Skip to main content
Erschienen in: Current Diabetes Reports 2/2013

01.04.2013 | Pathogenesis of Type 2 Diabetes and Insulin Resistance (RM Watanabe, Section Editor)

Maternal/Fetal Determinants of Insulin Resistance in Women During Pregnancy and in Offspring Over Life

verfasst von: Marilyn Lacroix, Eralda Kina, Marie-France Hivert

Erschienen in: Current Diabetes Reports | Ausgabe 2/2013

Einloggen, um Zugang zu erhalten

Abstract

Insulin resistance is a component of the pathophysiology of both type 2 diabetes and gestational diabetes mellitus (GDM), but is also characteristic of normal glycemic physiology during pregnancy. In recent years, many studies have tried to understand determinants of insulin resistance in normal pregnancy and GDM, revealing that the placenta is capable of secreting many cytokines and hormones, classically considered as adipokines. More specifically, it appears that leptin and TNFα could be implicated in gestational insulin resistance and GDM pathophysiology. In addition, the maternal metabolic milieu was also identified as a key determinant of later insulin resistance in offspring, a phenomenon often described as ‘fetal programming’. This article reviews the established risk factors and the more novel suspected biomarkers involved in maternal insulin resistance during pregnancy as well as the maternal and early life determinants of insulin resistance in offspring later in their life. We are also highlighting recent reports of the potential mechanisms involved in ‘programming’ of insulin resistance such as epigenetic modulation.
Literatur
1.
Zurück zum Zitat Rutter MK, Meigs JB, Sullivan LM, D’Agostino RBS, Wilson PW. Insulin resistance, the metabolic syndrome, and incident cardiovascular events in the Framingham Offspring Study. Diabetes. 2005;54:3252–7.PubMedCrossRef Rutter MK, Meigs JB, Sullivan LM, D’Agostino RBS, Wilson PW. Insulin resistance, the metabolic syndrome, and incident cardiovascular events in the Framingham Offspring Study. Diabetes. 2005;54:3252–7.PubMedCrossRef
2.
Zurück zum Zitat Catalano PM, Hoegh M, Minium J, Huston-Presley L, Bernard S, Kalhan S, et al. Adiponectin in human pregnancy: implications for regulation of glucose and lipid metabolism. Diabetologia. 2006;49:1677–85.PubMedCrossRef Catalano PM, Hoegh M, Minium J, Huston-Presley L, Bernard S, Kalhan S, et al. Adiponectin in human pregnancy: implications for regulation of glucose and lipid metabolism. Diabetologia. 2006;49:1677–85.PubMedCrossRef
3.
Zurück zum Zitat Catalano PM. Carbohydrate metabolism and gestational diabetes. Clin Obstet Gynecol. 1994;37:25–38.PubMedCrossRef Catalano PM. Carbohydrate metabolism and gestational diabetes. Clin Obstet Gynecol. 1994;37:25–38.PubMedCrossRef
4.
Zurück zum Zitat Godfrey KM, Barker DJ. Fetal nutrition and adult disease. Am J Clin Nutr. 2000;71:1344S–52S.PubMed Godfrey KM, Barker DJ. Fetal nutrition and adult disease. Am J Clin Nutr. 2000;71:1344S–52S.PubMed
5.
Zurück zum Zitat Nolan CJ, Damm P, Prentki M. Type 2 diabetes across generations: from pathophysiology to prevention and management. Lancet. 2011;378:169–81.PubMedCrossRef Nolan CJ, Damm P, Prentki M. Type 2 diabetes across generations: from pathophysiology to prevention and management. Lancet. 2011;378:169–81.PubMedCrossRef
6.
Zurück zum Zitat •• Bouchard L, Hivert MF, Guay SP, St-Pierre J, Perron P, Brisson D. Placental adiponectin gene DNA methylation levels are associated with mothers’ blood glucose concentration. Diabetes. 2012;61:1272–80. This article demonstrates that maternal glycemia is correlated with DNA methylation levels in the promoter region of ADIPOQ encoding for adiponectin, an adipokine potentially involved in insulin sensitivity pathways.PubMedCrossRef •• Bouchard L, Hivert MF, Guay SP, St-Pierre J, Perron P, Brisson D. Placental adiponectin gene DNA methylation levels are associated with mothers’ blood glucose concentration. Diabetes. 2012;61:1272–80. This article demonstrates that maternal glycemia is correlated with DNA methylation levels in the promoter region of ADIPOQ encoding for adiponectin, an adipokine potentially involved in insulin sensitivity pathways.PubMedCrossRef
7.
Zurück zum Zitat DeFronzo RA, Tobin JD, Andres R. Glucose clamp technique: a method for quantifying insulin secretion and resistance. Am J Physiol. 1979;237:E214–23.PubMed DeFronzo RA, Tobin JD, Andres R. Glucose clamp technique: a method for quantifying insulin secretion and resistance. Am J Physiol. 1979;237:E214–23.PubMed
8.
Zurück zum Zitat Matsuda M, DeFronzo RA. Insulin sensitivity indices obtained from oral glucose tolerance testing: comparison with the euglycemic insulin clamp. Diabetes Care. 1999;22:1462–70.PubMedCrossRef Matsuda M, DeFronzo RA. Insulin sensitivity indices obtained from oral glucose tolerance testing: comparison with the euglycemic insulin clamp. Diabetes Care. 1999;22:1462–70.PubMedCrossRef
9.
Zurück zum Zitat Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 1985;28:412–9.PubMedCrossRef Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 1985;28:412–9.PubMedCrossRef
10.
Zurück zum Zitat Chen X, Scholl TO. Ethnic differences in C-peptide/insulin/glucose dynamics in young pregnant women. J Clin Endocrinol Metab. 2002;87:4642–6.PubMedCrossRef Chen X, Scholl TO. Ethnic differences in C-peptide/insulin/glucose dynamics in young pregnant women. J Clin Endocrinol Metab. 2002;87:4642–6.PubMedCrossRef
11.
Zurück zum Zitat Retnakaran R, Hanley AJ, Connelly PW, Sermer M, Zinman B. Ethnicity modifies the effect of obesity on insulin resistance in pregnancy: a comparison of Asian, South Asian, and Caucasian women. J Clin Endocrinol Metab. 2006;91:93–7.PubMedCrossRef Retnakaran R, Hanley AJ, Connelly PW, Sermer M, Zinman B. Ethnicity modifies the effect of obesity on insulin resistance in pregnancy: a comparison of Asian, South Asian, and Caucasian women. J Clin Endocrinol Metab. 2006;91:93–7.PubMedCrossRef
12.
Zurück zum Zitat Gunton JE, Hitchman R, McElduff A. Effects of ethnicity on glucose tolerance, insulin resistance and beta cell function in 223 women with an abnormal glucose challenge test during pregnancy. Aust N Z J Obstet Gynaecol. 2001;41:182–6.PubMedCrossRef Gunton JE, Hitchman R, McElduff A. Effects of ethnicity on glucose tolerance, insulin resistance and beta cell function in 223 women with an abnormal glucose challenge test during pregnancy. Aust N Z J Obstet Gynaecol. 2001;41:182–6.PubMedCrossRef
13.
Zurück zum Zitat Catalano PM, Roman-Drago NM, Amini SB, Sims EA. Longitudinal changes in body composition and energy balance in lean women with normal and abnormal glucose tolerance during pregnancy. Am J Obstet Gynecol. 1998;179:156–65.PubMedCrossRef Catalano PM, Roman-Drago NM, Amini SB, Sims EA. Longitudinal changes in body composition and energy balance in lean women with normal and abnormal glucose tolerance during pregnancy. Am J Obstet Gynecol. 1998;179:156–65.PubMedCrossRef
14.
Zurück zum Zitat • McIntyre HD, Chang AM, Callaway LK, Cowley DM, Dyer AR, Radaelli T, et al. Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study Cooperative Research Group. Hormonal and metabolic factors associated with variations in insulin sensitivity in human pregnancy. Diabetes Care. 2010;33:356–60. This article reports that leptin, triglycerides, and IGFBP1 levels were associated with insulin resistance measured in a subsample of the women participating to the landmark HAPO study.PubMedCrossRef • McIntyre HD, Chang AM, Callaway LK, Cowley DM, Dyer AR, Radaelli T, et al. Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study Cooperative Research Group. Hormonal and metabolic factors associated with variations in insulin sensitivity in human pregnancy. Diabetes Care. 2010;33:356–60. This article reports that leptin, triglycerides, and IGFBP1 levels were associated with insulin resistance measured in a subsample of the women participating to the landmark HAPO study.PubMedCrossRef
15.
Zurück zum Zitat Retnakaran R, Qi Y, Sermer M, Connelly PW, Zinman B, Hanley AJ. Pre-gravid physical activity and reduced risk of glucose intolerance in pregnancy: the role of insulin sensitivity. Clin Endocrinol. 2009;70:615–22.CrossRef Retnakaran R, Qi Y, Sermer M, Connelly PW, Zinman B, Hanley AJ. Pre-gravid physical activity and reduced risk of glucose intolerance in pregnancy: the role of insulin sensitivity. Clin Endocrinol. 2009;70:615–22.CrossRef
16.
Zurück zum Zitat Barbour LA, McCurdy CE, Hernandez TL, Kirwan JP, Catalano PM, Friedman JE. Cellular mechanisms for insulin resistance in normal pregnancy and gestational diabetes. Diabetes Care. 2007;30 Suppl 2:S112–9.PubMedCrossRef Barbour LA, McCurdy CE, Hernandez TL, Kirwan JP, Catalano PM, Friedman JE. Cellular mechanisms for insulin resistance in normal pregnancy and gestational diabetes. Diabetes Care. 2007;30 Suppl 2:S112–9.PubMedCrossRef
17.
Zurück zum Zitat Birnbaum MJ. Turning down insulin signaling. J Clin Invest. 2001;108:655–9.PubMed Birnbaum MJ. Turning down insulin signaling. J Clin Invest. 2001;108:655–9.PubMed
18.
Zurück zum Zitat Manning AK, Hivert MF, Scott RA, Grimsby JL, Bouatia-Naji N, Chen H, et al. A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance. Nat Genet. 2012;44:659–69.PubMedCrossRef Manning AK, Hivert MF, Scott RA, Grimsby JL, Bouatia-Naji N, Chen H, et al. A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance. Nat Genet. 2012;44:659–69.PubMedCrossRef
19.
Zurück zum Zitat Tok EC, Ertunc D, Bilgin O, Erdal EM, Kaplanoglu M, Dilek S. Association of insulin receptor substrate-1 G972R variant with baseline characteristics of the patients with gestational diabetes mellitus. Am J Obstet Gynecol. 2006;194:868–72.PubMedCrossRef Tok EC, Ertunc D, Bilgin O, Erdal EM, Kaplanoglu M, Dilek S. Association of insulin receptor substrate-1 G972R variant with baseline characteristics of the patients with gestational diabetes mellitus. Am J Obstet Gynecol. 2006;194:868–72.PubMedCrossRef
20.
Zurück zum Zitat Friedman JE, Ishizuka T, Shao J, Huston L, Highman T, Catalano P. Impaired glucose transport and insulin receptor tyrosine phosphorylation in skeletal muscle from obese women with gestational diabetes. Diabetes. 1999;48:1807–14.PubMedCrossRef Friedman JE, Ishizuka T, Shao J, Huston L, Highman T, Catalano P. Impaired glucose transport and insulin receptor tyrosine phosphorylation in skeletal muscle from obese women with gestational diabetes. Diabetes. 1999;48:1807–14.PubMedCrossRef
21.
Zurück zum Zitat Friedman JE, Kirwan JP, Jing M, Presley L, Catalano PM. Increased skeletal muscle tumor necrosis factor-alpha and impaired insulin signaling persist in obese women with gestational diabetes mellitus 1 year postpartum. Diabetes. 2008;57:606–13.PubMedCrossRef Friedman JE, Kirwan JP, Jing M, Presley L, Catalano PM. Increased skeletal muscle tumor necrosis factor-alpha and impaired insulin signaling persist in obese women with gestational diabetes mellitus 1 year postpartum. Diabetes. 2008;57:606–13.PubMedCrossRef
22.
Zurück zum Zitat Hivert MF, Sullivan LM, Fox CS, Nathan DM, D’Agostino RBS, Wilson PW, et al. Associations of adiponectin, resistin, and tumor necrosis factor-alpha with insulin resistance. J Clin Endocrinol Metab. 2008;93:3165–72.PubMedCrossRef Hivert MF, Sullivan LM, Fox CS, Nathan DM, D’Agostino RBS, Wilson PW, et al. Associations of adiponectin, resistin, and tumor necrosis factor-alpha with insulin resistance. J Clin Endocrinol Metab. 2008;93:3165–72.PubMedCrossRef
23.
Zurück zum Zitat Kirwan JP, Hauguel-De Mouzon S, Lepercq J, Challier JC, Huston-Presley L, et al. TNF-alpha is a predictor of insulin resistance in human pregnancy. Diabetes. 2002;51:2207–13.PubMedCrossRef Kirwan JP, Hauguel-De Mouzon S, Lepercq J, Challier JC, Huston-Presley L, et al. TNF-alpha is a predictor of insulin resistance in human pregnancy. Diabetes. 2002;51:2207–13.PubMedCrossRef
24.
Zurück zum Zitat Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372:425–32.PubMedCrossRef Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372:425–32.PubMedCrossRef
25.
Zurück zum Zitat Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334:292–5.PubMedCrossRef Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334:292–5.PubMedCrossRef
26.
Zurück zum Zitat Hukshorn CJ, Saris WH, Westerterp-Plantenga MS, Farid AR, Smith FJ, Campfield LA. Weekly subcutaneous pegylated recombinant native human leptin (PEG-OB) administration in obese men. J Clin Endocrinol Metab. 2000;85:4003–9.PubMedCrossRef Hukshorn CJ, Saris WH, Westerterp-Plantenga MS, Farid AR, Smith FJ, Campfield LA. Weekly subcutaneous pegylated recombinant native human leptin (PEG-OB) administration in obese men. J Clin Endocrinol Metab. 2000;85:4003–9.PubMedCrossRef
28.
Zurück zum Zitat Hoggard N, Hunter L, Duncan JS, Williams LM, Trayhurn P, Mercer JG. Leptin and leptin receptor mRNA and protein expression in the murine fetus and placenta. Proc Natl Acad Sci U S A. 1997;94:11073–8.PubMedCrossRef Hoggard N, Hunter L, Duncan JS, Williams LM, Trayhurn P, Mercer JG. Leptin and leptin receptor mRNA and protein expression in the murine fetus and placenta. Proc Natl Acad Sci U S A. 1997;94:11073–8.PubMedCrossRef
29.
Zurück zum Zitat Masuzaki H, Ogawa Y, Sagawa N, Hosoda K, Matsumoto T, Mise H, et al. Nonadipose tissue production of leptin: leptin as a novel placenta-derived hormone in humans. Nat Med. 1997;3:1029–33.PubMedCrossRef Masuzaki H, Ogawa Y, Sagawa N, Hosoda K, Matsumoto T, Mise H, et al. Nonadipose tissue production of leptin: leptin as a novel placenta-derived hormone in humans. Nat Med. 1997;3:1029–33.PubMedCrossRef
30.
Zurück zum Zitat Lappas M, Yee K, Permezel M, Rice GE. Release and regulation of leptin, resistin and adiponectin from human placenta, fetal membranes, and maternal adipose tissue and skeletal muscle from normal and gestational diabetes mellitus-complicated pregnancies. J Endocrinol. 2005;186:457–65.PubMedCrossRef Lappas M, Yee K, Permezel M, Rice GE. Release and regulation of leptin, resistin and adiponectin from human placenta, fetal membranes, and maternal adipose tissue and skeletal muscle from normal and gestational diabetes mellitus-complicated pregnancies. J Endocrinol. 2005;186:457–65.PubMedCrossRef
31.
Zurück zum Zitat Radaelli T, Varastehpour A, Catalano P, Hauguel-de Mouzon S. Gestational diabetes induces placental genes for chronic stress and inflammatory pathways. Diabetes. 2003;52:2951–8.PubMedCrossRef Radaelli T, Varastehpour A, Catalano P, Hauguel-de Mouzon S. Gestational diabetes induces placental genes for chronic stress and inflammatory pathways. Diabetes. 2003;52:2951–8.PubMedCrossRef
32.
Zurück zum Zitat Meller M, Qiu C, Vadachkoria S, Abetew DF, Luthy DA, Williams MA. Changes in placental adipocytokine gene expression associated with gestational diabetes mellitus. Physiol Res. 2006;55:501–12.PubMed Meller M, Qiu C, Vadachkoria S, Abetew DF, Luthy DA, Williams MA. Changes in placental adipocytokine gene expression associated with gestational diabetes mellitus. Physiol Res. 2006;55:501–12.PubMed
33.
Zurück zum Zitat Enquobahrie DA, Williams MA, Qiu C, Meller M, Sorensen TK. Global placental gene expression in gestational diabetes mellitus. Am J Obstet Gynecol. 2009;200:206.e1–13.CrossRef Enquobahrie DA, Williams MA, Qiu C, Meller M, Sorensen TK. Global placental gene expression in gestational diabetes mellitus. Am J Obstet Gynecol. 2009;200:206.e1–13.CrossRef
34.
Zurück zum Zitat McLachlan KA, O’Neal D, Jenkins A, Alford FP. Do adiponectin, TNFalpha, leptin, and CRP relate to insulin resistance in pregnancy? Studies in women with and without gestational diabetes, during and after pregnancy. Diabetes Metab Res Rev. 2006;22:131–8.PubMedCrossRef McLachlan KA, O’Neal D, Jenkins A, Alford FP. Do adiponectin, TNFalpha, leptin, and CRP relate to insulin resistance in pregnancy? Studies in women with and without gestational diabetes, during and after pregnancy. Diabetes Metab Res Rev. 2006;22:131–8.PubMedCrossRef
35.
Zurück zum Zitat Vahamiko S, Isolauri E, Pesonen U, Koskinen P, Ekblad U, Laitinen K. Dietary sucrose intake is related to serum leptin concentration in overweight pregnant women. Eur J Nutr. 2010;49:83–90.PubMedCrossRef Vahamiko S, Isolauri E, Pesonen U, Koskinen P, Ekblad U, Laitinen K. Dietary sucrose intake is related to serum leptin concentration in overweight pregnant women. Eur J Nutr. 2010;49:83–90.PubMedCrossRef
36.
Zurück zum Zitat Pajvani UB, Du X, Combs TP, Berg AH, Rajala MW, Schulthess T, et al. Structure-function studies of the adipocyte-secreted hormone Acrp30/adiponectin. Implications fpr metabolic regulation and bioactivity. J Biol Chem. 2003;278:9073–85.PubMedCrossRef Pajvani UB, Du X, Combs TP, Berg AH, Rajala MW, Schulthess T, et al. Structure-function studies of the adipocyte-secreted hormone Acrp30/adiponectin. Implications fpr metabolic regulation and bioactivity. J Biol Chem. 2003;278:9073–85.PubMedCrossRef
37.
Zurück zum Zitat Lacroix M, Battista M, Doyon M, Ménard J, Ardilouze JL, Perron P, Hivert M. Lower adiponectin levels at first trimester of pregnancy are associated with increase insulin resistance and higher risk of gestational diabetes mellitus. Diabetes Care. 2013;36:1–7. Lacroix M, Battista M, Doyon M, Ménard J, Ardilouze JL, Perron P, Hivert M. Lower adiponectin levels at first trimester of pregnancy are associated with increase insulin resistance and higher risk of gestational diabetes mellitus. Diabetes Care. 2013;36:1–7.
38.
Zurück zum Zitat Retnakaran R, Hanley AJ, Raif N, Hirning CR, Connelly PW, Sermer M, et al. Adiponectin and beta cell dysfunction in gestational diabetes: pathophysiological implications. Diabetologia. 2005;48:993–1001.PubMedCrossRef Retnakaran R, Hanley AJ, Raif N, Hirning CR, Connelly PW, Sermer M, et al. Adiponectin and beta cell dysfunction in gestational diabetes: pathophysiological implications. Diabetologia. 2005;48:993–1001.PubMedCrossRef
39.
Zurück zum Zitat Ranheim T, Haugen F, Staff AC, Braekke K, Harsem NK, Drevon CA. Adiponectin is reduced in gestational diabetes mellitus in normal weight women. Acta Obstet Gynecol Scand. 2004;83:341–7.PubMed Ranheim T, Haugen F, Staff AC, Braekke K, Harsem NK, Drevon CA. Adiponectin is reduced in gestational diabetes mellitus in normal weight women. Acta Obstet Gynecol Scand. 2004;83:341–7.PubMed
40.
Zurück zum Zitat Mingrone G, Manco M, Mora ME, Guidone C, Iaconelli A, Gniuli D, et al. Influence of maternal obesity on insulin sensitivity and secretion in offspring. Diabetes Care. 2008;31:1872–6.PubMedCrossRef Mingrone G, Manco M, Mora ME, Guidone C, Iaconelli A, Gniuli D, et al. Influence of maternal obesity on insulin sensitivity and secretion in offspring. Diabetes Care. 2008;31:1872–6.PubMedCrossRef
41.
Zurück zum Zitat • Catalano PM, Presley L, Minium J, Hauguel-de Mouzon S. Fetuses of obese mothers develop insulin resistance in utero. Diabetes Care. 2009;32:1076–80. This study presented the impact of maternal obesity (with normal glucose tolerance) on maternal and newborn metabolic profiles: offspring from obese mothers had higher insulin resistance (HOMA-IR) and higher leptin levels measured in cord blood.PubMedCrossRef • Catalano PM, Presley L, Minium J, Hauguel-de Mouzon S. Fetuses of obese mothers develop insulin resistance in utero. Diabetes Care. 2009;32:1076–80. This study presented the impact of maternal obesity (with normal glucose tolerance) on maternal and newborn metabolic profiles: offspring from obese mothers had higher insulin resistance (HOMA-IR) and higher leptin levels measured in cord blood.PubMedCrossRef
42.
Zurück zum Zitat HAPO Study Cooperative Research Group. Hyperglycaemia and Adverse Pregnancy Outcome (HAPO) Study: associations with maternal body mass index. BJOG. 2010;117:575–84.CrossRef HAPO Study Cooperative Research Group. Hyperglycaemia and Adverse Pregnancy Outcome (HAPO) Study: associations with maternal body mass index. BJOG. 2010;117:575–84.CrossRef
43.
Zurück zum Zitat Pettitt DJ, Aleck KA, Baird HR, Carraher MJ, Bennett PH, Knowler WC. Congenital susceptibility to NIDDM. Role of intrauterine environment. Diabetes. 1988;37:622–8.PubMedCrossRef Pettitt DJ, Aleck KA, Baird HR, Carraher MJ, Bennett PH, Knowler WC. Congenital susceptibility to NIDDM. Role of intrauterine environment. Diabetes. 1988;37:622–8.PubMedCrossRef
44.
Zurück zum Zitat •• Krishnaveni GV, Veena SR, Hill JC, Kehoe S, Karat SC, Fall CH. Intrauterine exposure to maternal diabetes is associated with higher adiposity and insulin resistance and clustering of cardiovascular risk markers in Indian children. Diabetes Care. 2010;33:402–4. This article reports the association between GDM exposure and the risk of metabolic abnormality in children; insulin resistance was higher in children exposed to GDM (and the associations were stronger at 9 years of age than at 5 years of age).PubMedCrossRef •• Krishnaveni GV, Veena SR, Hill JC, Kehoe S, Karat SC, Fall CH. Intrauterine exposure to maternal diabetes is associated with higher adiposity and insulin resistance and clustering of cardiovascular risk markers in Indian children. Diabetes Care. 2010;33:402–4. This article reports the association between GDM exposure and the risk of metabolic abnormality in children; insulin resistance was higher in children exposed to GDM (and the associations were stronger at 9 years of age than at 5 years of age).PubMedCrossRef
45.
Zurück zum Zitat Boerschmann H, Pfluger M, Henneberger L, Ziegler AG, Hummel S. Prevalence and predictors of overweight and insulin resistance in offspring of mothers with gestational diabetes mellitus. Diabetes Care. 2010;33:1845–9.PubMedCrossRef Boerschmann H, Pfluger M, Henneberger L, Ziegler AG, Hummel S. Prevalence and predictors of overweight and insulin resistance in offspring of mothers with gestational diabetes mellitus. Diabetes Care. 2010;33:1845–9.PubMedCrossRef
46.
Zurück zum Zitat Egeland GM, Meltzer SJ. Following in mother’s footsteps? Mother-daughter risks for insulin resistance and cardiovascular disease 15 years after gestational diabetes. Diabet Med. 2010;27:257–65.PubMedCrossRef Egeland GM, Meltzer SJ. Following in mother’s footsteps? Mother-daughter risks for insulin resistance and cardiovascular disease 15 years after gestational diabetes. Diabet Med. 2010;27:257–65.PubMedCrossRef
47.
Zurück zum Zitat Boney CM, Verma A, Tucker R, Vohr BR. Metabolic syndrome in childhood: association with birth weight, maternal obesity, and gestational diabetes mellitus. Pediatrics. 2005;115:e290–6.PubMedCrossRef Boney CM, Verma A, Tucker R, Vohr BR. Metabolic syndrome in childhood: association with birth weight, maternal obesity, and gestational diabetes mellitus. Pediatrics. 2005;115:e290–6.PubMedCrossRef
48.
Zurück zum Zitat Gillman MW, Oakey H, Baghurst PA, Volkmer RE, Robinson JS, Crowther CA. Effect of treatment of gestational diabetes mellitus on obesity in the next generation. Diabetes Care. 2010;33:964–8.PubMedCrossRef Gillman MW, Oakey H, Baghurst PA, Volkmer RE, Robinson JS, Crowther CA. Effect of treatment of gestational diabetes mellitus on obesity in the next generation. Diabetes Care. 2010;33:964–8.PubMedCrossRef
49.
Zurück zum Zitat Bouchard L, Thibault S, Guay SP, Santure M, Monpetit A, St-Pierre J, et al. Leptin gene epigenetic adaptation to impaired glucose metabolism during pregnancy. Diabetes Care. 2010;33:2436–41.PubMedCrossRef Bouchard L, Thibault S, Guay SP, Santure M, Monpetit A, St-Pierre J, et al. Leptin gene epigenetic adaptation to impaired glucose metabolism during pregnancy. Diabetes Care. 2010;33:2436–41.PubMedCrossRef
50.
Zurück zum Zitat St-Pierre J, Hivert MF, Perron P, Poirier P, Guay SP, Brisson D. IGF2 DNA methylation is a modulator of newborn’s fetal growth and development. Epigenetics. 2012;7:1125–32.PubMedCrossRef St-Pierre J, Hivert MF, Perron P, Poirier P, Guay SP, Brisson D. IGF2 DNA methylation is a modulator of newborn’s fetal growth and development. Epigenetics. 2012;7:1125–32.PubMedCrossRef
51.
Zurück zum Zitat •• Chiavaroli V, Giannini C, D’Adamo E, de Giorgis T, Chiarelli F, Mohn A. Insulin resistance and oxidative stress in children born small and large for gestational age. Pediatrics. 2009;124:695–702. This article reports the higher insulin resistance observed in children at both ends of the spectrum for birth weight (small and large for gestational age) and the combined deleterious effect of current of excess weight at 6–8 years old.PubMedCrossRef •• Chiavaroli V, Giannini C, D’Adamo E, de Giorgis T, Chiarelli F, Mohn A. Insulin resistance and oxidative stress in children born small and large for gestational age. Pediatrics. 2009;124:695–702. This article reports the higher insulin resistance observed in children at both ends of the spectrum for birth weight (small and large for gestational age) and the combined deleterious effect of current of excess weight at 6–8 years old.PubMedCrossRef
52.
Zurück zum Zitat Hales CN, Barker DJ. Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis. Diabetologia. 1992;35:595–601.PubMedCrossRef Hales CN, Barker DJ. Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis. Diabetologia. 1992;35:595–601.PubMedCrossRef
53.
Zurück zum Zitat Phillips DI, Barker DJ, Hales CN, Hirst S, Osmond C. Thinness at birth and insulin resistance in adult life. Diabetologia. 1994;37:150–4.PubMedCrossRef Phillips DI, Barker DJ, Hales CN, Hirst S, Osmond C. Thinness at birth and insulin resistance in adult life. Diabetologia. 1994;37:150–4.PubMedCrossRef
54.
Zurück zum Zitat Ravelli AC, van der Meulen JH, Michels RP, Osmond C, Barker DJ, Hales CN, et al. Glucose tolerance in adults after prenatal exposure to famine. Lancet. 1998;351:173–7.PubMedCrossRef Ravelli AC, van der Meulen JH, Michels RP, Osmond C, Barker DJ, Hales CN, et al. Glucose tolerance in adults after prenatal exposure to famine. Lancet. 1998;351:173–7.PubMedCrossRef
55.
Zurück zum Zitat Flanagan DE, Moore VM, Godsland IF, Cockington RA, Robinson JS, Phillips DI. Fetal growth and the physiological control of glucose tolerance in adults: a minimal model analysis. Am J Physiol Endocrinol Metab. 2000;278:E700–6.PubMed Flanagan DE, Moore VM, Godsland IF, Cockington RA, Robinson JS, Phillips DI. Fetal growth and the physiological control of glucose tolerance in adults: a minimal model analysis. Am J Physiol Endocrinol Metab. 2000;278:E700–6.PubMed
56.
Zurück zum Zitat Jaquet D, Gaboriau A, Czernichow P, Levy-Marchal C. Insulin resistance early in adulthood in subjects born with intrauterine growth retardation. J Clin Endocrinol Metab. 2000;85:1401–6.PubMedCrossRef Jaquet D, Gaboriau A, Czernichow P, Levy-Marchal C. Insulin resistance early in adulthood in subjects born with intrauterine growth retardation. J Clin Endocrinol Metab. 2000;85:1401–6.PubMedCrossRef
57.
Zurück zum Zitat Bavdekar A, Yajnik CS, Fall CH, Bapat S, Pandit AN, Deshpande V, et al. Insulin resistance syndrome in 8-year-old Indian children: small at birth, big at 8 years, or both? Diabetes. 1999;48:2422–9.PubMedCrossRef Bavdekar A, Yajnik CS, Fall CH, Bapat S, Pandit AN, Deshpande V, et al. Insulin resistance syndrome in 8-year-old Indian children: small at birth, big at 8 years, or both? Diabetes. 1999;48:2422–9.PubMedCrossRef
58.
Zurück zum Zitat Levy-Marchal C, Jaquet D. Long-term metabolic consequences of being born small for gestational age. Pediatr Diabetes. 2004;5:147–53.PubMedCrossRef Levy-Marchal C, Jaquet D. Long-term metabolic consequences of being born small for gestational age. Pediatr Diabetes. 2004;5:147–53.PubMedCrossRef
59.
Zurück zum Zitat Ong KK, Petry CJ, Emmett PM, Sandhu MS, Kiess W, Hales CN, et al. ALSPAC study team. Insulin sensitivity and secretion in normal children related to size at birth, postnatal growth, and plasma insulin-like growth factor-I levels. Diabetologia. 2004;47:1064–70.PubMed Ong KK, Petry CJ, Emmett PM, Sandhu MS, Kiess W, Hales CN, et al. ALSPAC study team. Insulin sensitivity and secretion in normal children related to size at birth, postnatal growth, and plasma insulin-like growth factor-I levels. Diabetologia. 2004;47:1064–70.PubMed
60.
Zurück zum Zitat Veena SR, Krishnaveni GV, Wills AK, Hill JC, Karat SC, Fall CH. Glucose tolerance and insulin resistance in Indian children: relationship to infant feeding pattern. Diabetologia. 2011;54:2533–7.PubMedCrossRef Veena SR, Krishnaveni GV, Wills AK, Hill JC, Karat SC, Fall CH. Glucose tolerance and insulin resistance in Indian children: relationship to infant feeding pattern. Diabetologia. 2011;54:2533–7.PubMedCrossRef
61.
Zurück zum Zitat Williams DM, Martin RM, Davey Smith G, Alberti KG, Ben-Shlomo Y, McCarthy A. Associations of infant nutrition with insulin resistance measures in early adulthood: evidence from the Barry-Caerphilly Growth (BCG) study. PLoS One. 2012;7:e34161.PubMedCrossRef Williams DM, Martin RM, Davey Smith G, Alberti KG, Ben-Shlomo Y, McCarthy A. Associations of infant nutrition with insulin resistance measures in early adulthood: evidence from the Barry-Caerphilly Growth (BCG) study. PLoS One. 2012;7:e34161.PubMedCrossRef
62.
Zurück zum Zitat Slining MM, Kuzawa CW, Mayer-Davis EJ, Adair LS. Evaluating the indirect effect of infant weight velocity on insulin resistance in young adulthood: a birth cohort study from the Philippines. Am J Epidemiol. 2011;173:640–8.PubMedCrossRef Slining MM, Kuzawa CW, Mayer-Davis EJ, Adair LS. Evaluating the indirect effect of infant weight velocity on insulin resistance in young adulthood: a birth cohort study from the Philippines. Am J Epidemiol. 2011;173:640–8.PubMedCrossRef
63.
Zurück zum Zitat Raghupathy P, Antonisamy B, Geethanjali FS, Saperia J, Leary SD, Priya G, et al. Glucose tolerance, insulin resistance, and insulin secretion in young south Indian adults: relationships to parental size, neonatal size, and childhood body mass index. Diabetes Res Clin Pract. 2010;87:283–92.PubMedCrossRef Raghupathy P, Antonisamy B, Geethanjali FS, Saperia J, Leary SD, Priya G, et al. Glucose tolerance, insulin resistance, and insulin secretion in young south Indian adults: relationships to parental size, neonatal size, and childhood body mass index. Diabetes Res Clin Pract. 2010;87:283–92.PubMedCrossRef
64.
Zurück zum Zitat Pearce MS, Unwin NC, Parker L, Alberti KG. Life course determinants of insulin secretion and sensitivity at age 50 years: the Newcastle thousand families study. Diabetes Metab Res Rev. 2006;22:118–25.PubMedCrossRef Pearce MS, Unwin NC, Parker L, Alberti KG. Life course determinants of insulin secretion and sensitivity at age 50 years: the Newcastle thousand families study. Diabetes Metab Res Rev. 2006;22:118–25.PubMedCrossRef
Metadaten
Titel
Maternal/Fetal Determinants of Insulin Resistance in Women During Pregnancy and in Offspring Over Life
verfasst von
Marilyn Lacroix
Eralda Kina
Marie-France Hivert
Publikationsdatum
01.04.2013
Verlag
Current Science Inc.
Erschienen in
Current Diabetes Reports / Ausgabe 2/2013
Print ISSN: 1534-4827
Elektronische ISSN: 1539-0829
DOI
https://doi.org/10.1007/s11892-012-0360-x

Weitere Artikel der Ausgabe 2/2013

Current Diabetes Reports 2/2013 Zur Ausgabe

Issues in the Nutritional Treatment of Type 2 Diabetes and Obesity (E Mayer-Davis, Section Editor)

Omega-3 and Omega-6 Fatty Acids and Type 2 Diabetes

Health Care Delivery Systems in Diabetes (D Wexler, Section Editor)

The Role of Community Health Workers in Diabetes: Update on Current Literature

Issues in the Nutritional Treatment of Type 2 Diabetes and Obesity (E Mayer-Davis, Section Editor)

Role of Vitamin D in the Development of Insulin Resistance and Type 2 Diabetes

Issues in the Nutritional Treatment of Type 2 Diabetes and Obesity (E Mayer-Davis, Section Editor)

Meat Consumption, Diabetes, and Its Complications

Health Care Delivery Systems in Diabetes (D Wexler, Section Editor)

Pay-for-Performance: Impact on Diabetes

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

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