Skip to main content
Erschienen in: International Journal of Diabetes in Developing Countries 2/2019

07.03.2018 | Review Article

Gestational diabetes is associated to the development of brain insulin resistance in the offspring

verfasst von: Ricardo Augusto Leoni De Sousa

Erschienen in: International Journal of Diabetes in Developing Countries | Ausgabe 2/2019

Einloggen, um Zugang zu erhalten

Abstract

Gestational diabetes mellitus (GDM) is defined as a form of glucose intolerance. Evidences for late metabolic and behavioral consequences to offspring born from GDM are emerging. More recent and concerning evidences point to detrimental effects of GDM on the behavior and cognitive capacity of the offspring. We aimed to review what is known about the consequences of GDM to brain and behavior of the offspring. A research was made in PubMed using the words gestational diabetes, insulin resistance, memory, cognition, brain, and offspring. The most relevant papers according to citations and with results suggesting that hyperglycemia in pregnancy is associated with inflammation and some mechanisms which are potentially involved with the late brain insulin resistance and its consequences in the GDM’s offspring brain were selected. An increased risk of developing neuropsychiatric disorders to offspring from GDM mothers has been suggested. Transient intra-uterine exposure to high glucose levels can interfere with neuronal integrity, survival and connectivity to offspring’s brain. Fighting neuroinflammation during gestational period may avoid susceptibility for neurodegenerative disorders in the offspring.
Literatur
3.
Zurück zum Zitat Desisto CL, Kim SY, Sharma AJ. Prevalence estimates of gestational diabetes mellitus in the United States, pregnancy risk assessment monitoring system ( PRAMS ), 2007–2010. Prev Chronic Dis. 2014;11(12):1–9. Desisto CL, Kim SY, Sharma AJ. Prevalence estimates of gestational diabetes mellitus in the United States, pregnancy risk assessment monitoring system ( PRAMS ), 2007–2010. Prev Chronic Dis. 2014;11(12):1–9.
5.
Zurück zum Zitat Kim SY, England L, Sappenfield W, Hoyt G, Bish CL, Salihu HM, et al. Racial/ethnic differences in the percentage of gestational diabetes mellitus cases attributable to overweight and obesity, Florida, 2004-2007. Prevention. 2012;9(6):2012. Kim SY, England L, Sappenfield W, Hoyt G, Bish CL, Salihu HM, et al. Racial/ethnic differences in the percentage of gestational diabetes mellitus cases attributable to overweight and obesity, Florida, 2004-2007. Prevention. 2012;9(6):2012.
6.
Zurück zum Zitat Cypryk K, Szymczak W, Czupryniak L, Sobczak M. Gestational diabetes mellitus—an analysis of risk factors Cukrzyca ciążowa—analiza czynników ryzyka. Endokrynol Pol. 2008;59(5):393–7.PubMed Cypryk K, Szymczak W, Czupryniak L, Sobczak M. Gestational diabetes mellitus—an analysis of risk factors Cukrzyca ciążowa—analiza czynników ryzyka. Endokrynol Pol. 2008;59(5):393–7.PubMed
7.
Zurück zum Zitat Mills JL, Baker L, Goldman AS. Malformations in infants of diabetic mothers occur before the seventh gestational week implications for treatment. Diabetes. 1979;28:292–3.CrossRefPubMed Mills JL, Baker L, Goldman AS. Malformations in infants of diabetic mothers occur before the seventh gestational week implications for treatment. Diabetes. 1979;28:292–3.CrossRefPubMed
8.
Zurück zum Zitat Sharpe PB, Chan A, Haan E A, Hiller JE. Maternal diabetes and congenital anomalies in South Australia 1986-2000: a population-based cohort study. Birth Defects Res A Clin Mol Teratol [Internet]. 2005 [cited 2016 Dec 18];73(9):605–11. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16007590 . Sharpe PB, Chan A, Haan E A, Hiller JE. Maternal diabetes and congenital anomalies in South Australia 1986-2000: a population-based cohort study. Birth Defects Res A Clin Mol Teratol [Internet]. 2005 [cited 2016 Dec 18];73(9):605–11. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16007590 .
9.
10.
Zurück zum Zitat Frías JL, Frías JP, Frías PA, Frías MLM. Infrequently studied congenital anomalies as clues to the diagnosis of maternal diabetes mellitus. Am J Med Genet Part A. 2007;2909:2904–9. Frías JL, Frías JP, Frías PA, Frías MLM. Infrequently studied congenital anomalies as clues to the diagnosis of maternal diabetes mellitus. Am J Med Genet Part A. 2007;2909:2904–9.
11.
Zurück zum Zitat Jing Y, Song Y, Yao Y, Yin J, Wang D, Gao L. Retardation of fetal dendritic development induced by gestational hyperglycemia is associated with brain insulin/IGF-I signals. Int J Dev Neurosci. 2014;37:15–20.CrossRefPubMed Jing Y, Song Y, Yao Y, Yin J, Wang D, Gao L. Retardation of fetal dendritic development induced by gestational hyperglycemia is associated with brain insulin/IGF-I signals. Int J Dev Neurosci. 2014;37:15–20.CrossRefPubMed
12.
Zurück zum Zitat Boulet SL, Alexander GR, Salihu HM, Pass M. Macrosomic births in the United States: determinants, outcomes, and proposed grades of risk. Am J Obstet Gynecol. 2003;188:1372–8.CrossRefPubMed Boulet SL, Alexander GR, Salihu HM, Pass M. Macrosomic births in the United States: determinants, outcomes, and proposed grades of risk. Am J Obstet Gynecol. 2003;188:1372–8.CrossRefPubMed
13.
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 [Internet]. 2006 [cited 2017 Feb 5];22(2):131–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16170833 . 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 [Internet]. 2006 [cited 2017 Feb 5];22(2):131–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16170833 .
14.
Zurück zum Zitat Fu J, Tay SSW, Ling E A, Dheen ST. High glucose alters the expression of genes involved in proliferation and cell-fate specification of embryonic neural stem cells. Diabetologia [Internet]. 2006 [cited 2016 Dec 18];49(5):1027–38. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16508779 . Fu J, Tay SSW, Ling E A, Dheen ST. High glucose alters the expression of genes involved in proliferation and cell-fate specification of embryonic neural stem cells. Diabetologia [Internet]. 2006 [cited 2016 Dec 18];49(5):1027–38. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16508779 .
15.
Zurück zum Zitat Liao DM, Ng YK, Tay SSW, Ling E A, Dheen ST. Altered gene expression with abnormal patterning of the telencephalon in embryos of diabetic Albino Swiss mice. Diabetologia [Internet]. 2004 [cited 2016 Dec 18];47(3):523–31. Available from: http://www.ncbi.nlm.nih.gov/pubmed/14963649 . Liao DM, Ng YK, Tay SSW, Ling E A, Dheen ST. Altered gene expression with abnormal patterning of the telencephalon in embryos of diabetic Albino Swiss mice. Diabetologia [Internet]. 2004 [cited 2016 Dec 18];47(3):523–31. Available from: http://www.ncbi.nlm.nih.gov/pubmed/14963649 .
16.
Zurück zum Zitat Bomfim TR, Forny-germano L, Sathler LB, Brito-moreira J, Houzel J, Decker H, et al. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease-associated Aβ oligomers. J Clin Invest. 2012;122(4):1339–53.CrossRefPubMedPubMedCentral Bomfim TR, Forny-germano L, Sathler LB, Brito-moreira J, Houzel J, Decker H, et al. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease-associated Aβ oligomers. J Clin Invest. 2012;122(4):1339–53.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Clarke JR, Lyra NM, Figueiredo CP, Frozza RL, Ledo JH, Beckman D, et al. Alzheimer-associated Aβ oligomers impact the central nervous system to induce peripheral metabolic deregulation. EMBO Mol Med. 2015;7(2):190–211.CrossRefPubMedPubMedCentral Clarke JR, Lyra NM, Figueiredo CP, Frozza RL, Ledo JH, Beckman D, et al. Alzheimer-associated Aβ oligomers impact the central nervous system to induce peripheral metabolic deregulation. EMBO Mol Med. 2015;7(2):190–211.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Ferreira ST, Clarke JR, Bomfim TR, De Felice FG. Inflammation, defective insulin signaling, and neuronal dysfunction in Alzheimer’s disease. Alzheimers Dement [Internet]. Elsevier; 2014 [cited 2015 Mar 28];10(1 Suppl):S76–83. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24529528 . Ferreira ST, Clarke JR, Bomfim TR, De Felice FG. Inflammation, defective insulin signaling, and neuronal dysfunction in Alzheimer’s disease. Alzheimers Dement [Internet]. Elsevier; 2014 [cited 2015 Mar 28];10(1 Suppl):S76–83. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24529528 .
19.
20.
Zurück zum Zitat Chandna AR, Kuhlmann N, Bryce C A, Greba Q, Campanucci V A, Howland JG. Chronic maternal hyperglycemia induced during mid-pregnancy in rats increases RAGE expression, augments hippocampal excitability, and alters behavior of the offspring. Neuroscience [Internet]. IBRO; 2015 [cited 2016 Dec 20];303:241–60. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26151680 . Chandna AR, Kuhlmann N, Bryce C A, Greba Q, Campanucci V A, Howland JG. Chronic maternal hyperglycemia induced during mid-pregnancy in rats increases RAGE expression, augments hippocampal excitability, and alters behavior of the offspring. Neuroscience [Internet]. IBRO; 2015 [cited 2016 Dec 20];303:241–60. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26151680 .
22.
Zurück zum Zitat Hami J, Sadr-nabavi A, Sankian M, Baladi-Mood M, Haghir H. The effects of maternal diabetes on expression of insulin-like growth factor-1 and insulin receptors in male developing rat hippocampus. Brain Strcture Funct. 2013;218:73–84.CrossRef Hami J, Sadr-nabavi A, Sankian M, Baladi-Mood M, Haghir H. The effects of maternal diabetes on expression of insulin-like growth factor-1 and insulin receptors in male developing rat hippocampus. Brain Strcture Funct. 2013;218:73–84.CrossRef
23.
Zurück zum Zitat Linder K, Schleger F, Kiefer-Schmidt I, Fritsche L, Kümmel S, Heni M, et al. Gestational diabetes impairs human fetal postprandial brain activity. J Clin Endocrinol Metab [Internet]. 2015 [cited 2016 Oct 22];100(11):4029–36. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26465393 . Linder K, Schleger F, Kiefer-Schmidt I, Fritsche L, Kümmel S, Heni M, et al. Gestational diabetes impairs human fetal postprandial brain activity. J Clin Endocrinol Metab [Internet]. 2015 [cited 2016 Oct 22];100(11):4029–36. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26465393 .
24.
25.
Zurück zum Zitat Nelson CA, Wewerka S, Thomas KM, Tribby-walbridge S, Georgieff M. Neurocognitive sequelae of infants of diabetic mothers. Behav Neurosci. 2000;114(5):950–6.CrossRefPubMed Nelson CA, Wewerka S, Thomas KM, Tribby-walbridge S, Georgieff M. Neurocognitive sequelae of infants of diabetic mothers. Behav Neurosci. 2000;114(5):950–6.CrossRefPubMed
26.
Zurück zum Zitat Tanigawa K, Kawaguchi M, Tanaka O, Kato Y. Skeletal malformations in rat offspring long-term effect of maternal insulin-induced hypoglycemia during organogenesis. Diabetes. 1991;40:1115–21.CrossRefPubMed Tanigawa K, Kawaguchi M, Tanaka O, Kato Y. Skeletal malformations in rat offspring long-term effect of maternal insulin-induced hypoglycemia during organogenesis. Diabetes. 1991;40:1115–21.CrossRefPubMed
27.
Zurück zum Zitat Vind BF, Birk JB, Vienberg SG, Andersen B, Beck-Nielsen H, Wojtaszewski JFP, et al. Hyperglycaemia normalises insulin action on glucose metabolism but not the impaired activation of AKT and glycogen synthase in the skeletal muscle of patients with type 2 diabetes. Diabetologia [Internet]. 2012 [cited 2014 Jul 18];55(5):1435–45. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22322917 . Vind BF, Birk JB, Vienberg SG, Andersen B, Beck-Nielsen H, Wojtaszewski JFP, et al. Hyperglycaemia normalises insulin action on glucose metabolism but not the impaired activation of AKT and glycogen synthase in the skeletal muscle of patients with type 2 diabetes. Diabetologia [Internet]. 2012 [cited 2014 Jul 18];55(5):1435–45. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22322917 .
29.
Zurück zum Zitat Fröjdö S, Vidal H, Pirola L. Alterations of insulin signaling in type 2 diabetes: a review of the current evidence from humans. Biochim Biophys Acta [Internet]. Elsevier B.V.; 2009 [cited 2014 Jul 18];1792(2):83–92. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19041393 . Fröjdö S, Vidal H, Pirola L. Alterations of insulin signaling in type 2 diabetes: a review of the current evidence from humans. Biochim Biophys Acta [Internet]. Elsevier B.V.; 2009 [cited 2014 Jul 18];1792(2):83–92. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19041393 .
30.
Zurück zum Zitat Carvalho CRO, Carvalheira JBC, Lima MHM, Zimmerman SF, Caperuto LC, Amanso A, et al. Novel signal transduction pathway for luteinizing hormone and its interaction with insulin: activation of Janus kinase/signal transducer and activator of transcription and phosphoinositol 3-kinase/Akt pathways. Endocrinology [Internet]. 2003 [cited 2015 Jan 2];144(2):638–47. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12538627 . Carvalho CRO, Carvalheira JBC, Lima MHM, Zimmerman SF, Caperuto LC, Amanso A, et al. Novel signal transduction pathway for luteinizing hormone and its interaction with insulin: activation of Janus kinase/signal transducer and activator of transcription and phosphoinositol 3-kinase/Akt pathways. Endocrinology [Internet]. 2003 [cited 2015 Jan 2];144(2):638–47. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12538627 .
31.
32.
Zurück zum Zitat Folli F, Saad MJ, Backer JM, Kahn CR. Insulin stimulation of phosphatidylinositol 3-kinase activity and association with insulin receptor substrate 1 in liver and muscle of the intact rat *. J Biol Chem. 1992;267:22171–7.PubMed Folli F, Saad MJ, Backer JM, Kahn CR. Insulin stimulation of phosphatidylinositol 3-kinase activity and association with insulin receptor substrate 1 in liver and muscle of the intact rat *. J Biol Chem. 1992;267:22171–7.PubMed
33.
Zurück zum Zitat Chowdhury KK, Legare DJ, Lautt WW. Exercise enhancement of hepatic insulin-sensitising substance-mediated glucose uptake in diet-induced prediabetic rats. Br J Nutr [Internet]. 2013 [cited 2014 Jul 18];109(5):844–52. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23021417 . Chowdhury KK, Legare DJ, Lautt WW. Exercise enhancement of hepatic insulin-sensitising substance-mediated glucose uptake in diet-induced prediabetic rats. Br J Nutr [Internet]. 2013 [cited 2014 Jul 18];109(5):844–52. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23021417 .
34.
Zurück zum Zitat Backer JM, Myers MG Jr, Shoelson SE, Chin DJ, Sun X, Hu P, et al. Phosphatidylinositol 3′-kinase is activated by association with IRS-1 during insulin stimulation. EMBO J. 1992;11(9):3469–79.CrossRefPubMedPubMedCentral Backer JM, Myers MG Jr, Shoelson SE, Chin DJ, Sun X, Hu P, et al. Phosphatidylinositol 3′-kinase is activated by association with IRS-1 during insulin stimulation. EMBO J. 1992;11(9):3469–79.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Fantin VR, Wang Q, Lienhard GE, Keller SR, Valeria R. Mice lacking insulin receptor substrate 4 exhibit mild defects in growth, reproduction, and glucose homeostasis. Am J Physiol Endocrinol Metab. 2000;278:127–33.CrossRef Fantin VR, Wang Q, Lienhard GE, Keller SR, Valeria R. Mice lacking insulin receptor substrate 4 exhibit mild defects in growth, reproduction, and glucose homeostasis. Am J Physiol Endocrinol Metab. 2000;278:127–33.CrossRef
36.
Zurück zum Zitat Saad MJA, Araki E, Rothenberg PL, White MF, Kahn CR. Regulation of insulin receptor substrate-1 in liver and muscle of animal models of insulin resistance. J Clin Invest. 1992;90:1839–49.CrossRefPubMedPubMedCentral Saad MJA, Araki E, Rothenberg PL, White MF, Kahn CR. Regulation of insulin receptor substrate-1 in liver and muscle of animal models of insulin resistance. J Clin Invest. 1992;90:1839–49.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren J, Previs S, et al. Disruption of IRS-2 causes type 2 diabetes in mice. Nature. 1998;391:900–4.CrossRefPubMed Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren J, Previs S, et al. Disruption of IRS-2 causes type 2 diabetes in mice. Nature. 1998;391:900–4.CrossRefPubMed
38.
39.
Zurück zum Zitat Parsons A, Sorenson L. Dwarf mice: effect of lactogenic hormones * of. Endocr Soc. 2014;136(5):2013–22.CrossRef Parsons A, Sorenson L. Dwarf mice: effect of lactogenic hormones * of. Endocr Soc. 2014;136(5):2013–22.CrossRef
42.
Zurück zum Zitat Cheung NW, Byth K. Population health significance of gestational diabetes. Diabetes Care. 2005;26(7):2005–9.CrossRef Cheung NW, Byth K. Population health significance of gestational diabetes. Diabetes Care. 2005;26(7):2005–9.CrossRef
43.
Zurück zum Zitat Perkins JM, Dunn JP, Jagasia SM. Perspectives in gestational diabetes mellitus: a review of screening, diagnosis, and treatment. Clin Diabetes. 2007;25(2):57–62.CrossRef Perkins JM, Dunn JP, Jagasia SM. Perspectives in gestational diabetes mellitus: a review of screening, diagnosis, and treatment. Clin Diabetes. 2007;25(2):57–62.CrossRef
44.
Zurück zum Zitat Plagemann A, Dorner G. Alterations of hypothalamic catecholamines in the newborn offspring of gestational diabetic mother rats. Dev Brain Res. 1998;109:201–9.CrossRef Plagemann A, Dorner G. Alterations of hypothalamic catecholamines in the newborn offspring of gestational diabetic mother rats. Dev Brain Res. 1998;109:201–9.CrossRef
46.
Zurück zum Zitat Steculorum SM, Bouret SG. Maternal diabetes compromises the organization of hypothalamic feeding circuits and impairs leptin sensitivity in offspring. Endocrinology. 2015;152:4171–9.CrossRef Steculorum SM, Bouret SG. Maternal diabetes compromises the organization of hypothalamic feeding circuits and impairs leptin sensitivity in offspring. Endocrinology. 2015;152:4171–9.CrossRef
47.
Zurück zum Zitat Steculorum SM, Bouret SG. Maternal diabetes compromises the Organization of Hypothalamic Feeding Circuits and Impairs Leptin Sensitivity in offspring. Endocrinology. 2011;152(11):4171–9.CrossRefPubMedPubMedCentral Steculorum SM, Bouret SG. Maternal diabetes compromises the Organization of Hypothalamic Feeding Circuits and Impairs Leptin Sensitivity in offspring. Endocrinology. 2011;152(11):4171–9.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat De la Monte SM. Insulin resistance and neurodegeneration: progress towards the development of new therapeutics for Alzheimer’s disease. Drugs [Internet]. Springer International Publishing; 2017 [cited 2017 Feb 9];77(1):47–65. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27988872 . De la Monte SM. Insulin resistance and neurodegeneration: progress towards the development of new therapeutics for Alzheimer’s disease. Drugs [Internet]. Springer International Publishing; 2017 [cited 2017 Feb 9];77(1):47–65. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27988872 .
49.
Zurück zum Zitat Kinney BA, Rabe MB, Jensen RA, Steger RW. Maternal hyperglycemia leads to gender-dependent deficits in learning and memory in offspring. Exp Biol Med. 2003;228(2):152–9.CrossRef Kinney BA, Rabe MB, Jensen RA, Steger RW. Maternal hyperglycemia leads to gender-dependent deficits in learning and memory in offspring. Exp Biol Med. 2003;228(2):152–9.CrossRef
51.
52.
Zurück zum Zitat Van Lieshout RJ, Voruganti LP. Examen critique Diabetes mellitus during pregnancy and increased risk of schizophrenia in offspring: a review of the evidence and putative mechanisms. J Psychiatry Neurosci. 2008;33(5):395–404.PubMedPubMedCentral Van Lieshout RJ, Voruganti LP. Examen critique Diabetes mellitus during pregnancy and increased risk of schizophrenia in offspring: a review of the evidence and putative mechanisms. J Psychiatry Neurosci. 2008;33(5):395–404.PubMedPubMedCentral
53.
Zurück zum Zitat Bolaños L, Matute E, Ramírez-Dueñas MDL, Zarabozo D. Neuropsychological Impairment in School-Aged Children Born to Mothers With Gestational Diabetes. J Child Neurol [Internet]. 2015 [cited 2017 Feb 6];30(12):1616–24. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25814475 . Bolaños L, Matute E, Ramírez-Dueñas MDL, Zarabozo D. Neuropsychological Impairment in School-Aged Children Born to Mothers With Gestational Diabetes. J Child Neurol [Internet]. 2015 [cited 2017 Feb 6];30(12):1616–24. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25814475 .
56.
Zurück zum Zitat Haghir H, Hami J, Lotf N, Peyvandi M, Ghasemi S, Hosseini M. Expression of apoptosis-regulatory genes in the hippocampus of rat neonates born to mothers with diabetes. Metab Brain Dis. 2017:1–12. Haghir H, Hami J, Lotf N, Peyvandi M, Ghasemi S, Hosseini M. Expression of apoptosis-regulatory genes in the hippocampus of rat neonates born to mothers with diabetes. Metab Brain Dis. 2017:1–12.
58.
Zurück zum Zitat Wan Q, Xiong ZG, Man HY, Ackerley CA. Recruitment of functional GABA A receptors to postsynaptic domains by insulin. Nature. 1997;388(August):686–90.CrossRefPubMed Wan Q, Xiong ZG, Man HY, Ackerley CA. Recruitment of functional GABA A receptors to postsynaptic domains by insulin. Nature. 1997;388(August):686–90.CrossRefPubMed
59.
60.
Zurück zum Zitat Chiu S-L, Chen C-M, Cline HT. Insulin receptor signaling regulates synapse number, dendritic plasticity and circuit function in vivo. Neuron. 2011;58(5):708–19.CrossRef Chiu S-L, Chen C-M, Cline HT. Insulin receptor signaling regulates synapse number, dendritic plasticity and circuit function in vivo. Neuron. 2011;58(5):708–19.CrossRef
62.
Zurück zum Zitat Silverman BL, Metzger BE, Cho NH, Loeb CA. Impaired glucose tolerance in adolescent offspring of diabetic mothers. Diabetes Care. 1995;18(5):611–7.CrossRefPubMed Silverman BL, Metzger BE, Cho NH, Loeb CA. Impaired glucose tolerance in adolescent offspring of diabetic mothers. Diabetes Care. 1995;18(5):611–7.CrossRefPubMed
63.
Zurück zum Zitat Ross MG, Desai M, Khorram O, Mcknight RA, Lane RH, Torday J. Gestational programming of offspring obesity: a potential contributor to Alzheimer’s disease. Curr Alzheimer Reasearch. 2007;4:213–7.CrossRef Ross MG, Desai M, Khorram O, Mcknight RA, Lane RH, Torday J. Gestational programming of offspring obesity: a potential contributor to Alzheimer’s disease. Curr Alzheimer Reasearch. 2007;4:213–7.CrossRef
64.
Zurück zum Zitat Lourenco M V, Clarke JR, Frozza RL, Bomfim TR, Forny-Germano L, Batista AF, et al. TNF-α mediates PKR-dependent memory impairment and brain IRS-1 inhibition induced by Alzheimer’s β-amyloid oligomers in mice and monkeys. Cell Metab [Internet]. 2013 [cited 2015 Feb 4];18(6):831–43. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24315369 . Lourenco M V, Clarke JR, Frozza RL, Bomfim TR, Forny-Germano L, Batista AF, et al. TNF-α mediates PKR-dependent memory impairment and brain IRS-1 inhibition induced by Alzheimer’s β-amyloid oligomers in mice and monkeys. Cell Metab [Internet]. 2013 [cited 2015 Feb 4];18(6):831–43. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24315369 .
66.
Zurück zum Zitat Leloup C, Magnan C, Alquier T, Mistry S. Intrauterine hyperglycemia increases insulin binding sites but not glucose transporter expression in discrete brain areas in term rat fetuses. Pediatr Res. 2004;56(2):263–7.CrossRefPubMed Leloup C, Magnan C, Alquier T, Mistry S. Intrauterine hyperglycemia increases insulin binding sites but not glucose transporter expression in discrete brain areas in term rat fetuses. Pediatr Res. 2004;56(2):263–7.CrossRefPubMed
69.
Zurück zum Zitat Maurer MH, Geomor HK, Bürgers HF, Schelshorn DW, Kuschinsky W. Adult neural stem cells express glucose transporters GLUT1 and GLUT3 and regulate GLUT3 expression. FEBS Lett [Internet]. 2006 [cited 2017 Feb 9];580(18):4430–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16854415 . Maurer MH, Geomor HK, Bürgers HF, Schelshorn DW, Kuschinsky W. Adult neural stem cells express glucose transporters GLUT1 and GLUT3 and regulate GLUT3 expression. FEBS Lett [Internet]. 2006 [cited 2017 Feb 9];580(18):4430–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16854415 .
70.
Zurück zum Zitat Freeman L, Zitlin V, Stevens C, Granholm A-C. Diet induced effects on neuronal and glial elements in the middle-aged rat hippocampus. Nutr Neurosci. 2011;14(1):32–44.CrossRefPubMedPubMedCentral Freeman L, Zitlin V, Stevens C, Granholm A-C. Diet induced effects on neuronal and glial elements in the middle-aged rat hippocampus. Nutr Neurosci. 2011;14(1):32–44.CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat Harik N, Harik S, Kuo N-T, Sakai K, Przybylski R, LaManna J. Time-course and reversibility of the hypoxia-induced alterations in cerebral vascularity and cerebral capillary glucose transporter density. Brain Res. 1996;737:335–8.CrossRefPubMed Harik N, Harik S, Kuo N-T, Sakai K, Przybylski R, LaManna J. Time-course and reversibility of the hypoxia-induced alterations in cerebral vascularity and cerebral capillary glucose transporter density. Brain Res. 1996;737:335–8.CrossRefPubMed
72.
Zurück zum Zitat Eidelman AL, Samueloff A. The pathophysiology of the fetus of the diabetic mother. Semin Perinatol. 2002;26(3):232–6.CrossRefPubMed Eidelman AL, Samueloff A. The pathophysiology of the fetus of the diabetic mother. Semin Perinatol. 2002;26(3):232–6.CrossRefPubMed
74.
Zurück zum Zitat Mcquillen PS, Sheldon RA, Shatz CJ, Ferriero DM. Selective vulnerability of subplate neurons after early neonatal hypoxia-ischemia. J Neurosci. 2003;23(8):3308–15.CrossRefPubMedPubMedCentral Mcquillen PS, Sheldon RA, Shatz CJ, Ferriero DM. Selective vulnerability of subplate neurons after early neonatal hypoxia-ischemia. J Neurosci. 2003;23(8):3308–15.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Buzina-suboticanec K, Buzina R, Stavljenic A, Tadinac-babic M. Effects of iron supplementation on iron nutrition status and cognitive functions in children. Food Nutr Bull. 1998;19(4):298–306.CrossRef Buzina-suboticanec K, Buzina R, Stavljenic A, Tadinac-babic M. Effects of iron supplementation on iron nutrition status and cognitive functions in children. Food Nutr Bull. 1998;19(4):298–306.CrossRef
76.
Zurück zum Zitat Petry C, Eaton M, Wobken J, Mills MM, Johnson D, Georgieff M. Iron deficiency of liver, heart, and brain in newborn infants of diabetic mothers. J Pediatr. 1992;121:109–14.CrossRefPubMed Petry C, Eaton M, Wobken J, Mills MM, Johnson D, Georgieff M. Iron deficiency of liver, heart, and brain in newborn infants of diabetic mothers. J Pediatr. 1992;121:109–14.CrossRefPubMed
77.
Zurück zum Zitat Pritchard WS. Cognitive event-related potential correlates of schizophrenia. Psychol Bull. 1986;100(1):43–66.CrossRefPubMed Pritchard WS. Cognitive event-related potential correlates of schizophrenia. Psychol Bull. 1986;100(1):43–66.CrossRefPubMed
78.
Zurück zum Zitat Beard JL, Felt B, Schallert T, Burhans M, Connor JR, Georgieff MK. Moderate iron deficiency in infancy: biology and behavior in young rats. Behav Brain Res [Internet]. 2006 [cited 2017 Feb 5];170(2):224–32. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16569441 . Beard JL, Felt B, Schallert T, Burhans M, Connor JR, Georgieff MK. Moderate iron deficiency in infancy: biology and behavior in young rats. Behav Brain Res [Internet]. 2006 [cited 2017 Feb 5];170(2):224–32. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16569441 .
79.
Zurück zum Zitat Vaughn J, Brown J, Carter JP. The effects of maternal anemia on infant behavior. J Natl Med Assoc. 1986;78(10):963–8.PubMedPubMedCentral Vaughn J, Brown J, Carter JP. The effects of maternal anemia on infant behavior. J Natl Med Assoc. 1986;78(10):963–8.PubMedPubMedCentral
80.
Zurück zum Zitat Kinalski M, Telejko B, Kuz M, Kre A, Kinalska I. Tumor necrosis factor alpha system and plasma adiponectin concentration in women with gestational diabetes. Horm Metab Res. 2005;37:450–4.CrossRefPubMed Kinalski M, Telejko B, Kuz M, Kre A, Kinalska I. Tumor necrosis factor alpha system and plasma adiponectin concentration in women with gestational diabetes. Horm Metab Res. 2005;37:450–4.CrossRefPubMed
82.
Zurück zum Zitat Barker AJ, Ullian EM. New roles for astrocytes in developing synaptic circuits. Commun Integr Biol 2008;207–11, 1, 211. Barker AJ, Ullian EM. New roles for astrocytes in developing synaptic circuits. Commun Integr Biol 2008;207–11, 1, 211.
83.
Zurück zum Zitat Tomassoni D, Nwankwo IE, Gabrielli MG, Bhatt S, Muhammad AB, Lokhandwala MF, et al. Astrogliosis in the brain of obese Zucker rat: a model of metabolic syndrome. Neurosci Lett [Internet]. Elsevier Ireland Ltd. 2013;543:136–41. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23545209 Tomassoni D, Nwankwo IE, Gabrielli MG, Bhatt S, Muhammad AB, Lokhandwala MF, et al. Astrogliosis in the brain of obese Zucker rat: a model of metabolic syndrome. Neurosci Lett [Internet]. Elsevier Ireland Ltd. 2013;543:136–41. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23545209
84.
Zurück zum Zitat Gehrmann J, Matsumoto Y, Kreutzberg GW. REVIEWS Microglia: intrinsic immuneffector cell of the brain. Brain Res Rev. 1995;20:269–87.CrossRefPubMed Gehrmann J, Matsumoto Y, Kreutzberg GW. REVIEWS Microglia: intrinsic immuneffector cell of the brain. Brain Res Rev. 1995;20:269–87.CrossRefPubMed
86.
Zurück zum Zitat Fein A, Kostina E, Savion S, Orenstein H, Shepshelovich J, Torchinsky A, et al. Expression of tumor necrosis factor-a in the pregnant uterus of diabetic mice: effect of maternal immunopotentiation. Am J Reprod Immunol. 2001;46:161–8.CrossRef Fein A, Kostina E, Savion S, Orenstein H, Shepshelovich J, Torchinsky A, et al. Expression of tumor necrosis factor-a in the pregnant uterus of diabetic mice: effect of maternal immunopotentiation. Am J Reprod Immunol. 2001;46:161–8.CrossRef
87.
Zurück zum Zitat Vuong B, Odero G, Rozbacher S, Stevenson M, Kereliuk SM, Pereira TJ, et al. Exposure to gestational diabetes mellitus induces neuroinflammation, derangement of hippocampal neurons, and cognitive changes in rat offspring. J Neuroinflammation. J Neuroinflammation. 2017:1–13. Vuong B, Odero G, Rozbacher S, Stevenson M, Kereliuk SM, Pereira TJ, et al. Exposure to gestational diabetes mellitus induces neuroinflammation, derangement of hippocampal neurons, and cognitive changes in rat offspring. J Neuroinflammation. J Neuroinflammation. 2017:1–13.
88.
Zurück zum Zitat Cuffe JSM, Xu C, Anthony V. Biomarkers of oxidative stress in pregnancy complications. Biomark Med. 2017;11:295–306.CrossRefPubMed Cuffe JSM, Xu C, Anthony V. Biomarkers of oxidative stress in pregnancy complications. Biomark Med. 2017;11:295–306.CrossRefPubMed
89.
Zurück zum Zitat Toljic M, Egic A, Munjas J, Orlic NK, Milovanovic Z, Radenkovic A, et al. Increased oxidative stress and cytokinesis-block micronucleus cytome assay parameters in pregnant women with gestational diabetes mellitus and gestational arterial hypertension. Reprod Toxicol [Internet]. Elsevier Inc.; 2017; Available from https://doi.org/10.1016/j.reprotox.2017.04.002 Toljic M, Egic A, Munjas J, Orlic NK, Milovanovic Z, Radenkovic A, et al. Increased oxidative stress and cytokinesis-block micronucleus cytome assay parameters in pregnant women with gestational diabetes mellitus and gestational arterial hypertension. Reprod Toxicol [Internet]. Elsevier Inc.; 2017; Available from https://​doi.​org/​10.​1016/​j.​reprotox.​2017.​04.​002
90.
Zurück zum Zitat Usluoğullari B, Usluogullari CA, Balkan F, Orkmez M. Role of serum levels of irisin and oxidative stress markers in pregnant women with and without gestational diabetes Role of serum levels of irisin and oxidative stress markers in pregnant women with and without gestational diabetes. Gynecol Endocrinol [Internet]. Informa UK Limited, trading as Taylor 8 Francis Group; 2017;0(0):-000. Available from https://doi.org/10.1080/09513590.2017.1284789 Usluoğullari B, Usluogullari CA, Balkan F, Orkmez M. Role of serum levels of irisin and oxidative stress markers in pregnant women with and without gestational diabetes Role of serum levels of irisin and oxidative stress markers in pregnant women with and without gestational diabetes. Gynecol Endocrinol [Internet]. Informa UK Limited, trading as Taylor 8 Francis Group; 2017;0(0):-000. Available from https://​doi.​org/​10.​1080/​09513590.​2017.​1284789
91.
Zurück zum Zitat Li H, Yin Q, Li N, Ouyang Z, Zhong M. Plasma markers of oxidative stress in patients with gestational diabetes mellitus in the second and third trimester. Obstet Gynecol Int Hindawi Publishing Corporation. 2016;2016:1–8.CrossRef Li H, Yin Q, Li N, Ouyang Z, Zhong M. Plasma markers of oxidative stress in patients with gestational diabetes mellitus in the second and third trimester. Obstet Gynecol Int Hindawi Publishing Corporation. 2016;2016:1–8.CrossRef
93.
Zurück zum Zitat Aziz HSA, John CM, Yusof NIMS, Nordin M, Ramasamy R, Adam A, et al. Animal model of gestational diabetes mellitus with pathophysiological resemblance to the human condition induced by multiple factors ( nutritional, pharmacological, and stress ) in rats. Biomed Hindawi Publishing Corporation; 2016;2016. Aziz HSA, John CM, Yusof NIMS, Nordin M, Ramasamy R, Adam A, et al. Animal model of gestational diabetes mellitus with pathophysiological resemblance to the human condition induced by multiple factors ( nutritional, pharmacological, and stress ) in rats. Biomed Hindawi Publishing Corporation; 2016;2016.
94.
Zurück zum Zitat Davies KJA. Oxidative stress: the paradox of aerobic life. Buochem Soc Symp. 1995;61:1–31.CrossRef Davies KJA. Oxidative stress: the paradox of aerobic life. Buochem Soc Symp. 1995;61:1–31.CrossRef
95.
Zurück zum Zitat Stanner SA, Hughes J, Kelly CNM, Buttriss J. A review of the epidemiological evidence for the “antioxidant hypothesis.”. Public Health Nutr. 2017;7:407–22.CrossRef Stanner SA, Hughes J, Kelly CNM, Buttriss J. A review of the epidemiological evidence for the “antioxidant hypothesis.”. Public Health Nutr. 2017;7:407–22.CrossRef
96.
Zurück zum Zitat Sena E, Wheble P, Sandercock P, Macleod M. Systematic review and meta-analysis of the efficacy of tirilazad in experimental stroke. Stroke. 2007:388–95. Sena E, Wheble P, Sandercock P, Macleod M. Systematic review and meta-analysis of the efficacy of tirilazad in experimental stroke. Stroke. 2007:388–95.
97.
Zurück zum Zitat Bannister J, Bannister W, Rotilio G. Aspects of the structure, function, and applications of superoxide dismutase. Crit Rev Biomed. 1987;22:111–80. Bannister J, Bannister W, Rotilio G. Aspects of the structure, function, and applications of superoxide dismutase. Crit Rev Biomed. 1987;22:111–80.
98.
Zurück zum Zitat Chelikani P, Fita I, Loewen P. Diversity of structures and properties among catalases. Cell. 2009;61:192–208. Chelikani P, Fita I, Loewen P. Diversity of structures and properties among catalases. Cell. 2009;61:192–208.
99.
Zurück zum Zitat Sedighi O, Makhlough A, Shokrzadeh M, Hoorshad S. Association between plasma selenium and glutathione peroxidase levels and severity of diabetic nephropathy in patients with type two diabetes mellitus. Nephro Urol Mon. 2014;6(5):10–3.CrossRef Sedighi O, Makhlough A, Shokrzadeh M, Hoorshad S. Association between plasma selenium and glutathione peroxidase levels and severity of diabetic nephropathy in patients with type two diabetes mellitus. Nephro Urol Mon. 2014;6(5):10–3.CrossRef
100.
Zurück zum Zitat Renault KM, Carlsen EM, Nilas L, Secher NJ, Cortes D, Olsen SF, et al. Accepted article preview: published ahead of advance online publication. Int J Obes 2017; Renault KM, Carlsen EM, Nilas L, Secher NJ, Cortes D, Olsen SF, et al. Accepted article preview: published ahead of advance online publication. Int J Obes 2017;
101.
Zurück zum Zitat Inzucchi SE, Bergenstal RM, Buse JB, Diamant M, Ferrannini E, Nauck M, et al. Management of hyperglycaemia in type 2 diabetes: a patient-centered approach. Position statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia [Internet]. 2012 [cited 2014 Jul 12];55(6):1577–96. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22526604 . Inzucchi SE, Bergenstal RM, Buse JB, Diamant M, Ferrannini E, Nauck M, et al. Management of hyperglycaemia in type 2 diabetes: a patient-centered approach. Position statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia [Internet]. 2012 [cited 2014 Jul 12];55(6):1577–96. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22526604 .
Metadaten
Titel
Gestational diabetes is associated to the development of brain insulin resistance in the offspring
verfasst von
Ricardo Augusto Leoni De Sousa
Publikationsdatum
07.03.2018
Verlag
Springer India
Erschienen in
International Journal of Diabetes in Developing Countries / Ausgabe 2/2019
Print ISSN: 0973-3930
Elektronische ISSN: 1998-3832
DOI
https://doi.org/10.1007/s13410-018-0618-1

Weitere Artikel der Ausgabe 2/2019

International Journal of Diabetes in Developing Countries 2/2019 Zur Ausgabe