catalano PM, Lowe LP, Metzger BE (2019) Hyperglycemia and adverse pregnancy outcome follow-up Study (HAPO FUS): maternal gestational diabetes mellitus and childhood glucose metabolism. Diabetes Care 42:372–380PubMedPubMedCentralCrossRef Lowe WL, Scholtens DM, Kuang A, Linder B, Lawrence JM, Lebenthal Y, McCance D, Hamilton J, Nodzenski M, Talbot O, Brickman WJ, Clayton P, Ma RC, Tam WH, Dyer AR, Catalano PM, Lowe LP, Metzger BE (2019) Hyperglycemia and adverse pregnancy outcome follow-up Study (HAPO FUS): maternal gestational diabetes mellitus and childhood glucose metabolism. Diabetes Care 42:372–380PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Boerschmann H, Pflüger M, Henneberger L, Ziegler A-G, Hummel S (2010) Prevalence and predictors of overweight and insulin resistance in offspring of mothers with gestational diabetes mellitus. Diabetes Care 33:1845–1849PubMedPubMedCentralCrossRef Boerschmann H, Pflüger M, Henneberger L, Ziegler A-G, Hummel S (2010) Prevalence and predictors of overweight and insulin resistance in offspring of mothers with gestational diabetes mellitus. Diabetes Care 33:1845–1849PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Vääräsmäki M, Pouta A, Elliot P, Tapanainen P, Sovio U, Ruokonen A, Hartikainen A-L, McCarthy M, Järvelin M-R (2009) Adolescent manifestations of metabolic syndrome among children born to women with gestational diabetes in a general-population birth cohort. Am J Epidemiol 169:1209–1215PubMedCrossRef Vääräsmäki M, Pouta A, Elliot P, Tapanainen P, Sovio U, Ruokonen A, Hartikainen A-L, McCarthy M, Järvelin M-R (2009) Adolescent manifestations of metabolic syndrome among children born to women with gestational diabetes in a general-population birth cohort. Am J Epidemiol 169:1209–1215PubMedCrossRef
5.
Zurück zum Zitat Ding G-L, Wang F-F, Shu J, Tian S, Jiang Y, Zhang D, Wang N, Luo Q, Zhang Y, Jin F, Leung PCK, Sheng J-Z, Huang H-F (2012) Transgenerational glucose intolerance with Igf2/H19 epigenetic alterations in mouse islet induced by intrauterine hyperglycemia. Diabetes 61:1133–1142PubMedPubMedCentralCrossRef Ding G-L, Wang F-F, Shu J, Tian S, Jiang Y, Zhang D, Wang N, Luo Q, Zhang Y, Jin F, Leung PCK, Sheng J-Z, Huang H-F (2012) Transgenerational glucose intolerance with Igf2/H19 epigenetic alterations in mouse islet induced by intrauterine hyperglycemia. Diabetes 61:1133–1142PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Zhu H, Chen B, Cheng Y, Zhou Y, Yan YS, Luo Q, Jiang Y, Sheng JZ, Ding GL, Huang HF (2019) Insulin therapy for gestational diabetes mellitus does not fully protect offspring from diet-induced metabolic disorders. Diabetes 68:696–708PubMedCrossRef Zhu H, Chen B, Cheng Y, Zhou Y, Yan YS, Luo Q, Jiang Y, Sheng JZ, Ding GL, Huang HF (2019) Insulin therapy for gestational diabetes mellitus does not fully protect offspring from diet-induced metabolic disorders. Diabetes 68:696–708PubMedCrossRef
7.
Zurück zum Zitat van Dijk SJ, Tellam RL, Morrison JL, Muhlhausler BS, Molloy PL (2015) Recent developments on the role of epigenetics in obesity and metabolic disease. Clin Epigenetics 7:66PubMedPubMedCentralCrossRef van Dijk SJ, Tellam RL, Morrison JL, Muhlhausler BS, Molloy PL (2015) Recent developments on the role of epigenetics in obesity and metabolic disease. Clin Epigenetics 7:66PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Perng W, Kelsey MM, Sauder KA, Dabelea D (2021) How does exposure to overnutrition in utero lead to childhood adiposity? Testing the insulin hypersecretion hypothesis in the EPOCH cohort. Diabetologia 64:2237–2246PubMedCrossRef Perng W, Kelsey MM, Sauder KA, Dabelea D (2021) How does exposure to overnutrition in utero lead to childhood adiposity? Testing the insulin hypersecretion hypothesis in the EPOCH cohort. Diabetologia 64:2237–2246PubMedCrossRef
9.
Zurück zum Zitat Hockett CW, Harrall KK, Moore BF, Starling AP, Bellatorre A, Sauder KA, Perng W, Scherzinger A, Garg K, Ringham BM, Glueck DH, Dabelea D (2019) Persistent effects of in utero overnutrition on offspring adiposity: the exploring perinatal outcomes among Children (EPOCH) study. Diabetologia 62:2017–2024PubMedPubMedCentralCrossRef Hockett CW, Harrall KK, Moore BF, Starling AP, Bellatorre A, Sauder KA, Perng W, Scherzinger A, Garg K, Ringham BM, Glueck DH, Dabelea D (2019) Persistent effects of in utero overnutrition on offspring adiposity: the exploring perinatal outcomes among Children (EPOCH) study. Diabetologia 62:2017–2024PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat A. Bellatorre, A. Scherzinger, E. Stamm, M. Martinez, B. Ringham, and D. Dabelea, (2018) Fetal Overnutrition and Adolescent Hepatic Fat Fraction: the Exploring Perinatal Outcomes in Children Study. J Pediatr 192. A. Bellatorre, A. Scherzinger, E. Stamm, M. Martinez, B. Ringham, and D. Dabelea, (2018) Fetal Overnutrition and Adolescent Hepatic Fat Fraction: the Exploring Perinatal Outcomes in Children Study. J Pediatr 192.
11.
Zurück zum Zitat Lefebvre P, Staels B (2021) Hepatic sexual dimorphism - implications for non-alcoholic fatty liver disease. Nat Rev Endocrinol 17:662–670PubMedCrossRef Lefebvre P, Staels B (2021) Hepatic sexual dimorphism - implications for non-alcoholic fatty liver disease. Nat Rev Endocrinol 17:662–670PubMedCrossRef
12.
Zurück zum Zitat M. Schiffrin, C. Winkler, L. Quignodon, A. Naldi, M. Trötzmüller, H. Köfeler, H. Henry, P. Parini, B. Desvergne, and F. Gilardi, (2021) Sex Dimorphism of Nonalcoholic Fatty Liver Disease (NAFLD) in -Null Mice. Int J Mol Sci 22. M. Schiffrin, C. Winkler, L. Quignodon, A. Naldi, M. Trötzmüller, H. Köfeler, H. Henry, P. Parini, B. Desvergne, and F. Gilardi, (2021) Sex Dimorphism of Nonalcoholic Fatty Liver Disease (NAFLD) in -Null Mice. Int J Mol Sci 22.
13.
Zurück zum Zitat Shaheen M, Schrode KM, Pan D, Kermah D, Puri V, Zarrinpar A, Elisha D, Najjar SM, Friedman TC (2021) Sex-specific differences in the association between race/ethnicity and NAFLD among US population. Front Med (Lausanne) 8:795421PubMedCrossRef Shaheen M, Schrode KM, Pan D, Kermah D, Puri V, Zarrinpar A, Elisha D, Najjar SM, Friedman TC (2021) Sex-specific differences in the association between race/ethnicity and NAFLD among US population. Front Med (Lausanne) 8:795421PubMedCrossRef
14.
Zurück zum Zitat Cooper AJ, Gupta SR, Moustafa AF, Chao AM (2021) Sex/gender differences in obesity prevalence, comorbidities, and treatment. Curr Obes Rep 10:458–466PubMedCrossRef Cooper AJ, Gupta SR, Moustafa AF, Chao AM (2021) Sex/gender differences in obesity prevalence, comorbidities, and treatment. Curr Obes Rep 10:458–466PubMedCrossRef
15.
Zurück zum Zitat Tsai S-Y, Rodriguez AA, Dastidar SG, Del Greco E, Carr KL, Sitzmann JM, Academia EC, Viray CM, Martinez LL, Kaplowitz BS, Ashe TD, La Spada AR, Kennedy BK (2016) Increased 4E-BP1 expression protects against diet-induced obesity and insulin resistance in male mice. Cell Rep 16:1903–1914PubMedPubMedCentralCrossRef Tsai S-Y, Rodriguez AA, Dastidar SG, Del Greco E, Carr KL, Sitzmann JM, Academia EC, Viray CM, Martinez LL, Kaplowitz BS, Ashe TD, La Spada AR, Kennedy BK (2016) Increased 4E-BP1 expression protects against diet-induced obesity and insulin resistance in male mice. Cell Rep 16:1903–1914PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Gao A, Su J, Liu R, Zhao S, Li W, Xu X, Li D, Shi J, Gu B, Zhang J, Li Q, Wang X, Zhang Y, Xu Y, Lu J, Ning G, Hong J, Bi Y, Gu W, Wang J, Wang W (2021) Sexual dimorphism in glucose metabolism is shaped by androgen-driven gut microbiome. Nat Commun 12:7080PubMedPubMedCentralCrossRef Gao A, Su J, Liu R, Zhao S, Li W, Xu X, Li D, Shi J, Gu B, Zhang J, Li Q, Wang X, Zhang Y, Xu Y, Lu J, Ning G, Hong J, Bi Y, Gu W, Wang J, Wang W (2021) Sexual dimorphism in glucose metabolism is shaped by androgen-driven gut microbiome. Nat Commun 12:7080PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Pettersson US, Waldén TB, Carlsson P-O, Jansson L, Phillipson M (2012) Female mice are protected against high-fat diet induced metabolic syndrome and increase the regulatory T cell population in adipose tissue. PLoS ONE 7:e46057PubMedPubMedCentralCrossRef Pettersson US, Waldén TB, Carlsson P-O, Jansson L, Phillipson M (2012) Female mice are protected against high-fat diet induced metabolic syndrome and increase the regulatory T cell population in adipose tissue. PLoS ONE 7:e46057PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Age- and sex-specific prevalences of diabetes and impaired glucose regulation in 13 European cohorts. Diabetes Care 26 (2003) 61–69. Age- and sex-specific prevalences of diabetes and impaired glucose regulation in 13 European cohorts. Diabetes Care 26 (2003) 61–69.
19.
Zurück zum Zitat Barker DJP (2007) The origins of the developmental origins theory. J Intern Med 261:412–417PubMedCrossRef Barker DJP (2007) The origins of the developmental origins theory. J Intern Med 261:412–417PubMedCrossRef
20.
Zurück zum Zitat C.N. Hales, and D.J. Barker, (2001) The thrifty phenotype hypothesis. British Medical Bulletin 60. C.N. Hales, and D.J. Barker, (2001) The thrifty phenotype hypothesis. British Medical Bulletin 60.
21.
Zurück zum Zitat Lowe WL, Lowe LP, Kuang A, Catalano PM, Nodzenski M, Talbot O, Tam W-H, Sacks DA, McCance D, Linder B, Lebenthal Y, Lawrence JM, Lashley M, Josefson JL, Hamilton J, Deerochanawong C, Clayton P, Brickman WJ, Dyer AR, Scholtens DM, Metzger BE (2019) Maternal glucose levels during pregnancy and childhood adiposity in the hyperglycemia and adverse pregnancy outcome follow-up study. Diabetologia 62:598–610PubMedPubMedCentralCrossRef Lowe WL, Lowe LP, Kuang A, Catalano PM, Nodzenski M, Talbot O, Tam W-H, Sacks DA, McCance D, Linder B, Lebenthal Y, Lawrence JM, Lashley M, Josefson JL, Hamilton J, Deerochanawong C, Clayton P, Brickman WJ, Dyer AR, Scholtens DM, Metzger BE (2019) Maternal glucose levels during pregnancy and childhood adiposity in the hyperglycemia and adverse pregnancy outcome follow-up study. Diabetologia 62:598–610PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Gillman MW, Oakey H, Baghurst PA, Volkmer RE, Robinson JS, Crowther CA (2010) Effect of treatment of gestational diabetes mellitus on obesity in the next generation. Diabetes Care 33:964–968PubMedPubMedCentralCrossRef Gillman MW, Oakey H, Baghurst PA, Volkmer RE, Robinson JS, Crowther CA (2010) Effect of treatment of gestational diabetes mellitus on obesity in the next generation. Diabetes Care 33:964–968PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Fried SK, Lee M-J, Karastergiou K (2015) Shaping fat distribution: new insights into the molecular determinants of depot- and sex-dependent adipose biology. Obesity (Silver Spring) 23:1345–1352PubMedCrossRef Fried SK, Lee M-J, Karastergiou K (2015) Shaping fat distribution: new insights into the molecular determinants of depot- and sex-dependent adipose biology. Obesity (Silver Spring) 23:1345–1352PubMedCrossRef
26.
Zurück zum Zitat Nishizawa H, Shimomura I, Kishida K, Maeda N, Kuriyama H, Nagaretani H, Matsuda M, Kondo H, Furuyama N, Kihara S, Nakamura T, Tochino Y, Funahashi T, Matsuzawa Y (2002) Androgens decrease plasma adiponectin, an insulin-sensitizing adipocyte-derived protein. Diabetes 51:2734–2741PubMedCrossRef Nishizawa H, Shimomura I, Kishida K, Maeda N, Kuriyama H, Nagaretani H, Matsuda M, Kondo H, Furuyama N, Kihara S, Nakamura T, Tochino Y, Funahashi T, Matsuzawa Y (2002) Androgens decrease plasma adiponectin, an insulin-sensitizing adipocyte-derived protein. Diabetes 51:2734–2741PubMedCrossRef
27.
Zurück zum Zitat Pereira TJ, Fonseca MA, Campbell KE, Moyce BL, Cole LK, Hatch GM, Doucette CA, Klein J, Aliani M, Dolinsky VW (2015) Maternal obesity characterized by gestational diabetes increases the susceptibility of rat offspring to hepatic steatosis via a disrupted liver metabolome. J Physiol 593:3181–3197PubMedPubMedCentralCrossRef Pereira TJ, Fonseca MA, Campbell KE, Moyce BL, Cole LK, Hatch GM, Doucette CA, Klein J, Aliani M, Dolinsky VW (2015) Maternal obesity characterized by gestational diabetes increases the susceptibility of rat offspring to hepatic steatosis via a disrupted liver metabolome. J Physiol 593:3181–3197PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Stevanović-Silva J, Beleza J, Coxito P, Pereira S, Rocha H, Gaspar TB, Gärtner F, Correia R, Martins MJ, Guimarães T, Martins S, Oliveira PJ, Ascensão A, Magalhães J (2021) Maternal high-fat high-sucrose diet and gestational exercise modulate hepatic fat accumulation and liver mitochondrial respiratory capacity in mothers and male offspring. Metabolism 116:154704PubMedCrossRef Stevanović-Silva J, Beleza J, Coxito P, Pereira S, Rocha H, Gaspar TB, Gärtner F, Correia R, Martins MJ, Guimarães T, Martins S, Oliveira PJ, Ascensão A, Magalhães J (2021) Maternal high-fat high-sucrose diet and gestational exercise modulate hepatic fat accumulation and liver mitochondrial respiratory capacity in mothers and male offspring. Metabolism 116:154704PubMedCrossRef
29.
Zurück zum Zitat Oliveira AC, Andreotti S, Chimin P, Sertié RAL, Farias TDSM, Torres-Leal FL, de Proença ARG, Campaña AB, D’Avila LSP, Oliveira KA, Lima FB (2015) Neonatal streptozotocin-induced diabetes in mothers promotes metabolic programming of adipose tissue in male rat offspring. Life Sci 136:151–156PubMedCrossRef Oliveira AC, Andreotti S, Chimin P, Sertié RAL, Farias TDSM, Torres-Leal FL, de Proença ARG, Campaña AB, D’Avila LSP, Oliveira KA, Lima FB (2015) Neonatal streptozotocin-induced diabetes in mothers promotes metabolic programming of adipose tissue in male rat offspring. Life Sci 136:151–156PubMedCrossRef
30.
Zurück zum Zitat Fornes D, Heinecke F, Roberti SL, White V, Capobianco E, Jawerbaum A (2020) Proinflammation in maternal and fetal livers and circulating miR-122 dysregulation in a GDM rat model induced by intrauterine programming. Mol Cell Endocrinol 510:110824PubMedCrossRef Fornes D, Heinecke F, Roberti SL, White V, Capobianco E, Jawerbaum A (2020) Proinflammation in maternal and fetal livers and circulating miR-122 dysregulation in a GDM rat model induced by intrauterine programming. Mol Cell Endocrinol 510:110824PubMedCrossRef
31.
Zurück zum Zitat Jiang Y, Zhu H, Chen Z, Yu Y-C, Guo X-H, Chen Y, Yang M-M, Chen B-W, Sagnelli M, Xu D, Zhao B-H, Luo Q (2022) Hepatic IGF2/H19 epigenetic alteration induced glucose intolerance in gestational diabetes mellitus offspring FoxO1 mediation. Front Endocrinol (Lausanne) 13:844707PubMedCrossRef Jiang Y, Zhu H, Chen Z, Yu Y-C, Guo X-H, Chen Y, Yang M-M, Chen B-W, Sagnelli M, Xu D, Zhao B-H, Luo Q (2022) Hepatic IGF2/H19 epigenetic alteration induced glucose intolerance in gestational diabetes mellitus offspring FoxO1 mediation. Front Endocrinol (Lausanne) 13:844707PubMedCrossRef
32.
Zurück zum Zitat Fornes D, White V, Higa R, Heinecke F, Capobianco E, Jawerbaum A (2018) Sex-dependent changes in lipid metabolism, PPAR pathways and microRNAs that target PPARs in the fetal liver of rats with gestational diabetes. Mol Cell Endocrinol 461:12–21PubMedCrossRef Fornes D, White V, Higa R, Heinecke F, Capobianco E, Jawerbaum A (2018) Sex-dependent changes in lipid metabolism, PPAR pathways and microRNAs that target PPARs in the fetal liver of rats with gestational diabetes. Mol Cell Endocrinol 461:12–21PubMedCrossRef
33.
Zurück zum Zitat Isganaitis E, Woo M, Ma H, Chen M, Kong W, Lytras A, Sales V, Decoste-Lopez J, Lee K-J, Leatherwood C, Lee D, Fitzpatrick C, Gall W, Watkins S, Patti M-E (2014) Developmental programming by maternal insulin resistance: hyperinsulinemia, glucose intolerance, and dysregulated lipid metabolism in male offspring of insulin-resistant mice. Diabetes 63:688–700PubMedPubMedCentralCrossRef Isganaitis E, Woo M, Ma H, Chen M, Kong W, Lytras A, Sales V, Decoste-Lopez J, Lee K-J, Leatherwood C, Lee D, Fitzpatrick C, Gall W, Watkins S, Patti M-E (2014) Developmental programming by maternal insulin resistance: hyperinsulinemia, glucose intolerance, and dysregulated lipid metabolism in male offspring of insulin-resistant mice. Diabetes 63:688–700PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Tam WH, Ma RCW, Ozaki R, Li AM, Chan MHM, Yuen LY, Lao TTH, Yang X, Ho CS, Tutino GE, Chan JCN (2017) In Utero exposure to maternal hyperglycemia increases childhood cardiometabolic risk in offspring. Diabetes Care 40:679–686PubMedPubMedCentralCrossRef Tam WH, Ma RCW, Ozaki R, Li AM, Chan MHM, Yuen LY, Lao TTH, Yang X, Ho CS, Tutino GE, Chan JCN (2017) In Utero exposure to maternal hyperglycemia increases childhood cardiometabolic risk in offspring. Diabetes Care 40:679–686PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Bider-Canfield Z, Martinez MP, Wang X, Yu W, Bautista MP, Brookey J, Page KA, Buchanan TA, Xiang AH (2017) Maternal obesity, gestational diabetes, breastfeeding and childhood overweight at age 2 years. Pediatr Obes 12:171–178PubMedCrossRef Bider-Canfield Z, Martinez MP, Wang X, Yu W, Bautista MP, Brookey J, Page KA, Buchanan TA, Xiang AH (2017) Maternal obesity, gestational diabetes, breastfeeding and childhood overweight at age 2 years. Pediatr Obes 12:171–178PubMedCrossRef
36.
Zurück zum Zitat Antikainen L, Jääskeläinen J, Nordman H, Voutilainen R, Huopio H (2021) Boys but not girls exposed to maternal gestational diabetes mellitus have unfavorable fat distribution. Horm Res Paediatr 94:194–200PubMedCrossRef Antikainen L, Jääskeläinen J, Nordman H, Voutilainen R, Huopio H (2021) Boys but not girls exposed to maternal gestational diabetes mellitus have unfavorable fat distribution. Horm Res Paediatr 94:194–200PubMedCrossRef
37.
Zurück zum Zitat Kaseva N, Vääräsmäki M, Matinolli HM, Sipola-Leppänen M, Tikanmäki M, Heinonen K, Lano A, Wolke D, Andersson S, Järvelin MR, Räikkönen K, Eriksson JG, Kajantie E (2018) Pre-pregnancy overweight or obesity and gestational diabetes as predictors of body composition in offspring twenty years later: evidence from two birth cohort studies. Int J Obes (Lond) 42:872–879PubMedCrossRef Kaseva N, Vääräsmäki M, Matinolli HM, Sipola-Leppänen M, Tikanmäki M, Heinonen K, Lano A, Wolke D, Andersson S, Järvelin MR, Räikkönen K, Eriksson JG, Kajantie E (2018) Pre-pregnancy overweight or obesity and gestational diabetes as predictors of body composition in offspring twenty years later: evidence from two birth cohort studies. Int J Obes (Lond) 42:872–879PubMedCrossRef
38.
Zurück zum Zitat Du Q, Sompolinsky Y, Walfisch A, Zhong H, Liu Y, Feng W (2020) The sex specific association between maternal gestational diabetes and offspring metabolic status at 1 year of age. Front Endocrinol (Lausanne) 11:608125PubMedCrossRef Du Q, Sompolinsky Y, Walfisch A, Zhong H, Liu Y, Feng W (2020) The sex specific association between maternal gestational diabetes and offspring metabolic status at 1 year of age. Front Endocrinol (Lausanne) 11:608125PubMedCrossRef
39.
Zurück zum Zitat Gingras V, Rifas-Shiman SL, Derks IPM, Aris IM, Oken E, Hivert M-F (2018) Associations of gestational glucose tolerance with offspring body composition and estimated insulin resistance in early adolescence. Diabetes Care 41:e164–e166PubMedPubMedCentralCrossRef Gingras V, Rifas-Shiman SL, Derks IPM, Aris IM, Oken E, Hivert M-F (2018) Associations of gestational glucose tolerance with offspring body composition and estimated insulin resistance in early adolescence. Diabetes Care 41:e164–e166PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Le Moullec N, Fianu A, Maillard O, Chazelle E, Naty N, Schneebeli C, Gérardin P, Huiart L, Charles M-A, Favier F (2018) Sexual dimorphism in the association between gestational diabetes mellitus and overweight in offspring at 5–7 years: The OBEGEST cohort study. PLoS ONE 13:e0195531PubMedPubMedCentralCrossRef Le Moullec N, Fianu A, Maillard O, Chazelle E, Naty N, Schneebeli C, Gérardin P, Huiart L, Charles M-A, Favier F (2018) Sexual dimorphism in the association between gestational diabetes mellitus and overweight in offspring at 5–7 years: The OBEGEST cohort study. PLoS ONE 13:e0195531PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Shapiro ALB, Sauder KA, Tregellas JR, Legget KT, Gravitz SL, Ringham BM, Glueck DH, Johnson SL, Dabelea D (2017) Exposure to maternal diabetes in utero and offspring eating behavior: The EPOCH study. Appetite 116:610–615PubMedPubMedCentralCrossRef Shapiro ALB, Sauder KA, Tregellas JR, Legget KT, Gravitz SL, Ringham BM, Glueck DH, Johnson SL, Dabelea D (2017) Exposure to maternal diabetes in utero and offspring eating behavior: The EPOCH study. Appetite 116:610–615PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Wang A, Luo B, Chen Z, Xia Y, Chen C, Qi H, Baker PN, Saffery R, Han T-L, Zhang H (2022) Rodents on a high-fat diet born to mothers with gestational diabetes exhibit sex-specific lipidomic changes in reproductive organs. Acta Biochim Biophys Sin (Shanghai) 54:736–747PubMedCrossRef Wang A, Luo B, Chen Z, Xia Y, Chen C, Qi H, Baker PN, Saffery R, Han T-L, Zhang H (2022) Rodents on a high-fat diet born to mothers with gestational diabetes exhibit sex-specific lipidomic changes in reproductive organs. Acta Biochim Biophys Sin (Shanghai) 54:736–747PubMedCrossRef
43.
Zurück zum Zitat Mansell T, Vlahos A, Collier F, Ponsonby A-L, Vuillermin P, Ellul S, Tang MLK, Burgner D, Saffery R (2022) The newborn metabolome: associations with gestational diabetes, sex, gestation, birth mode, and birth weight. Pediatr Res 91:1864–1873PubMedCrossRef Mansell T, Vlahos A, Collier F, Ponsonby A-L, Vuillermin P, Ellul S, Tang MLK, Burgner D, Saffery R (2022) The newborn metabolome: associations with gestational diabetes, sex, gestation, birth mode, and birth weight. Pediatr Res 91:1864–1873PubMedCrossRef
44.
Zurück zum Zitat Spaziani M, Radicioni AF (2020) Metabolic and cardiovascular risk factors in Klinefelter syndrome. Am J Med Genet C Semin Med Genet 184:334–343PubMedCrossRef Spaziani M, Radicioni AF (2020) Metabolic and cardiovascular risk factors in Klinefelter syndrome. Am J Med Genet C Semin Med Genet 184:334–343PubMedCrossRef
45.
Zurück zum Zitat K. O'Neill, J. Alexander, R. Azuma, R. Xiao, N.W. Snyder, C.A. Mesaros, I.A. Blair, and S.E. Pinney, (2018) Gestational Diabetes Alters the Metabolomic Profile in 2nd Trimester Amniotic Fluid in a Sex-Specific Manner. Int J Mol Sci 19. K. O'Neill, J. Alexander, R. Azuma, R. Xiao, N.W. Snyder, C.A. Mesaros, I.A. Blair, and S.E. Pinney, (2018) Gestational Diabetes Alters the Metabolomic Profile in 2nd Trimester Amniotic Fluid in a Sex-Specific Manner. Int J Mol Sci 19.
46.
Zurück zum Zitat Yang Y, Pan Z, Guo F, Wang H, Long W, Wang H, Yu B (2021) Placental metabolic profiling in gestational diabetes mellitus: an important role of fatty acids. J Clin Lab Anal 35:e24096PubMedPubMedCentralCrossRef Yang Y, Pan Z, Guo F, Wang H, Long W, Wang H, Yu B (2021) Placental metabolic profiling in gestational diabetes mellitus: an important role of fatty acids. J Clin Lab Anal 35:e24096PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Nadif R, Dilworth MR, Sibley CP, Baker PN, Davidge ST, Gibson JM, Aplin JD, Westwood M (2015) The maternal environment programs postnatal weight gain and glucose tolerance of male offspring, but placental and fetal growth are determined by fetal genotype in the Leprdb/+ model of gestational diabetes. Endocrinology 156:360–366PubMedCrossRef Nadif R, Dilworth MR, Sibley CP, Baker PN, Davidge ST, Gibson JM, Aplin JD, Westwood M (2015) The maternal environment programs postnatal weight gain and glucose tolerance of male offspring, but placental and fetal growth are determined by fetal genotype in the Leprdb/+ model of gestational diabetes. Endocrinology 156:360–366PubMedCrossRef
48.
Zurück zum Zitat Chen B, Du Y-R, Zhu H, Sun M-L, Wang C, Cheng Y, Pang H, Ding G, Gao J, Tan Y, Tong X, Lv P, Zhou F, Zhan Q, Xu Z-M, Wang L, Luo D, Ye Y, Jin L, Zhang S, Zhu Y, Lin X, Wu Y, Jin L, Zhou Y, Yan C, Sheng J, Flatt PR, Xu G-L, Huang H (2022) Maternal inheritance of glucose intolerance via oocyte TET3 insufficiency. Nature 605:761–766PubMedCrossRef Chen B, Du Y-R, Zhu H, Sun M-L, Wang C, Cheng Y, Pang H, Ding G, Gao J, Tan Y, Tong X, Lv P, Zhou F, Zhan Q, Xu Z-M, Wang L, Luo D, Ye Y, Jin L, Zhang S, Zhu Y, Lin X, Wu Y, Jin L, Zhou Y, Yan C, Sheng J, Flatt PR, Xu G-L, Huang H (2022) Maternal inheritance of glucose intolerance via oocyte TET3 insufficiency. Nature 605:761–766PubMedCrossRef
49.
Zurück zum Zitat Zhu H, Luo S-S, Cheng Y, Yan Y-S, Zou K-X, Ding G-L, Jin L, Huang H-F (2021) Intrauterine hyperglycemia alters the metabolomic profile in fetal mouse pancreas in a gender-specific manner. Front Endocrinol (Lausanne) 12:710221PubMedCrossRef Zhu H, Luo S-S, Cheng Y, Yan Y-S, Zou K-X, Ding G-L, Jin L, Huang H-F (2021) Intrauterine hyperglycemia alters the metabolomic profile in fetal mouse pancreas in a gender-specific manner. Front Endocrinol (Lausanne) 12:710221PubMedCrossRef
50.
Zurück zum Zitat S. Renner, A.S. Martins, E. Streckel, C. Braun-Reichhart, M. Backman, C. Prehn, N. Klymiuk, A. Bähr, A. Blutke, C. Landbrecht-Schessl, A. Wünsch, B. Kessler, M. Kurome, A. Hinrichs, S.-J. Koopmans, S. Krebs, E. Kemter, B. Rathkolb, H. Nagashima, H. Blum, M. Ritzmann, R. Wanke, B. Aigner, J. Adamski, M. Hrabě de Angelis, and E. Wolf, (2019) Mild maternal hyperglycemia in transgenic pigs causes impaired glucose tolerance and metabolic alterations in neonatal offspring. Dis Model Mech 12. S. Renner, A.S. Martins, E. Streckel, C. Braun-Reichhart, M. Backman, C. Prehn, N. Klymiuk, A. Bähr, A. Blutke, C. Landbrecht-Schessl, A. Wünsch, B. Kessler, M. Kurome, A. Hinrichs, S.-J. Koopmans, S. Krebs, E. Kemter, B. Rathkolb, H. Nagashima, H. Blum, M. Ritzmann, R. Wanke, B. Aigner, J. Adamski, M. Hrabě de Angelis, and E. Wolf, (2019) Mild maternal hyperglycemia in transgenic pigs causes impaired glucose tolerance and metabolic alterations in neonatal offspring. Dis Model Mech 12.
51.
Zurück zum Zitat Krishnaveni GV, Hill JC, Leary SD, Veena SR, Saperia J, Saroja A, Karat SC, Fall CHD (2005) Anthropometry, glucose tolerance, and insulin concentrations in Indian children: relationships to maternal glucose and insulin concentrations during pregnancy. Diabetes Care 28:2919–2925PubMedCrossRef Krishnaveni GV, Hill JC, Leary SD, Veena SR, Saperia J, Saroja A, Karat SC, Fall CHD (2005) Anthropometry, glucose tolerance, and insulin concentrations in Indian children: relationships to maternal glucose and insulin concentrations during pregnancy. Diabetes Care 28:2919–2925PubMedCrossRef
52.
Zurück zum Zitat Krishnaveni GV, Veena SR, Hill JC, Kehoe S, Karat SC, Fall CHD (2010) Intrauterine exposure to maternal diabetes is associated with higher adiposity and insulin resistance and clustering of cardiovascular risk markers in Indian children. Diabetes Care 33:402–404PubMedCrossRef Krishnaveni GV, Veena SR, Hill JC, Kehoe S, Karat SC, Fall CHD (2010) Intrauterine exposure to maternal diabetes is associated with higher adiposity and insulin resistance and clustering of cardiovascular risk markers in Indian children. Diabetes Care 33:402–404PubMedCrossRef
53.
Zurück zum Zitat J.-F. Gautier, L.S. Fetita, J.-P. Riveline, F. Ibrahim, R. Porcher, C. Abi Khalil, G. Velho, S.-P. Choukem, S. Hadjadj, E. Larger, R. Roussel, P. Boudou, M. Marre, E. Ravussin, and F. Mauvais-Jarvis, (2018) Sex Difference In the Effect of Fetal Exposure to Maternal Diabetes on Insulin Secretion. J Endocr Soc 2 391–397. J.-F. Gautier, L.S. Fetita, J.-P. Riveline, F. Ibrahim, R. Porcher, C. Abi Khalil, G. Velho, S.-P. Choukem, S. Hadjadj, E. Larger, R. Roussel, P. Boudou, M. Marre, E. Ravussin, and F. Mauvais-Jarvis, (2018) Sex Difference In the Effect of Fetal Exposure to Maternal Diabetes on Insulin Secretion. J Endocr Soc 2 391–397.
54.
Zurück zum Zitat Krieger J-P, Arnold M, Pettersen KG, Lossel P, Langhans W, Lee SJ (2016) Knockdown of GLP-1 receptors in vagal afferents affects normal food intake and glycemia. Diabetes 65:34–43PubMedCrossRef Krieger J-P, Arnold M, Pettersen KG, Lossel P, Langhans W, Lee SJ (2016) Knockdown of GLP-1 receptors in vagal afferents affects normal food intake and glycemia. Diabetes 65:34–43PubMedCrossRef
55.
Zurück zum Zitat Smith EP, An Z, Wagner C, Lewis AG, Cohen EB, Li B, Mahbod P, Sandoval D, Perez-Tilve D, Tamarina N, Philipson LH, Stoffers DA, Seeley RJ, D’Alessio DA (2014) The role of β cell glucagon-like peptide-1 signaling in glucose regulation and response to diabetes drugs. Cell Metab 19:1050–1057PubMedPubMedCentralCrossRef Smith EP, An Z, Wagner C, Lewis AG, Cohen EB, Li B, Mahbod P, Sandoval D, Perez-Tilve D, Tamarina N, Philipson LH, Stoffers DA, Seeley RJ, D’Alessio DA (2014) The role of β cell glucagon-like peptide-1 signaling in glucose regulation and response to diabetes drugs. Cell Metab 19:1050–1057PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Navarro G, Xu W, Jacobson DA, Wicksteed B, Allard C, Zhang G, De Gendt K, Kim SH, Wu H, Zhang H, Verhoeven G, Katzenellenbogen JA, Mauvais-Jarvis F (2016) Extranuclear actions of the androgen receptor enhance glucose-stimulated insulin secretion in the male. Cell Metab 23:837–851PubMedPubMedCentralCrossRef Navarro G, Xu W, Jacobson DA, Wicksteed B, Allard C, Zhang G, De Gendt K, Kim SH, Wu H, Zhang H, Verhoeven G, Katzenellenbogen JA, Mauvais-Jarvis F (2016) Extranuclear actions of the androgen receptor enhance glucose-stimulated insulin secretion in the male. Cell Metab 23:837–851PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Perng W, Ringham BM, Smith HA, Michelotti G, Kechris KM, Dabelea D (2020) A prospective study of associations between in utero exposure to gestational diabetes mellitus and metabolomic profiles during late childhood and adolescence. Diabetologia 63:296–312PubMedCrossRef Perng W, Ringham BM, Smith HA, Michelotti G, Kechris KM, Dabelea D (2020) A prospective study of associations between in utero exposure to gestational diabetes mellitus and metabolomic profiles during late childhood and adolescence. Diabetologia 63:296–312PubMedCrossRef
58.
Zurück zum Zitat Hockett CW, Bedrick EJ, Zeitler P, Crume TL, Daniels S, Dabelea D (2019) Exposure to diabetes in utero is associated with earlier pubertal timing and faster pubertal growth in the offspring: the EPOCH study. J Pediatr 206:105–112PubMedCrossRef Hockett CW, Bedrick EJ, Zeitler P, Crume TL, Daniels S, Dabelea D (2019) Exposure to diabetes in utero is associated with earlier pubertal timing and faster pubertal growth in the offspring: the EPOCH study. J Pediatr 206:105–112PubMedCrossRef
59.
Zurück zum Zitat Kubo A, Ferrara A, Laurent CA, Windham GC, Greenspan LC, Deardorff J, Hiatt RA, Quesenberry CP, Kushi LH (2016) Associations between maternal pregravid obesity and gestational diabetes and the timing of pubarche in daughters. Am J Epidemiol 184(1):14CrossRef Kubo A, Ferrara A, Laurent CA, Windham GC, Greenspan LC, Deardorff J, Hiatt RA, Quesenberry CP, Kushi LH (2016) Associations between maternal pregravid obesity and gestational diabetes and the timing of pubarche in daughters. Am J Epidemiol 184(1):14CrossRef
60.
Zurück zum Zitat Lauridsen LLB, Arendt LH, Ernst A, Brix N, Parner ET, Olsen J, Ramlau-Hansen CH (2018) Maternal diabetes mellitus and timing of pubertal development in daughters and sons: a nationwide cohort study. Fertil Steril 110:35–44PubMedCrossRef Lauridsen LLB, Arendt LH, Ernst A, Brix N, Parner ET, Olsen J, Ramlau-Hansen CH (2018) Maternal diabetes mellitus and timing of pubertal development in daughters and sons: a nationwide cohort study. Fertil Steril 110:35–44PubMedCrossRef
61.
Zurück zum Zitat Yoshikawa H, Tajiri Y, Sako Y, Hashimoto T, Umeda F, Nawata H (2001) Effects of free fatty acids on beta-cell functions: a possible involvement of peroxisome proliferator-activated receptors alpha or pancreatic/duodenal homeobox. Metabolism 50:613–618PubMedCrossRef Yoshikawa H, Tajiri Y, Sako Y, Hashimoto T, Umeda F, Nawata H (2001) Effects of free fatty acids on beta-cell functions: a possible involvement of peroxisome proliferator-activated receptors alpha or pancreatic/duodenal homeobox. Metabolism 50:613–618PubMedCrossRef
62.
Zurück zum Zitat A.K. Nur Zati Iwani, M.Y. Jalaludin, A. Yahya, F. Mansor, F. MD Zain, J.Y.H. Hong, R.M. Wan Mohd Zin, and A.H. Mokhtar, (2022) TG: HDL-C Ratio as Insulin Resistance Marker for Metabolic Syndrome in Children With Obesity. Front Endocrinol (Lausanne) 13 852290 A.K. Nur Zati Iwani, M.Y. Jalaludin, A. Yahya, F. Mansor, F. MD Zain, J.Y.H. Hong, R.M. Wan Mohd Zin, and A.H. Mokhtar, (2022) TG: HDL-C Ratio as Insulin Resistance Marker for Metabolic Syndrome in Children With Obesity. Front Endocrinol (Lausanne) 13 852290
63.
Zurück zum Zitat Franssen R, Monajemi H, Stroes ESG, Kastelein JJP (2011) Obesity and dyslipidemia. Med Clin North Am 95:893–902PubMedCrossRef Franssen R, Monajemi H, Stroes ESG, Kastelein JJP (2011) Obesity and dyslipidemia. Med Clin North Am 95:893–902PubMedCrossRef
64.
Zurück zum Zitat Randle PJ, Garland PB, Hales CN, Newsholme EA (1963) The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1:785–789PubMedCrossRef Randle PJ, Garland PB, Hales CN, Newsholme EA (1963) The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1:785–789PubMedCrossRef
65.
Zurück zum Zitat Y. Lu, Y. Jia, J. Lu, J. Liu, Y. Xu, Y. Liu, and K. Chen, (2021) Progenies of gestational diabetes mellitus exhibit sex disparity in metabolism after respective therapies of insulin, glibenclamide, and metformin in dams during pregnancy. Arch Physiol Biochem. Y. Lu, Y. Jia, J. Lu, J. Liu, Y. Xu, Y. Liu, and K. Chen, (2021) Progenies of gestational diabetes mellitus exhibit sex disparity in metabolism after respective therapies of insulin, glibenclamide, and metformin in dams during pregnancy. Arch Physiol Biochem.
66.
Zurück zum Zitat Thaeomor A, Teangphuck P, Chaisakul J, Seanthaweesuk S, Somparn N, Roysommuti S (2017) Perinatal taurine supplementation prevents metabolic and cardiovascular effects of maternal diabetes in adult rat offspring. Adv Exp Med Biol 975(Pt 1):295–305PubMedCrossRef Thaeomor A, Teangphuck P, Chaisakul J, Seanthaweesuk S, Somparn N, Roysommuti S (2017) Perinatal taurine supplementation prevents metabolic and cardiovascular effects of maternal diabetes in adult rat offspring. Adv Exp Med Biol 975(Pt 1):295–305PubMedCrossRef
67.
Zurück zum Zitat Brawerman GM, Kereliuk SM, Brar N, Cole LK, Seshadri N, Pereira TJ, Xiang B, Hunt KL, Fonseca MA, Hatch GM, Doucette CA, Dolinsky VW (2019) Maternal resveratrol administration protects against gestational diabetes-induced glucose intolerance and islet dysfunction in the rat offspring. J Physiol 597:4175–4192PubMedCrossRef Brawerman GM, Kereliuk SM, Brar N, Cole LK, Seshadri N, Pereira TJ, Xiang B, Hunt KL, Fonseca MA, Hatch GM, Doucette CA, Dolinsky VW (2019) Maternal resveratrol administration protects against gestational diabetes-induced glucose intolerance and islet dysfunction in the rat offspring. J Physiol 597:4175–4192PubMedCrossRef
68.
Zurück zum Zitat Landon MB, Rice MM, Varner MW, Casey BM, Reddy UM, Wapner RJ, Rouse DJ, Biggio JR, Thorp JM, Chien EK, Saade G, Peaceman AM, Blackwell SC, VanDorsten JP (2015) Mild gestational diabetes mellitus and long-term child health. Diabetes Care 38:445–452PubMedCrossRef Landon MB, Rice MM, Varner MW, Casey BM, Reddy UM, Wapner RJ, Rouse DJ, Biggio JR, Thorp JM, Chien EK, Saade G, Peaceman AM, Blackwell SC, VanDorsten JP (2015) Mild gestational diabetes mellitus and long-term child health. Diabetes Care 38:445–452PubMedCrossRef
69.
Zurück zum Zitat Zhang W, Su R, Feng H, Lin L, Wang C, Yang H (2019) Transgenerational obesity and alteration of ARHGEF11 in the rat liver induced by intrauterine hyperglycemia. J Diabetes Res 2019:6320839PubMedPubMedCentralCrossRef Zhang W, Su R, Feng H, Lin L, Wang C, Yang H (2019) Transgenerational obesity and alteration of ARHGEF11 in the rat liver induced by intrauterine hyperglycemia. J Diabetes Res 2019:6320839PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Hanafi MY, Abdelkhalek TM, Saad MI, Saleh MM, Haiba MM, Kamel MA (2016) Diabetes-induced perturbations are subject to intergenerational transmission through maternal line. J Physiol Biochem 72:315–326PubMedCrossRef Hanafi MY, Abdelkhalek TM, Saad MI, Saleh MM, Haiba MM, Kamel MA (2016) Diabetes-induced perturbations are subject to intergenerational transmission through maternal line. J Physiol Biochem 72:315–326PubMedCrossRef
71.
Zurück zum Zitat M. Huerta-Cervantes, D.J. Peña-Montes, M.Á. López-Vázquez, R. Montoya-Pérez, C. Cortés-Rojo, M.E. (2021) Olvera-Cortés, and A. Saavedra-Molina, Effects of Gestational Diabetes in Cognitive Behavior, Oxidative Stress and Metabolism on the Second-Generation Off-Spring of Rats. Nutrients 13. M. Huerta-Cervantes, D.J. Peña-Montes, M.Á. López-Vázquez, R. Montoya-Pérez, C. Cortés-Rojo, M.E. (2021) Olvera-Cortés, and A. Saavedra-Molina, Effects of Gestational Diabetes in Cognitive Behavior, Oxidative Stress and Metabolism on the Second-Generation Off-Spring of Rats. Nutrients 13.
72.
Zurück zum Zitat Anderson LA, McTernan PG, Barnett AH, Kumar S (2001) The effects of androgens and estrogens on preadipocyte proliferation in human adipose tissue: influence of gender and site. J Clin Endocrinol Metab 86:5045–5051PubMedCrossRef Anderson LA, McTernan PG, Barnett AH, Kumar S (2001) The effects of androgens and estrogens on preadipocyte proliferation in human adipose tissue: influence of gender and site. J Clin Endocrinol Metab 86:5045–5051PubMedCrossRef
73.
74.
Zurück zum Zitat Medrikova D, Jilkova ZM, Bardova K, Janovska P, Rossmeisl M, Kopecky J (2012) Sex differences during the course of diet-induced obesity in mice: adipose tissue expandability and glycemic control. Int J Obes (Lond) 36:262–272PubMedCrossRef Medrikova D, Jilkova ZM, Bardova K, Janovska P, Rossmeisl M, Kopecky J (2012) Sex differences during the course of diet-induced obesity in mice: adipose tissue expandability and glycemic control. Int J Obes (Lond) 36:262–272PubMedCrossRef
75.
Zurück zum Zitat Mårin P, Odén B, Björntorp P (1995) Assimilation and mobilization of triglycerides in subcutaneous abdominal and femoral adipose tissue in vivo in men: effects of androgens. J Clin Endocrinol Metab 80:239–243PubMed Mårin P, Odén B, Björntorp P (1995) Assimilation and mobilization of triglycerides in subcutaneous abdominal and femoral adipose tissue in vivo in men: effects of androgens. J Clin Endocrinol Metab 80:239–243PubMed
76.
Zurück zum Zitat Zitzmann M (2009) Testosterone deficiency, insulin resistance and the metabolic syndrome. Nat Rev Endocrinol 5:673–681PubMedCrossRef Zitzmann M (2009) Testosterone deficiency, insulin resistance and the metabolic syndrome. Nat Rev Endocrinol 5:673–681PubMedCrossRef
77.
Zurück zum Zitat Le May C, Chu K, Hu M, Ortega CS, Simpson ER, Korach KS, Tsai M-J, Mauvais-Jarvis F (2006) Estrogens protect pancreatic beta-cells from apoptosis and prevent insulin-deficient diabetes mellitus in mice. P Natl Acad Sci USA 103:9232–9237CrossRef Le May C, Chu K, Hu M, Ortega CS, Simpson ER, Korach KS, Tsai M-J, Mauvais-Jarvis F (2006) Estrogens protect pancreatic beta-cells from apoptosis and prevent insulin-deficient diabetes mellitus in mice. P Natl Acad Sci USA 103:9232–9237CrossRef
78.
Zurück zum Zitat Wong WPS, Tiano JP, Liu S, Hewitt SC, Le May C, Dalle S, Katzenellenbogen JA, Katzenellenbogen BS, Korach KS, Mauvais-Jarvis F (2010) Extranuclear estrogen receptor-alpha stimulates NeuroD1 binding to the insulin promoter and favors insulin synthesis. P Natl Acad Sci USA 107:13057–13062CrossRef Wong WPS, Tiano JP, Liu S, Hewitt SC, Le May C, Dalle S, Katzenellenbogen JA, Katzenellenbogen BS, Korach KS, Mauvais-Jarvis F (2010) Extranuclear estrogen receptor-alpha stimulates NeuroD1 binding to the insulin promoter and favors insulin synthesis. P Natl Acad Sci USA 107:13057–13062CrossRef
79.
Zurück zum Zitat Ribas V, Nguyen MTA, Henstridge DC, Nguyen A-K, Beaven SW, Watt MJ, Hevener AL (2010) Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERalpha-deficient mice. Am J Physiol Endocrinol Metab 298:E304–E319PubMedCrossRef Ribas V, Nguyen MTA, Henstridge DC, Nguyen A-K, Beaven SW, Watt MJ, Hevener AL (2010) Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERalpha-deficient mice. Am J Physiol Endocrinol Metab 298:E304–E319PubMedCrossRef
80.
Zurück zum Zitat Alonso-Magdalena P, Ropero AB, Carrera MP, Cederroth CR, Baquié M, Gauthier BR, Nef S, Stefani E, Nadal A (2008) Pancreatic insulin content regulation by the estrogen receptor ER alpha. PLoS ONE 3:e2069PubMedPubMedCentralCrossRef Alonso-Magdalena P, Ropero AB, Carrera MP, Cederroth CR, Baquié M, Gauthier BR, Nef S, Stefani E, Nadal A (2008) Pancreatic insulin content regulation by the estrogen receptor ER alpha. PLoS ONE 3:e2069PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Camporez JP, Lyu K, Goldberg EL, Zhang D, Cline GW, Jurczak MJ, Dixit VD, Petersen KF, Shulman GI (2019) Anti-inflammatory effects of oestrogen mediate the sexual dimorphic response to lipid-induced insulin resistance. J Physiol 597:3885–3903PubMedCrossRef Camporez JP, Lyu K, Goldberg EL, Zhang D, Cline GW, Jurczak MJ, Dixit VD, Petersen KF, Shulman GI (2019) Anti-inflammatory effects of oestrogen mediate the sexual dimorphic response to lipid-induced insulin resistance. J Physiol 597:3885–3903PubMedCrossRef
82.
Zurück zum Zitat Camporez JPG, Jornayvaz FR, Lee H-Y, Kanda S, Guigni BA, Kahn M, Samuel VT, Carvalho CRO, Petersen KF, Jurczak MJ, Shulman GI (2013) Cellular mechanism by which estradiol protects female ovariectomized mice from high-fat diet-induced hepatic and muscle insulin resistance. Endocrinology 154:1021–1028PubMedPubMedCentralCrossRef Camporez JPG, Jornayvaz FR, Lee H-Y, Kanda S, Guigni BA, Kahn M, Samuel VT, Carvalho CRO, Petersen KF, Jurczak MJ, Shulman GI (2013) Cellular mechanism by which estradiol protects female ovariectomized mice from high-fat diet-induced hepatic and muscle insulin resistance. Endocrinology 154:1021–1028PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Umano GR, Shabanova V, Pierpont B, Mata M, Nouws J, Tricò D, Galderisi A, Santoro N, Caprio S (2019) A low visceral fat proportion, independent of total body fat mass, protects obese adolescent girls against fatty liver and glucose dysregulation: a longitudinal study. Int J Obes (Lond) 43:673–682PubMedCrossRef Umano GR, Shabanova V, Pierpont B, Mata M, Nouws J, Tricò D, Galderisi A, Santoro N, Caprio S (2019) A low visceral fat proportion, independent of total body fat mass, protects obese adolescent girls against fatty liver and glucose dysregulation: a longitudinal study. Int J Obes (Lond) 43:673–682PubMedCrossRef
84.
Zurück zum Zitat G. Navarro, C. Allard, J.J. Morford, W. Xu, S. Liu, A. Molinas, Jr., S.M. Butcher, N.H. Fine, M. Blandino-Rosano, V.N. Sure, S. Yu, R. Zhang, H. Münzberg, D.A. Jacobson, P.V. Katakam, D.J. Hodson, E. Bernal-Mizrachi, A. Zsombok, and F. Mauvais-Jarvis, (2018) Androgen excess in pancreatic β cells and neurons predisposes female mice to type 2 diabetes. JCI Insight 3. G. Navarro, C. Allard, J.J. Morford, W. Xu, S. Liu, A. Molinas, Jr., S.M. Butcher, N.H. Fine, M. Blandino-Rosano, V.N. Sure, S. Yu, R. Zhang, H. Münzberg, D.A. Jacobson, P.V. Katakam, D.J. Hodson, E. Bernal-Mizrachi, A. Zsombok, and F. Mauvais-Jarvis, (2018) Androgen excess in pancreatic β cells and neurons predisposes female mice to type 2 diabetes. JCI Insight 3.
85.
Zurück zum Zitat S. Handgraaf, R. Dusaulcy, F. Visentin, J. Philippe, and Y. Gosmain, (2018) 17-β Estradiol regulates proglucagon-derived peptide secretion in mouse and human α- and L cells. JCI Insight 3. S. Handgraaf, R. Dusaulcy, F. Visentin, J. Philippe, and Y. Gosmain, (2018) 17-β Estradiol regulates proglucagon-derived peptide secretion in mouse and human α- and L cells. JCI Insight 3.
86.
Zurück zum Zitat Kvist H, Chowdhury B, Grangård U, Tylén U, Sjöström L (1988) Total and visceral adipose-tissue volumes derived from measurements with computed tomography in adult men and women: predictive equations. Am J Clin Nutr 48:1351–1361PubMedCrossRef Kvist H, Chowdhury B, Grangård U, Tylén U, Sjöström L (1988) Total and visceral adipose-tissue volumes derived from measurements with computed tomography in adult men and women: predictive equations. Am J Clin Nutr 48:1351–1361PubMedCrossRef
87.
Zurück zum Zitat Lemieux S, Prud’homme D, Bouchard C, Tremblay A, Després JP (1993) Sex differences in the relation of visceral adipose tissue accumulation to total body fatness. Am J Clin Nutr 58:463–467PubMedCrossRef Lemieux S, Prud’homme D, Bouchard C, Tremblay A, Després JP (1993) Sex differences in the relation of visceral adipose tissue accumulation to total body fatness. Am J Clin Nutr 58:463–467PubMedCrossRef
88.
Zurück zum Zitat L. Dearden, S.G. Bouret, and S.E. Ozanne, (2018) Sex and gender differences in developmental programming of metabolism. Mol Metab 15. L. Dearden, S.G. Bouret, and S.E. Ozanne, (2018) Sex and gender differences in developmental programming of metabolism. Mol Metab 15.
89.
Zurück zum Zitat Snijder MB, Dekker JM, Visser M, Bouter LM, Stehouwer CDA, Yudkin JS, Heine RJ, Nijpels G, Seidell JC (2004) Trunk fat and leg fat have independent and opposite associations with fasting and postload glucose levels: the Hoorn study. Diabetes Care 27:372–377PubMedCrossRef Snijder MB, Dekker JM, Visser M, Bouter LM, Stehouwer CDA, Yudkin JS, Heine RJ, Nijpels G, Seidell JC (2004) Trunk fat and leg fat have independent and opposite associations with fasting and postload glucose levels: the Hoorn study. Diabetes Care 27:372–377PubMedCrossRef
90.
Zurück zum Zitat Macotela Y, Boucher J, Tran TT, Kahn CR (2009) Sex and depot differences in adipocyte insulin sensitivity and glucose metabolism. Diabetes 58:803–812PubMedPubMedCentralCrossRef Macotela Y, Boucher J, Tran TT, Kahn CR (2009) Sex and depot differences in adipocyte insulin sensitivity and glucose metabolism. Diabetes 58:803–812PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Gallagher D, Visser M, De Meersman RE, Sepúlveda D, Baumgartner RN, Pierson RN, Harris T, Heymsfield SB (1985) Appendicular skeletal muscle mass: effects of age, gender, and ethnicity. J Appl Physiol 83(1997):229–239 Gallagher D, Visser M, De Meersman RE, Sepúlveda D, Baumgartner RN, Pierson RN, Harris T, Heymsfield SB (1985) Appendicular skeletal muscle mass: effects of age, gender, and ethnicity. J Appl Physiol 83(1997):229–239
92.
Zurück zum Zitat Janssen I, Heymsfield SB, Wang ZM, Ross R (1985) Skeletal muscle mass and distribution in 468 men and women aged 18–88 yr. J Appl Physiol 89(2000):81–88 Janssen I, Heymsfield SB, Wang ZM, Ross R (1985) Skeletal muscle mass and distribution in 468 men and women aged 18–88 yr. J Appl Physiol 89(2000):81–88
93.
Zurück zum Zitat Shulman GI (2014) Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease. N Engl J Med 371:1131–1141PubMedCrossRef Shulman GI (2014) Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease. N Engl J Med 371:1131–1141PubMedCrossRef
94.
Zurück zum Zitat Machann J, Thamer C, Schnoedt B, Stefan N, Stumvoll M, Haring HU, Claussen CD, Fritsche A, Schick F (2005) Age and gender related effects on adipose tissue compartments of subjects with increased risk for type 2 diabetes: a whole body MRI/MRS study. MAGMA 18:128–137PubMedCrossRef Machann J, Thamer C, Schnoedt B, Stefan N, Stumvoll M, Haring HU, Claussen CD, Fritsche A, Schick F (2005) Age and gender related effects on adipose tissue compartments of subjects with increased risk for type 2 diabetes: a whole body MRI/MRS study. MAGMA 18:128–137PubMedCrossRef
95.
Zurück zum Zitat Marchese E, Rodeghier C, Monson RS, McCracken B, Shi T, Schrock W, Martellotto J, Oberholzer J, Danielson KK (2015) Enumerating β-Cells in whole human islets: sex differences and associations with clinical outcomes after islet transplantation. Diabetes Care 38:e176–e177PubMedPubMedCentralCrossRef Marchese E, Rodeghier C, Monson RS, McCracken B, Shi T, Schrock W, Martellotto J, Oberholzer J, Danielson KK (2015) Enumerating β-Cells in whole human islets: sex differences and associations with clinical outcomes after islet transplantation. Diabetes Care 38:e176–e177PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Hall E, Volkov P, Dayeh T, Esguerra JLS, Salö S, Eliasson L, Rönn T, Bacos K, Ling C (2014) Sex differences in the genome-wide DNA methylation pattern and impact on gene expression, microRNA levels and insulin secretion in human pancreatic islets. Genome Biol 15:522PubMedPubMedCentralCrossRef Hall E, Volkov P, Dayeh T, Esguerra JLS, Salö S, Eliasson L, Rönn T, Bacos K, Ling C (2014) Sex differences in the genome-wide DNA methylation pattern and impact on gene expression, microRNA levels and insulin secretion in human pancreatic islets. Genome Biol 15:522PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Emanuelli B, Eberlé D, Suzuki R, Kahn CR (2008) Overexpression of the dual-specificity phosphatase MKP-4/DUSP-9 protects against stress-induced insulin resistance. P Natl Acad Sci USA 105:3545–3550CrossRef Emanuelli B, Eberlé D, Suzuki R, Kahn CR (2008) Overexpression of the dual-specificity phosphatase MKP-4/DUSP-9 protects against stress-induced insulin resistance. P Natl Acad Sci USA 105:3545–3550CrossRef
98.
Zurück zum Zitat Gonzalez TL, Sun T, Koeppel AF, Lee B, Wang ET, Farber CR, Rich SS, Sundheimer LW, Buttle RA, Chen Y-DI, Rotter JI, Turner SD, Williams J, Goodarzi MO, Pisarska MD (2018) Sex differences in the late first trimester human placenta transcriptome. Biol Sex Differ 9:4PubMedPubMedCentralCrossRef Gonzalez TL, Sun T, Koeppel AF, Lee B, Wang ET, Farber CR, Rich SS, Sundheimer LW, Buttle RA, Chen Y-DI, Rotter JI, Turner SD, Williams J, Goodarzi MO, Pisarska MD (2018) Sex differences in the late first trimester human placenta transcriptome. Biol Sex Differ 9:4PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat O’Connell BA, Moritz KM, Walker DW, Dickinson H (2013) Sexually dimorphic placental development throughout gestation in the spiny mouse (Acomys cahirinus). Placenta 34:119–126PubMedCrossRef O’Connell BA, Moritz KM, Walker DW, Dickinson H (2013) Sexually dimorphic placental development throughout gestation in the spiny mouse (Acomys cahirinus). Placenta 34:119–126PubMedCrossRef
101.
Zurück zum Zitat Howerton CL, Bale TL (2014) Targeted placental deletion of OGT recapitulates the prenatal stress phenotype including hypothalamic mitochondrial dysfunction. P Natl Acad Sci USA 111:9639–9644CrossRef Howerton CL, Bale TL (2014) Targeted placental deletion of OGT recapitulates the prenatal stress phenotype including hypothalamic mitochondrial dysfunction. P Natl Acad Sci USA 111:9639–9644CrossRef
102.
Zurück zum Zitat P.S. Burgoyne, A.R. Thornhill, S.K. Boudrean, S.M. Darling, C.E. Bishop, and E.P. Evans, (1995) The genetic basis of XX-XY differences present before gonadal sex differentiation in the mouse. Philos Trans R Soc Lond B Biol Sci 350. P.S. Burgoyne, A.R. Thornhill, S.K. Boudrean, S.M. Darling, C.E. Bishop, and E.P. Evans, (1995) The genetic basis of XX-XY differences present before gonadal sex differentiation in the mouse. Philos Trans R Soc Lond B Biol Sci 350.
103.
104.
Zurück zum Zitat Pérez-Crespo M, Ramírez MA, Fernández-González R, Rizos D, Lonergan P, Pintado B, Gutiérrez-Adán A (2005) Differential sensitivity of male and female mouse embryos to oxidative induced heat-stress is mediated by glucose-6-phosphate dehydrogenase gene expression. Mol Reprod Dev 72:502–510PubMedCrossRef Pérez-Crespo M, Ramírez MA, Fernández-González R, Rizos D, Lonergan P, Pintado B, Gutiérrez-Adán A (2005) Differential sensitivity of male and female mouse embryos to oxidative induced heat-stress is mediated by glucose-6-phosphate dehydrogenase gene expression. Mol Reprod Dev 72:502–510PubMedCrossRef
105.
Zurück zum Zitat Galetzka D, Weis E, Tralau T, Seidmann L, Haaf T (2007) Sex-specific windows for high mRNA expression of DNA methyltransferases 1 and 3A and methyl-CpG-binding domain proteins 2 and 4 in human fetal gonads. Mol Reprod Dev 74:233–241PubMedCrossRef Galetzka D, Weis E, Tralau T, Seidmann L, Haaf T (2007) Sex-specific windows for high mRNA expression of DNA methyltransferases 1 and 3A and methyl-CpG-binding domain proteins 2 and 4 in human fetal gonads. Mol Reprod Dev 74:233–241PubMedCrossRef
106.
Zurück zum Zitat Xiao Y, Word B, Starlard-Davenport A, Haefele A, Lyn-Cook BD, Hammons G (2008) Age and gender affect DNMT3a and DNMT3b expression in human liver. Cell Biol Toxicol 24:265–272PubMedCrossRef Xiao Y, Word B, Starlard-Davenport A, Haefele A, Lyn-Cook BD, Hammons G (2008) Age and gender affect DNMT3a and DNMT3b expression in human liver. Cell Biol Toxicol 24:265–272PubMedCrossRef
107.
Zurück zum Zitat Shah KB, Chernausek SD, Teague AM, Bard DE, Tryggestad JB (2021) Maternal diabetes alters microRNA expression in fetal exosomes, human umbilical vein endothelial cells and placenta. Pediatr Res 89:1157–1163PubMedCrossRef Shah KB, Chernausek SD, Teague AM, Bard DE, Tryggestad JB (2021) Maternal diabetes alters microRNA expression in fetal exosomes, human umbilical vein endothelial cells and placenta. Pediatr Res 89:1157–1163PubMedCrossRef
Metadaten
Titel
Sex differences in glycolipidic disorders after exposure to maternal hyperglycemia during early development
verfasst von
S.-s. Luo
H. Zhu
H-f. Huang
G.-l. Ding
Publikationsdatum
01.08.2023
Verlag
Springer International Publishing
Erschienen in
Journal of Endocrinological Investigation / Ausgabe 8/2023
Elektronische ISSN: 1720-8386
DOI
https://doi.org/10.1007/s40618-023-02069-5

Weitere Artikel der Ausgabe 8/2023

Journal of Endocrinological Investigation 8/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.