Acessibilidade / Reportar erro

Consequences of gestational diabetes to the brain and behavior of the offspring

ABSTRACT

Gestational diabetes mellitus (GD) is a form of insulin resistance triggered during the second/third trimesters of pregnancy in previously normoglycemic women. It is currently estimated that 10% of all pregnancies in the United States show this condition. For many years, the transient nature of GD has led researchers and physicians to assume that long-term consequences were absent. However, GD diagnosis leads to a six-fold increase in the risk of developing type 2 diabetes (T2D) in women and incidence of obesity and T2D is also higher among their infants. Recent and concerning evidences point to detrimental effects of GD on the behavior and cognition of the offspring, which often persist until adolescence or adulthood. Considering that the perinatal period is critical for determination of adult behavior, it is expected that the intra-uterine exposure to hyperglycemia, hyperinsulinemia and pro-inflammatory mediators, hallmark features of GD, might affect brain development. Here, we review early clinical and experimental evidence linking GD to consequences on the behavior of the offspring, focusing on memory and mood disorders. We also discuss initial evidence suggesting that downregulation of insulin signaling cascades are seen in the brains of GD offspring and could contribute to the consequences on their behavior.

Key words:
insulin resistance; hippocampus; inflammation; depression; learning; memory; programing

INTRODUCTION

Gestational diabetes (GD) is defined as a form of insulin resistance that initially manifests during the second or third trimesters of pregnancy in previously normoglycemic women. It is expected that GD occurs in up to 10% of all pregnancies, reaching higher incidence in developed countries, especially in the United States (DeSisto et al. 2014). Epidemiological data also suggest an alarming increase in the number of cases over the last few years (Albrecht et al. 2010ALBRECHT SS, KUKLINA EV, BANSIL P, JAMIESON DJ, WHITEMAN MK, KOURTIS AP, POSNER SF AND CALLAGHAN WM. 2010. Diabetes trends among delivery hospitalizations in the U.S., 1994-2004. Diabetes Care 33: 768-773.). Risk factors for GD include family history of overweight and obesity, nonwhite race and advanced maternal age (Cypryk et al. 2008CYPRYK K, SZYMCZAK W, CZUPRYNIAK L, SOBCZAK M AND LEWINSKI A. 2008. Gestational diabetes mellitus - an analysis of risk factors. Endokrynol Pol 59: 393-397., Savitz et al. 2008SAVITZ DA, JANEVIC TM, ENGEL SM, KAUFMAN JS AND HERRING AH. 2008. Ethnicity and gestational diabetes in New York City, 1995-2003. BJOG 115: 969-978.), but independent of risk factor any pregnant woman may manifest this metabolic change.

GD has been associated to macrosomia of the offspring and to sporadic reports of neonatal hypoglycemia, hypocalcemia and respiratory distress syndrome (Frías et al. 2007). Despite these observations, until recently GD was considered a transient condition associated with no major consequence to long-term health of the mother or child, since it is expected that only 3-5% of women remain diabetic after labor (Gilmartin et al. 2008GILMARTIN AB, URAL SH AND REPKE JT. 2008. Gestational diabetes mellitus. Rev Obstet Gynecol 1: 129-134.) and longitudinal studies following this population are still rare. Therefore, whereas extensive research has focused on unraveling the consequences of obesity and type 2 diabetes (T2D), the long-term effects of GD have been poorly scrutinized and are possibly underestimated (Poston 2011POSTON L. 2011. Intergenerational transmission of insulin resistance and type 2 diabetes. Prog Biophys Mol Biol 106: 315-322.). It is now known that women who developed insulin resistance during pregnancy have a six-fold increase in the risk of developing T2D later in life compared to women who remained euglycemic (Cheung and Byth 2003CHEUNG NW AND BYTH K. 2003. Population health significance of gestational diabetes. Diabetes Care 26: 2005-2009.). However, more recent and concerning evidence point to detrimental effects of GD on the development, metabolism and behavior of the offspring (Daraki et al. 2017DARAKI V, ROUMELIOTAKI T, KOUTRA K, GEORGIOU V, KAMPOURI M, KYRIKLAKI A, VAFEIADI M, PAPAVASILIOU S, KOGEVINAS M AND CHATZI L. 2017. Effect of parental obesity and gestational diabetes on child neuropsychological and behavioral development at 4 years of age: the Rhea mother-child cohort, Crete, Greece. Eur Child Adolesc Psychiatry 26: 703-714., Yessoufou and Moutairou 2011YESSOUFOU A AND MOUTAIROU K. 2011. Maternal diabetes in pregnancy: early and long-term outcomes on the offspring and the concept of “metabolic memory”. Exp Diabetes Res 2011: 218598., Garcia-Vargas et al. 2012).

Early studies by Dorner and Mohnike (1976DÖRNER G AND MOHNIKE A. 1976. Further evidence for a predominantly maternal transmission of maturity-onset type diabetes. Endokrinologie 68: 121-124.) suggested a higher incidence of diabetes in adults born to mothers with GD, which has been further supported by several other studies (Silverman et al. 1995SILVERMAN BL, METZGER BE, CHO NH AND LOEB CA. 1995. Impaired glucose tolerance in adolescent offspring of diabetic mothers. Relationship to fetal hyperinsulinism. Diabetes Care 18: 611-617., Poston and Health 2010POSTON L. 2010. Developmental programming and diabetes - The human experience and insight from animal models. Best Pract Res Clin Endocrinol Metab 24: 541-552.). Animal and human studies suggest that the exaggerated glucose transportation across the placenta is the main responsible for fetal hyperglycemia, pancreatic hyperplasia and enhanced insulin secretion, which might generate life-long persistent effects on pancreatic secretory function (Plagemann et al. 1998PLAGEMANN A, HARDER T, LINDNER R, MELCHIOR K, RAKE A, RITTEL F, ROHDE W AND DORNER G. 1998. Alterations of hypothalamic catecholamines in the newborn offspring of gestational diabetic mother rats. Brain Res Dev Brain Res 109: 201-209.). Increased hypothalamic inflammation and disrupted insulin signaling in this brain region are classical mechanisms involved in the physiopathology of T2D, and recent studies using animal models have showed that similar alterations occur in the hypothalamus of GD offspring even before T2D manifests (Melo et al. 2014MELO AM, BENATTI RO, IGNACIO-SOUZA LM, OKINO C, TORSONI AS, MILANSKI M, VELLOSO LA AND TORSONI MA. 2014. Hypothalamic endoplasmic reticulum stress and insulin resistance in offspring of mice dams fed high-fat diet during pregnancy and lactation. Metabolism 63: 682-692., Steculorum and Bouret 2011STECULORUM SM AND BOURET SG. 2011. Maternal diabetes compromises the organization of hypothalamic feeding circuits and impairs leptin sensitivity in offspring. Endocrinology 152: 4171-4179.).

Considering that the perinatal period is critical for determination of adult behavior, including levels of anxiety, impulsivity and stress responses (Bolton and Bilbo 2014BOLTON JL AND BILBO SD. 2014. Developmental programming of brain and behavior by perinatal diet: focus on inflammatory mechanisms. Dialogues Clin Neurosci 16: 307-320.), it is expected that the intra-uterine exposure to hyperglicemia, hyperinsulinemia and proinflammatory mediators, hallmark features of GD, might have other consequences to both brain function and behavior. Animal models have been extremely useful in providing insights into this question. However, since GD is a transient and multifactorial condition, animal models that recapitulate all aspects of the disease remain challenging (Pasek and Gannon 2013PASEK RC AND GANNON M. 2013. Advancements and challenges in generating accurate animal models of gestational diabetes mellitus. Am J Physiol Endocrinol Metab 305: E1327-1338.).

Here, we review emerging clinical and experimental evidence linking GD to late consequences to the behavior of the offspring, especially concerning memory formation and mood disorders. Although preliminary, these early studies point out to GD as an important factor influencing offspring brain health. Moreover, studies from our group and others have recently described how inflammation and disrupted insulin signaling in memory-related brain regions occur in conditions that affect cognition (Bomfim et al. 2012BOMFIM TR ET AL. 2012. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease- associated Abeta oligomers. J Clin Invest 122: 1339-1353., Lourenço et al. 2013, Neves et al. 2016NEVES FS ET AL. 2016. Brain-Defective Insulin Signaling Is Associated to Late Cognitive Impairment in Post-Septic Mice. Mol Neurobiol.). Therefore, we also focus on the evidence suggesting that downregulation of insulin receptors and its intracellular cascade are seen in the brains of GD offspring and could contribute to affect their behavior.

ANIMAL MODELS OF GD

Currently available animal models of GD rely on surgical, chemical, nutritional or genetic approaches. For over a century, partial pancreatectomy performed before or during various stages of pregnancy has been described as an efficient method to surgically induce GD in different species, including rodents and dogs (Carlson and Drennan 1911CARLSON A AND DRENNAN F. 1911. The control of pancreatic diabetes in pregnancy by the passage of the internal secretion of the pancreas of the fetus to the blood of the mother. Am J Physiol 28: 391-395., Markowitz and Soskin 1927MARKOWITZ J AND SOSKIN S. 1927. Pancreatic diabetes and pregnancy. Am J Physiol 79: 553-558., Cuthbert et al. 1936CUTHBERT FP, IVY AC, ISAACS BL AND GRAY J. 1936. The relation of pregnancy and lactation to extirpation diabetis in the dog. Am J Physiol 115: 480-496., Jawerbaum et al. 1993JAWERBAUM A, CATAFAU JR, GONZALES ET, RODRIGUEZ RR, GELPI E, GOMEZ G, GIMENO AL AND GIMENO MA. 1993. Eicosanoid production by uterine strips and by embryos obtained from diabetic pregnant rats. Prostaglandins 45: 487-495.). Alternatively, models involving permanent damage to pancreatic β-cells can be induced through the administration of chemicals such as the nitrosurea derivative streptozotocin (STZ) and alloxan, a pyrimidine derivative (Junod et al. 1969JUNOD A, LAMBERT AE, STAUFFACHER W AND RENOLD AE. 1969. Diabetogenic action of streptozotocin: relationship of dose to metabolic response. J Clin Invest 48: 2129-2139., Lenzen and Panten 1988LENZEN S AND PANTEN U. 1988. Alloxan: history and mechanism of action. Diabetologia 31: 337-342.). Like pancreatectomy, these drugs induce an irreversible state of diabetes in experimental animals due to the drastic reduction of endogenous insulin, giving rise to a condition more closely related to T1D. As a consequence, such models provide limited information on the pathogenesis of GD, although they are useful to characterize the impact of hyperglycemia on the offspring.

As obesity is considered one of the main risk factors for GD, administration of high-fat (HFD), or glucose infusion to pregnant animals have been widely used as experimental models (Bihoreau et al. 1986BIHOREAU MT, KTORZA A, KINEBANYAN MF AND PICON L. 1986. Impaired glucose homeostasis in adult rats from hyperglycemic mothers. Diabetes 35: 979-984., Taylor et al. 2005TAYLOR PD ET AL. 2005. Impaired glucose homeostasis and mitochondrial abnormalities in offspring of rats fed a fat-rich diet in pregnancy. Am J Physiol Regul Integr Comp Physiol 288: R134-139., Srinivasan et al. 2006SRINIVASAN M, KATEWA SD, PALANIYAPPAN A, PANDYA JD AND PATEL MS. 2006. Maternal high-fat diet consumption results in fetal malprogramming predisposing to the onset of metabolic syndrome-like phenotype in adulthood. Am J Physiol Endocrinol Metab 291: E792-799.). Likewise, hyperglycemia and insulin resistance are hallmarks of a number of genetic models used for the study of metabolic diseases. Transgenic mice that do not express leptin or leptin receptors are characterized by an inability to adequately suppress feeding behavior and are classically used to model obesity and T2D. While homozygote knockouts for leptin or its receptor are infertile, heterozygous mice are glucose intolerant and develop GD (Lambin et al. 2007LAMBIN S, VAN BREE R, CALUWAERTS S, VERCRUYSSE L, VERGOTE I AND VERHAEGHE J. 2007. Adipose tissue in offspring of Lepr(db/+) mice: early-life environment vs. genotype. Am J Physiol Endocrinol Metab 292: E262-271.). It is important to mention that, although obese women have an increased risk for developing GD, only about 20% of all GD cases are attributable to obesity (Ferrara 2007FERRARA A. 2007. Increasing prevalence of gestational diabetes mellitus: a public health perspective. Diabetes Care 30(Suppl 2): S141-146., Kim et al. 2012aKIM SY, ENGLAND L, SAPPENFIELD W, WILSON HG, BISH CL, SALIHU HM AND SHARMA AJ. 2012a. Racial/ethnic differences in the percentage of gestational diabetes mellitus cases attributable to overweight and obesity, Florida, 2004-2007. Prev Chronic Dis 9: E88.). Many women develop GD despite being lean, meaning that factors other than increased body mass have an important role in the pathogenesis of the disease, and obesity-related models have limited construct validity. Moreover, HFD alters serum fatty acid profile (Liu et al. 2015LIU TW, HEDEN TD, MATTHEW MORRIS E, FRITSCHE KL, VIEIRA-POTTER VJ AND THYFAULT JP. 2015. High-Fat Diet Alters Serum Fatty Acid Profiles in Obesity Prone Rats: Implications for In Vitro Studies. Lipids 50: 997-1008.), possibly resulting in consequences on the offspring which are not necessarily related to GD.

Pregnancy is associated to a physiological decrease in insulin sensitivity in peripheral tissues, which are adaptive to allow increased access of the fetus to mother’s circulating glucose. Several studies indicate that pancreatic β-cell adaptations to pregnancy are crucial to maintain normoglycemia. Such adaptations include β-cell hypertrophy and proliferation, as well as increased insulin production and secretion (Baeyens et al. 2016BAEYENS L, HINDI S, SORENSON RL AND GERMAN MS. 2016. Beta-Cell adaptation in pregnancy. Diabetes Obes Metab 18(Suppl 1): 63-70.). Failure on this physiological pancreatic adaptation or an abnormally increased peripheral insulin resistance may contribute to generate GD. A number of GD mouse models are based on genetic manipulation of factors involved in β-cell adaptation during pregnancy, including prolactin receptor (PrlR) (Lee et al. 2009LEE CC, KUO YM, HUANG CC AND HSU KS. 2009. Insulin rescues amyloid beta-induced impairment of hippocampal long-term potentiation. Neurobiol Aging 30: 377-387.), c-Met, a tyrosine kinase receptor activated by hepatocyte growth factor (HGF) (Demirci et al. 2012DEMIRCI C, ERNST S, ALVAREZ-PEREZ JC, ROSA T, VALLE S, SHRIDHAR V, CASINELLI GP, ALONSO LC, VASAVADA RC AND GARCIA-OCANA A. 2012. Loss of HGF/c-Met signaling in pancreatic beta-cells leads to incomplete maternal beta-cell adaptation and gestational diabetes mellitus. Diabetes 61: 1143-1152.), the serotonin receptor 5Htr2b (Kim et al. 2010KIM H ET AL. 2010. Serotonin regulates pancreatic beta cell mass during pregnancy. Nat Med 16: 804-808.), and the nuclear factors menin (Karnik et al. 2007KARNIK SK, CHEN H, MCLEAN GW, HEIT JJ, GU X, ZHANG AY, FONTAINE M, YEN MH AND KIM SK. 2007. Menin controls growth of pancreatic beta-cells in pregnant mice and promotes gestational diabetes mellitus. Science 318: 806-809.), hepatocyte nuclear factor 4α (HNF-4α) (Gupta et al. 2007GUPTA RK, GAO N, GORSKI RK, WHITE P, HARDY OT, RAFIQ K, BRESTELLI JE, CHEN G, STOECKERT CJ JR AND KAESTNER KH. 2007. Expansion of adult beta-cell mass in response to increased metabolic demand is dependent on HNF-4alpha. Genes Dev 21: 756-769.), Forkhead box D3 (FoxD3) (Plank et al. 2011PLANK JL, FRIST AY, LEGRONE AW, MAGNUSON MA AND LABOSKY PA. 2011. Loss of Foxd3 results in decreased beta-cell proliferation and glucose intolerance during pregnancy. Endocrinology 152: 4589-4600.), and FoxM1 (Zhang et al. 2010ZHANG TY AND MEANEY MJ. 2010. Epigenetics and the environmental regulation of the genome and its function. Annu Rev Psychol 61: 439-466, C431-433.). Although these transgenic models may be useful in elucidating how GD impacts the offspring, they target specific signaling pathways, in contrast to the human GD which is polygenic and multifactorial in nature.

The generation of animal models that fully recapitulate GD is challenging, especially as a variety of risk factors, including ethnicity, weight, and family history can contribute to the development of the disease. Also, the severity of hyperglycemia found in some models is not typical of human GD. The main advantages and disadvantages of currently used animal models of GD are summarized in Table I. An ideal animal model would involve normoglycemic females at early pregnancy developing mild hyperglycemia during pregnancy, and returning to normal glucose levels shortly after labor. Another drawback is the fact that, once diagnosed, the condition is treated in humans, suggesting that normalization of glycemia in experimental GD could better reflect the influence of the disease in the offspring.

TABLE I
Advantages and disadvantages of currently available animal models of gestational diabetes.

METABOLIC AND BEHAVIORAL CONSEQUENCES OF BRAIN INSULIN SIGNALING DYSFUNCTION

Historically, the skeletal muscle, adipose tissue and liver were considered the main insulin-responsive tissues in control of peripheral metabolism. T2D was classically associated with impaired sensitivity to insulin in these tissues, decreasing glucose uptake and leading to hyperglycemia, even when insulin production and release were normal (Hotamisligil 2008HOTAMISLIGIL GS. 2008. Inflammation and endoplasmic reticulum stress in obesity and diabetes. Int J Obes (Lond) 32(Suppl 7): S52-54.). The brain was considered an insulin-insensitive organ until the late 1970’s, when it was demonstrated that i.c.v. infusion of insulin decreased food intake in experimental models (Woods et al. 1979WOODS SC, LOTTER EC, MCKAY LD AND PORTE D JR. 1979. Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature 282: 503-505.). After this landmark finding, the role of insulin signaling in brain regions that control peripheral metabolism have been extensively scrutinized. The hypothalamus is recognized as a key structure in control of whole body energy homeostasis in response to insulin and other hormones, and it is now known that hypothalamic insulin resistance plays a central role in obesity and T2D (Arruda et al. 2011ARRUDA AP, MILANSKI M, COOPE A, TORSONI AS, ROPELLE E, CARVALHO DP, CARVALHEIRA JB AND VELLOSO LA. 2011. Low-grade hypothalamic inflammation leads to defective thermogenesis, insulin resistance, and impaired insulin secretion. Endocrinology 152: 1314-1326., Thaler et al. 2012THALER JP ET AL. 2012. Obesity is associated with hypothalamic injury in rodents and humans. J Clin Invest 122: 153-162.).

Under physiological conditions, binding of insulin to its receptor triggers its intracellular tyrosine kinase activity. Insulin receptor substrate (IRS) proteins are a family of high molecular weight proteins, of which IRS-1 and IRS-2 are the most extensively studied. IRS-1 is targeted by insulin receptors and undergoes phosphorylation at tyrosine residues, a process recognized as the key initial step of the insulin signaling pathway, which is followed by PI3K activation and Akt phosphorylation. During T2D development, increased levels of pro-inflammatory mediators, especially TNF-α, act on the hypothalamus and peripheral tissues causing activation of intracellular stress kinases which can also target IRS-1, although they phosphorylate serine instead of tyrosine residues (Weissmann et al. 2014WEISSMANN L ET AL. 2014. IKKepsilon is key to induction of insulin resistance in the hypothalamus, and its inhibition reverses obesity. Diabetes 63: 3334-3345., Belgardt et al. 2010BELGARDT BF ET AL. 2010. Hypothalamic and pituitary c-Jun N-terminal kinase 1 signaling coordinately regulates glucose metabolism. Proc Natl Acad Sci U S A 107: 6028-6033.). Phosphorylation of IRS-1 at serine residues inhibits its phosphorylation at tyrosine residues and thus interferes with its ability to engage in insulin signaling even in the presence of insulin (Copps and White 2012COPPS KD AND WHITE MF. 2012. Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2. Diabetologia 55: 2565-2582.).

Insulin and insulin-like growth factor (IGF) receptors in the central nervous system, however, are not restricted to the hypothalamus, being widely distributed throughout the encephalon (Zhao et al. 2004ZHAO WQ, CHEN H, QUON MJ AND ALKON DL. 2004. Insulin and the insulin receptor in experimental models of learning and memory. Eur J Pharmacol 490: 71-81.). The hippocampus and cortex have extensive expression of theses receptors and are centrally involved in memory formation (Zhao and Alkon 2001). By acting on these brain regions, insulin was shown to be neuroprotective (Plum et al. 2005PLUM L, SCHUBERT M AND BRUNING JC. 2005. The role of insulin receptor signaling in the brain. Trends Endocrinol Metab 16: 59-65., Kovacs and Hajnal 2009KOVACS P AND HAJNAL A. 2009. In vivo electrophysiological effects of insulin in the rat brain. Neuropeptides 43: 283-293., Ott et al. 2012OTT V, BENEDICT C, SCHULTES B, BORN J AND HALLSCHMID M. 2012. Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism. Diabetes Obes Metab 14: 214-221., Bomfim et al. 2012BOMFIM TR ET AL. 2012. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease- associated Abeta oligomers. J Clin Invest 122: 1339-1353.) and to affect synapse plasticity (Wan et al. 1997WAN Q, XIONG ZG, MAN HY, ACKERLEY CA, BRAUNTON J, LU WY, BECKER LE, MACDONALD JF AND WANG YT. 1997. Recruitment of functional GABA(A) receptors to postsynaptic domains by insulin. Nature 388: 686-690., Biessels et al. 1996BIESSELS GJ, KAMAL A, RAMAKERS GM, URBAN IJ, SPRUIJT BM, ERKELENS DW AND GISPEN WH. 1996. Place learning and hippocampal synaptic plasticity in streptozotocin-induced diabetic rats. Diabetes 45: 1259-1266.) and cognitive function in healthy subjects (Ott et al. 2012, Benedict et al. 2004BENEDICT C, HALLSCHMID M, HATKE A, SCHULTES B, FEHM HL, BORN J AND KERN W. 2004. Intranasal insulin improves memory in humans. Psychoneuroendocrinology 29: 1326-1334.). Interestingly, growing evidence support that defective hippocampal insulin signaling is related to conditions that affect memory processing, particularly Alzheimer’s disease (AD) (Bomfim et al. 2012, Craft and Watson 2004CRAFT S AND WATSON GS. 2004. Insulin and neurodegenerative disease: shared and specific mechanisms. Lancet Neurol 3: 169-178., Ma et al. 2009MA QL ET AL. 2009. Beta-amyloid oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling: suppression by omega-3 fatty acids and curcumin. J Neurosci 29: 9078-9089.). TNF-α levels are elevated in the brains of AD patients and transgenic mouse models (Takeda et al. 2010TAKEDA S, SATO N, UCHIO-YAMADA K, SAWADA K, KUNIEDA T, TAKEUCHI D, KURINAMI H, SHINOHARA M, RAKUGI H AND MORISHITA R. 2010. Diabetes-accelerated memory dysfunction via cerebrovascular inflammation and Abeta deposition in an Alzheimer mouse model with diabetes. Proc Natl Acad Sci U S A 107: 7036-7041., Salkovic-Petrisic and Hoyer 2007). As in the hypothalamus of T2D patients, hippocampal activation of stress kinases (JNK, IKK and PKR) is also reported in response to increased levels of TNF-α in AD models (Lourenço et al. 2013, Forny-Germano et al. 2014, Ma et al. 2009). As a consequence, IRS-1 serine phosphorylation is increased and insulin signaling is impaired in the hippocampus, contributing to memory impairment in mouse models of sporadic and familial forms of AD (Bomfim et al. 2012). Our group has recently investigated whether similar molecular mechanisms also underlie cognitive impairment seen in sepsis survivors. Sepsis is accompanied by alterations in circulating glucose levels in acute stages and insulin administration was shown to increase survival rates (Gearhart and Parbhoo 2006GEARHART MM AND PARBHOO SK. 2006. Hyperglycemia in the critically ill patient. AACN Clin Issues 17: 50-55.). These patients often present late cognitive impairment and some of them never fully recover (Iwashyna et al. 2010IWASHYNA TJ, ELY EW, SMITH DM AND LANGA KM. 2010. Long-term cognitive impairment and functional disability among survivors of severe sepsis. JAMA 304: 1787-1794., Pandharipande et al. 2013PANDHARIPANDE PP ET AL. 2013. Long-term cognitive impairment after critical illness. N Engl J Med 369: 1306-1316., Semmler et al. 2012). Using an experimental model of sepsis, we were able to mimic the late cognitive impairment seen in patients and found that increased hippocampal expression of TNF-α and impaired insulin signaling in this brain region also accompany sepsis-associated late cognitive decline (Neves et al. 2016NEVES FS ET AL. 2016. Brain-Defective Insulin Signaling Is Associated to Late Cognitive Impairment in Post-Septic Mice. Mol Neurobiol.). Whether impaired brain insulin signaling is a common denominator of other conditions affecting memory remains to be established.

Even though GD represents a self-limited metabolic condition for the mother, factors such as the high permeability of placental barrier and the maternal pro-inflammatory and hyperglycemic status can be extremely deleterious to the fetus brain. Women with GD have increased plasma levels of inflammation markers, such as C-reactive protein, malondialdehyde (MDA) (Badehnoosh et al. 2017BADEHNOOSH B, KARAMALI M, ZARRATI M, JAMILIAN M, BAHMANI F, TAJABADI-EBRAHIMI M, JAFARI P, RAHMANI E AND ASEMI Z. 2017. The effects of probiotic supplementation on biomarkers of inflammation, oxidative stress and pregnancy outcomes in gestational diabetes. J Matern Fetal Neonatal Med: 1-9.), TNF-α (Friedman et al. 2008FRIEDMAN JE, KIRWAN JP, JING M, PRESLEY L AND CATALANO PM. 2008. Increased skeletal muscle tumor necrosis factor-alpha and impaired insulin signaling persist in obese women with gestational diabetes mellitus 1 year postpartum. Diabetes 57: 606-613.) among others (Lowe et al. 2010LOWE LP, METZGER BE, LOWE WL JR, DYER AR, MCDADE TW, MCINTYRE HD AND GROUP HSCR. 2010. Inflammatory mediators and glucose in pregnancy: results from a subset of the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study. J Clin Endocrinol Metab 95: 5427-5434.). Increased expression of the transcription factor peroxisome proliferator-activated receptor γ (PPARγ) has been described in leukocytes from GD patients compared to healthy pregnant women (Wójcik et al. 2015). Importantly, one study has shown that maternal overweight, but not exposure to intra-uterine hyperglycemia, was associated with increased plasma levels of IL-6 and C-reactive protein in 18-27-year-old offspring from GD mothers (Kelstrup et al. 2012KELSTRUP L, CLAUSEN TD, MATHIESEN ER, HANSEN T AND DAMM P. 2012. Low-grade inflammation in young adults exposed to intrauterine hyperglycemia. Diabetes Res Clin Pract 97: 322-330.). Animal studies have shown that both placenta and brains of GD fetuses have increased levels of pro-inflammatory markers (Tang et al. 2015TANG X, QIN Q, XIE X AND HE P. 2015. Protective effect of sRAGE on fetal development in pregnant rats with gestational diabetes mellitus. Cell Biochem Biophys 71: 549-556., Melo et al. 2014MELO AM, BENATTI RO, IGNACIO-SOUZA LM, OKINO C, TORSONI AS, MILANSKI M, VELLOSO LA AND TORSONI MA. 2014. Hypothalamic endoplasmic reticulum stress and insulin resistance in offspring of mice dams fed high-fat diet during pregnancy and lactation. Metabolism 63: 682-692.). In the hypothalamus, the pro-inflammatory profile appears to be long-lasting, since high expression of IL1-β mRNA and increased protein levels of NFκB/p-JNK were described in the hypothalamus of adult mice born from high-fat diet-fed mothers (Melo et al. 2014). Levels of endoplasmic reticulum stress markers were also higher in the brains of these adolescent animals delivered by GD females, suggesting that obesity-induced insulin resistance during pregnancy is associated to persistent changes in physiological protein synthesis (Melo et al. 2014). In addition, leptin resistance and reduced neural projections within hypothalamic nuclei of adult mice born from hyperglycemic dams were also reported (Steculorum and Bouret 2011STECULORUM SM AND BOURET SG. 2011. Maternal diabetes compromises the organization of hypothalamic feeding circuits and impairs leptin sensitivity in offspring. Endocrinology 152: 4171-4179.). These findings suggest that hypothalamic effects of GD in the offspring might be a consequence of inflammation or disrupted central insulin response. In this scenario, it seems plausible that the insulin signaling pathway in brain regions involved in learning and memory might also be affected, and this hypothesis has never been directly addressed.

EMERGING EVIDENCE OF LONG-LASTING INFLUENCE OF GD TO BRAIN AND BEHAVIOR OF THE OFFSPRING

MEMORY AND BRAIN INSULIN SIGNALING

Epidemiological, clinical and experimental studies support that GD can interfere with intra-uterine brain development and influence behavior later in life. Impaired performance in explicit memory tasks have been reported in 1-year-old babies (Deboer et al. 2005DEBOER T, WEWERKA S, BAUER PJ, GEORGIEFF MK AND NELSON CA. 2005. Explicit memory performance in infants of diabetic mothers at 1 year of age. Dev Med Child Neurol 47: 525-531., Riggins et al. 2010RIGGINS T, BAUER PJ, GEORGIEFF MK AND NELSON CA. 2010. Declarative memory performance in infants of diabetic mothers. Adv Child Dev Behav 38: 73-110.), whereas a slower development of cognition and language was found in 18-month-old babies from obese and diabetic mothers compared to the offspring of healthy subjects (Torres-Espinola et al. 2015). These cognitive deficits appear to be reversible, as older children from diabetic mothers have normal performance in different memory tasks (Riggins et al. 2010). These findings are suggestive of delayed neurocognitive development in the offspring of GD mothers.

Incipient studies have suggested that brain insulin signaling is involved in neurological deficits of offspring from diabetic mothers. One interesting study recorded fetal brain activity triggered by glucose ingestion in healthy or GD pregnant subjects, and associated diabetes to a slower brain response of the offspring (Linder et al. 2015LINDER K, SCHLEGER F, KIEFER-SCHMIDT I, FRITSCHE L, KUMMEL S, HENI M, WEISS M, HARING HU, PREISSL H AND FRITSCHE A. 2015. Gestational Diabetes Impairs Human Fetal Postprandial Brain Activity. J Clin Endocrinol Metab 100: 4029-4036.). Using a STZ rat model, Jing et al. (2014JING YH, SONG YF, YAO YM, YIN J, WANG DG AND GAO LP. 2014. Retardation of fetal dendritic development induced by gestational hyperglycemia is associated with brain insulin/IGF-I signals. Int J Dev Neurosci 37: 15-20.) found a decreased expression of IGF-1 and increased expression of insulin receptors in brains of E14, E16 and E18 fetuses from diabetic mothers, effects that were accompanied by reduced number of dendritic spines and smaller levels of the pre-synaptic protein synaptophysin. Authors found that these alterations were absent in fetuses from rats treated with insulin throughout pregnancy, suggesting that they are directly linked to hyperglycemia. Another study evaluated the expression of IGF-1 and insulin receptors specifically in the hippocampus of pups from STZ-treated pregnant rats. Authors found that IGF-1 receptor expression was decreased in the hippocampi of P7 and P14 male rats born from STZ-treated dams, whereas hippocampal insulin receptor expression was slightly increased at P0, but significantly reduced in P14 rats born from diabetic dams compared to control groups (Hami et al. 2013HAMI J, SADR-NABAVI A, SANKIAN M, BALALI-MOOD M AND HAGHIR H. 2013. The effects of maternal diabetes on expression of insulin-like growth factor-1 and insulin receptors in male developing rat hippocampus. Brain Struct Funct 218: 73-84.). Structural and electrophysiological alterations have also been described in the hippocampus of rodents born from GD mothers. A decreased number of neurons was found in the pyramidal layers of CA1 and CA3 hippocampal regions at postnatal days 7 and 21, in the offspring of STZ-treated rats (Golalipour et al. 2012GOLALIPOUR MJ, KAFSHGIRI SK AND GHAFARI S. 2012. Gestational diabetes induced neuronal loss in CA1 and CA3 subfields of rat hippocampus in early postnatal life. Folia Morphol (Warsz) 71: 71-77.). Chandna et al. (2015CHANDNA AR, KUHLMANN N, BRYCE CA, GREBA Q, CAMPANUCCI VA AND HOWLAND JG. 2015. Chronic maternal hyperglycemia induced during mid-pregnancy in rats increases RAGE expression, augments hippocampal excitability, and alters behavior of the offspring. Neuroscience 303: 241-260.) reported that hippocampal neurons in newborn pups from STZ-treated females showed altered action potential kinetics along with a more hyperpolarized resting membrane potential. Despite these changes in neonatal hippocampal excitability, animals in this study showed normal memory acquisition as adults.

MOOD DISORDERS

Classical studies in developmental psychobiology and physiology have shown how variations in perinatal environment are associated with changes in behavior that persist throughout life. Increased impulsivity, anxiety levels and depressive-like behavior, among other emotional behaviors, have been described as a consequence of the exposure to different stressful environments during development (Zhang and Meaney 2010ZHANG TY AND MEANEY MJ. 2010. Epigenetics and the environmental regulation of the genome and its function. Annu Rev Psychol 61: 439-466, C431-433.). Studies which directly investigated whether intra-uterine exposure to hyperglycemia and hyperinsulinemia were associated to altered anxiety levels showed confounding results. While some of them report no differences in anxiety levels among rats born from GD dams when diabetes was induced before pregnancy (Kinney et al. 2003KINNEY BA, RABE MB, JENSEN RA AND STEGER RW. 2003. Maternal hyperglycemia leads to gender-dependent deficits in learning and memory in offspring. Exp Biol Med (Maywood) 228: 152-159., Ramanathan et al. 2000RAMANATHAN M, JAISWAL AK AND BHATTACHARYA SK. 2000. Hyperglycaemia in pregnancy: effects on the offspring behaviour with special reference to anxiety paradigms. Indian J Exp Biol 38: 231-236.), one study reported decreased anxiety levels in the offspring when STZ was administered during mid pregnancy (Chandna et al. 2015CHANDNA AR, KUHLMANN N, BRYCE CA, GREBA Q, CAMPANUCCI VA AND HOWLAND JG. 2015. Chronic maternal hyperglycemia induced during mid-pregnancy in rats increases RAGE expression, augments hippocampal excitability, and alters behavior of the offspring. Neuroscience 303: 241-260.), a condition which more closely resembles the time course of GD development in humans.

Mood-relevant neurotransmitter systems in the fetus brain may also be affected by GD, since changes in cathecolamine system were observed in several hypothalamic nuclei of newborn and adolescent offspring from GD rats (Plagemann et al. 1998PLAGEMANN A, HARDER T, LINDNER R, MELCHIOR K, RAKE A, RITTEL F, ROHDE W AND DORNER G. 1998. Alterations of hypothalamic catecholamines in the newborn offspring of gestational diabetic mother rats. Brain Res Dev Brain Res 109: 201-209.). Until now, there are no studies directly evaluating whether animals or patients born from diabetic mothers show increased depressive-like behavior or whether they are more susceptible to becoming depressive following a second hit later in life, as previously described for other conditions.

NEUROPSYCHIATRIC DISORDERS

Early-life exposure to several common viruses and bacteria has been linked to the development of neuropsychiatric disorders. Manipulation of maternal immune system appears to be a common denominator important for determination of later outcomes (Estes and MacAllister 2016ESTES ML AND MCALLISTER AK. 2016. Maternal immune activation: Implications for neuropsychiatric disorders. Science 353: 772-777.). In fact, a broader range of environmental distresses during the prenatal period have been associated to the development of schizophrenia and autism later in life (Reisinger et al. 2015REISINGER S, KHAN D, KONG E, BERGER A, POLLAK A AND POLLAK DD. 2015. The poly(I:C)-induced maternal immune activation model in preclinical neuropsychiatric drug discovery. Pharmacol Ther 149: 213-226.). Clinical evidence suggest that the offspring of GD mothers have increased risk of developing schizophrenia (Van Lieshout and Voruganti 2008, Boksa 2004BOKSA P. 2004. Animal models of obstetric complications in relation to schizophrenia. Brain Res Brain Res Rev 45: 1-17.) and autism (Gardener et al. 2009GARDENER H, SPIEGELMAN D AND BUKA SL. 2009. Prenatal risk factors for autism: comprehensive meta-analysis. Br J Psychiatry 195: 7-14., Xiang et al. 2015XIANG AH, WANG X, MARTINEZ MP, WALTHALL JC, CURRY ES, PAGE K, BUCHANAN TA, COLEMAN KJ AND GETAHUN D. 2015. Association of maternal diabetes with autism in offspring. JAMA 313: 1425-1434.) during adolescence and adulthood. Interestingly, a schizophrenic-like phenotype was successfully reproduced in male rats born from STZ-treated dams, as these animals showed disruption of pre-pulse inhibition response in their young adulthood (Chandna et al. 2015CHANDNA AR, KUHLMANN N, BRYCE CA, GREBA Q, CAMPANUCCI VA AND HOWLAND JG. 2015. Chronic maternal hyperglycemia induced during mid-pregnancy in rats increases RAGE expression, augments hippocampal excitability, and alters behavior of the offspring. Neuroscience 303: 241-260.). This behavior persisted even when euglycemia was ensured by insulin treatment during pregnancy, but authors did not further evaluate the mechanisms underlying this interesting behavioral finding.

NEURODEGENERATIVE DISEASES

Alzheimer’s disease (AD) is the most common form of dementia in the elderly. Increasing evidence suggests that AD development is influenced by events that take place throughout life, manifesting itself as a consequence of cumulative factors (De Felice, 2013). It has been hypothesized that adverse environments early in life may influence how neurons interact with microglia and astrocytes, making cells over reactive when exposed to the amyloid-β peptide, which is generated in the brain under physiological conditions and form large extracellular deposits during disease development (Ferreira and Klein 2011FERREIRA ST AND KLEIN WL. 2011. The Abeta oligomer hypothesis for synapse failure and memory loss in Alzheimer’s disease. Neurobiol Learn Mem 96: 529-543.). Epidemiological data on how gestational diabetes and maternal obesity influence the development of AD and other neurodegenerative disease are still lacking. In an interesting study using a classical AD transgenic model (3xTg), female mice were treated with a high-fat diet during pregnancy and lactation. Maternal obesity did not increase Aβ load in the brains of adult offspring, but higher levels of phosphorylated Tau protein were found in the hippocampus of these animals, which was associated to a worsened performance in several memory tasks (Martin et al. 2014MARTIN SA, JAMESON CH, ALLAN SM AND LAWRENCE CB. 2014. Maternal high-fat diet worsens memory deficits in the triple-transgenic (3xTgAD) mouse model of Alzheimer’s disease. PLoS One 9: e99226.). Similar experiments were performed in Tg2576 AD transgenic mice, and an increased amyloid-β burden was found in the brains of the offspring as a result of maternal obesity (Nizari et al. 2016NIZARI S, CARARE RO AND HAWKES CA. 2016. Increased Abeta pathology in aged Tg2576 mice born to mothers fed a high fat diet. Sci Rep 6: 21981.). Interestingly, Hawkes et al. (2015HAWKES CA, GENTLEMAN SM, NICOLL JA AND CARARE RO. 2015. Prenatal high-fat diet alters the cerebrovasculature and clearance of beta-amyloid in adult offspring. J Pathol 235: 619-631.) showed that exposure of pregnant mice to high-fat diet leads to changes in multiple components of the neurovascular unit of the offspring, which impairs perivascular clearance of Aβ from their brains, favoring amyloid deposition. Although these studies clearly support a possible role of gestational health on the development of neurodegenerative diseases in adult life, more studies should be performed in order to directly investigate the consequences of intra-uterine exposure to hyperglycemia, hyperinsulinemia and inflammatory markers in the development of neurodegenerative disorders later in life as well as scrutinize the possible underlying mechanisms.

CONCLUSIONS

The developing brain is extremely sensitive to endogenous and exogenous signals. GD is a condition where fetuses are exposed to high circulating levels of glucose and increased pro-inflammatory mediators during a critical period of brain development (Melo et al. 2014MELO AM, BENATTI RO, IGNACIO-SOUZA LM, OKINO C, TORSONI AS, MILANSKI M, VELLOSO LA AND TORSONI MA. 2014. Hypothalamic endoplasmic reticulum stress and insulin resistance in offspring of mice dams fed high-fat diet during pregnancy and lactation. Metabolism 63: 682-692., Tang et al. 2015TANG X, QIN Q, XIE X AND HE P. 2015. Protective effect of sRAGE on fetal development in pregnant rats with gestational diabetes mellitus. Cell Biochem Biophys 71: 549-556.). Although gross malformations are reported in 3-5% of children delivered by GD mothers (Wren et al. 2003WREN C, BIRRELL G AND HAWTHORNE G. 2003. Cardiovascular malformations in infants of diabetic mothers. Heart 89: 1217-1220., Gharehbaghi and Ghaemi 2010GHAREHBAGHI MM AND GHAEMI MR. 2010. Goldenhar syndrome in an infant of diabetic mother. Iran J Pediatr 20: 131-134.), hypothalamic dysfunction and obesity are expected to affect 30-40% of the offspring (Kim et al. 2012bKIM SY, SHARMA AJ AND CALLAGHAN WM. 2012b. Gestational diabetes and childhood obesity: what is the link? Curr Opin Obstet Gynecol 24: 376-381.). Therefore, other changes to brain function, behavior and development of brain diseases could be expected in the offspring of GD (Figure 1). Defects in brain insulin signaling might explain at least in part this delayed cognitive development of GD offspring, and this hypothesis has never been directly addressed. However, downregulation of insulin signaling mediators has already been reported in experimental models of GD. GD is a transient and multifactorial condition which makes it challenging to experimentally recapitulate. We believe that further studies should be performed in order to enable the development of new animal models for GD, so that long-term consequences to both mothers and offspring can be assessed in light of the disease’s complexity.

Figure 1
Consequences of gestational diabetes (GD) to the offspring. Intra-uterine exposure to hyperglycemia, hyperinsulinemia, inflammation and oxidative stress, hallmarks of GD, is associated to anatomical malformations (green circle) and hypothalamic dysfunction/obesity (red circle) in the offspring. Emerging evidence suggest that GD might be associated to behavioral and neurophysiological defects in the offspring (dashed blue circle), which may only manifest at childhood, adulthood or old age.

In developed countries, screening for GD is mandatory and treatment of this condition reaches a high percentage of these patients, suggesting that glycemia is kept within normal levels during the rest of gestation. In animal models, maintenance of euglycemia by insulin treatment has been associated to reversion of several effects in the central nervous system of the offspring (Jing et al. 2014JING YH, SONG YF, YAO YM, YIN J, WANG DG AND GAO LP. 2014. Retardation of fetal dendritic development induced by gestational hyperglycemia is associated with brain insulin/IGF-I signals. Int J Dev Neurosci 37: 15-20., Hami et al. 2013HAMI J, SADR-NABAVI A, SANKIAN M, BALALI-MOOD M AND HAGHIR H. 2013. The effects of maternal diabetes on expression of insulin-like growth factor-1 and insulin receptors in male developing rat hippocampus. Brain Struct Funct 218: 73-84.). However, standard treatment regimens are not always effective in prevention of other classical complications of GD in patients (Crowther et al. 2005CROWTHER CA, HILLER JE, MOSS JR, MCPHEE AJ, JEFFRIES WS, ROBINSON JS AND AUSTRALIAN CARBOHYDRATE INTOLERANCE STUDY IN PREGNANT WOMEN TRIAL G. 2005. Effect of treatment of gestational diabetes mellitus on pregnancy outcomes. N Engl J Med 352: 2477-2486., Landon et al. 2009LANDON MB ET AL. 2009. A multicenter, randomized trial of treatment for mild gestational diabetes. N Engl J Med 361: 1339-1348.), and there remains a need to improve treatment of diabetic pregnant women. Moreover, considering that increased levels of pro-inflammatory markers may have a key role in brain development and central insulin resistance, normalization of these markers to physiological levels in response to the classical treatment used for GD should be addressed.

Lifestyle intervention in pregnant obese women was also shown to reduce circulating levels of inflammation markers (Renault et al. 2015RENAULT KM, CARLSEN EM, NORGAARD K, NILAS L, PRYDS O, SECHER NJ, OLSEN SF AND HALLDORSSON TI. 2015. Intake of Sweets, Snacks and Soft Drinks Predicts Weight Gain in Obese Pregnant Women: Detailed Analysis of the Results of a Randomised Controlled Trial. PLoS One 10: e0133041.) and it is expected that nearly 50% of GD cases could potentially be prevented if we reduced the risk of overweight and obesity to that of normal-weight women (Kim et al. 2012aKIM SY, ENGLAND L, SAPPENFIELD W, WILSON HG, BISH CL, SALIHU HM AND SHARMA AJ. 2012a. Racial/ethnic differences in the percentage of gestational diabetes mellitus cases attributable to overweight and obesity, Florida, 2004-2007. Prev Chronic Dis 9: E88.). Therefore, changes in lifestyle habits, such as frequent exercise and healthy diet, remain the best known ways of preventing GD and its undesired long-term effects.

REFERENCES

  • ALBRECHT SS, KUKLINA EV, BANSIL P, JAMIESON DJ, WHITEMAN MK, KOURTIS AP, POSNER SF AND CALLAGHAN WM. 2010. Diabetes trends among delivery hospitalizations in the U.S., 1994-2004. Diabetes Care 33: 768-773.
  • ARRUDA AP, MILANSKI M, COOPE A, TORSONI AS, ROPELLE E, CARVALHO DP, CARVALHEIRA JB AND VELLOSO LA. 2011. Low-grade hypothalamic inflammation leads to defective thermogenesis, insulin resistance, and impaired insulin secretion. Endocrinology 152: 1314-1326.
  • BADEHNOOSH B, KARAMALI M, ZARRATI M, JAMILIAN M, BAHMANI F, TAJABADI-EBRAHIMI M, JAFARI P, RAHMANI E AND ASEMI Z. 2017. The effects of probiotic supplementation on biomarkers of inflammation, oxidative stress and pregnancy outcomes in gestational diabetes. J Matern Fetal Neonatal Med: 1-9.
  • BAEYENS L, HINDI S, SORENSON RL AND GERMAN MS. 2016. Beta-Cell adaptation in pregnancy. Diabetes Obes Metab 18(Suppl 1): 63-70.
  • BELGARDT BF ET AL. 2010. Hypothalamic and pituitary c-Jun N-terminal kinase 1 signaling coordinately regulates glucose metabolism. Proc Natl Acad Sci U S A 107: 6028-6033.
  • BENEDICT C, HALLSCHMID M, HATKE A, SCHULTES B, FEHM HL, BORN J AND KERN W. 2004. Intranasal insulin improves memory in humans. Psychoneuroendocrinology 29: 1326-1334.
  • BIESSELS GJ, KAMAL A, RAMAKERS GM, URBAN IJ, SPRUIJT BM, ERKELENS DW AND GISPEN WH. 1996. Place learning and hippocampal synaptic plasticity in streptozotocin-induced diabetic rats. Diabetes 45: 1259-1266.
  • BIHOREAU MT, KTORZA A, KINEBANYAN MF AND PICON L. 1986. Impaired glucose homeostasis in adult rats from hyperglycemic mothers. Diabetes 35: 979-984.
  • BOKSA P. 2004. Animal models of obstetric complications in relation to schizophrenia. Brain Res Brain Res Rev 45: 1-17.
  • BOLTON JL AND BILBO SD. 2014. Developmental programming of brain and behavior by perinatal diet: focus on inflammatory mechanisms. Dialogues Clin Neurosci 16: 307-320.
  • BOMFIM TR ET AL. 2012. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease- associated Abeta oligomers. J Clin Invest 122: 1339-1353.
  • CARLSON A AND DRENNAN F. 1911. The control of pancreatic diabetes in pregnancy by the passage of the internal secretion of the pancreas of the fetus to the blood of the mother. Am J Physiol 28: 391-395.
  • CHANDNA AR, KUHLMANN N, BRYCE CA, GREBA Q, CAMPANUCCI VA AND HOWLAND JG. 2015. Chronic maternal hyperglycemia induced during mid-pregnancy in rats increases RAGE expression, augments hippocampal excitability, and alters behavior of the offspring. Neuroscience 303: 241-260.
  • CHEUNG NW AND BYTH K. 2003. Population health significance of gestational diabetes. Diabetes Care 26: 2005-2009.
  • COPPS KD AND WHITE MF. 2012. Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2. Diabetologia 55: 2565-2582.
  • CRAFT S AND WATSON GS. 2004. Insulin and neurodegenerative disease: shared and specific mechanisms. Lancet Neurol 3: 169-178.
  • CROWTHER CA, HILLER JE, MOSS JR, MCPHEE AJ, JEFFRIES WS, ROBINSON JS AND AUSTRALIAN CARBOHYDRATE INTOLERANCE STUDY IN PREGNANT WOMEN TRIAL G. 2005. Effect of treatment of gestational diabetes mellitus on pregnancy outcomes. N Engl J Med 352: 2477-2486.
  • CUTHBERT FP, IVY AC, ISAACS BL AND GRAY J. 1936. The relation of pregnancy and lactation to extirpation diabetis in the dog. Am J Physiol 115: 480-496.
  • CYPRYK K, SZYMCZAK W, CZUPRYNIAK L, SOBCZAK M AND LEWINSKI A. 2008. Gestational diabetes mellitus - an analysis of risk factors. Endokrynol Pol 59: 393-397.
  • DARAKI V, ROUMELIOTAKI T, KOUTRA K, GEORGIOU V, KAMPOURI M, KYRIKLAKI A, VAFEIADI M, PAPAVASILIOU S, KOGEVINAS M AND CHATZI L. 2017. Effect of parental obesity and gestational diabetes on child neuropsychological and behavioral development at 4 years of age: the Rhea mother-child cohort, Crete, Greece. Eur Child Adolesc Psychiatry 26: 703-714.
  • DEBOER T, WEWERKA S, BAUER PJ, GEORGIEFF MK AND NELSON CA. 2005. Explicit memory performance in infants of diabetic mothers at 1 year of age. Dev Med Child Neurol 47: 525-531.
  • DE FELICE FG. 2013. Alzheimer’s disease and insulin resistance: translating basic science into clinical applications. J Clin Invest 123: 531-539.
  • DEMIRCI C, ERNST S, ALVAREZ-PEREZ JC, ROSA T, VALLE S, SHRIDHAR V, CASINELLI GP, ALONSO LC, VASAVADA RC AND GARCIA-OCANA A. 2012. Loss of HGF/c-Met signaling in pancreatic beta-cells leads to incomplete maternal beta-cell adaptation and gestational diabetes mellitus. Diabetes 61: 1143-1152.
  • DESISTO CL, KIM SY AND SHARMA AJ. 2014. Prevalence estimates of gestational diabetes mellitus in the United States, Pregnancy Risk Assessment Monitoring System (PRAMS), 2007-2010. Prev Chronic Dis 11: E104.
  • DÖRNER G AND MOHNIKE A. 1976. Further evidence for a predominantly maternal transmission of maturity-onset type diabetes. Endokrinologie 68: 121-124.
  • ESTES ML AND MCALLISTER AK. 2016. Maternal immune activation: Implications for neuropsychiatric disorders. Science 353: 772-777.
  • FERRARA A. 2007. Increasing prevalence of gestational diabetes mellitus: a public health perspective. Diabetes Care 30(Suppl 2): S141-146.
  • FERREIRA ST AND KLEIN WL. 2011. The Abeta oligomer hypothesis for synapse failure and memory loss in Alzheimer’s disease. Neurobiol Learn Mem 96: 529-543.
  • FORNY-GERMANO L ET AL. 2014. Alzheimer’s disease-like pathology induced by amyloid-beta oligomers in nonhuman primates. J Neurosci 34: 13629-13643.
  • FRIAS JL, FRIAS JP, FRIAS PA AND MARTINEZ-FRIAS ML. 2007. Infrequently studied congenital anomalies as clues to the diagnosis of maternal diabetes mellitus. Am J Med Genet A 143A: 2904-2909.
  • FRIEDMAN JE, KIRWAN JP, JING M, PRESLEY L AND CATALANO PM. 2008. Increased skeletal muscle tumor necrosis factor-alpha and impaired insulin signaling persist in obese women with gestational diabetes mellitus 1 year postpartum. Diabetes 57: 606-613.
  • GARCIA-VARGAS L, ADDISON SS, NISTALA R, KURUKULASURIYA D AND SOWERS JR. 2012. Gestational Diabetes and the Offspring: Implications in the Development of the Cardiorenal Metabolic Syndrome in Offspring. Cardiorenal Med 2: 134-142.
  • GARDENER H, SPIEGELMAN D AND BUKA SL. 2009. Prenatal risk factors for autism: comprehensive meta-analysis. Br J Psychiatry 195: 7-14.
  • GEARHART MM AND PARBHOO SK. 2006. Hyperglycemia in the critically ill patient. AACN Clin Issues 17: 50-55.
  • GHAREHBAGHI MM AND GHAEMI MR. 2010. Goldenhar syndrome in an infant of diabetic mother. Iran J Pediatr 20: 131-134.
  • GILMARTIN AB, URAL SH AND REPKE JT. 2008. Gestational diabetes mellitus. Rev Obstet Gynecol 1: 129-134.
  • GOLALIPOUR MJ, KAFSHGIRI SK AND GHAFARI S. 2012. Gestational diabetes induced neuronal loss in CA1 and CA3 subfields of rat hippocampus in early postnatal life. Folia Morphol (Warsz) 71: 71-77.
  • GUPTA RK, GAO N, GORSKI RK, WHITE P, HARDY OT, RAFIQ K, BRESTELLI JE, CHEN G, STOECKERT CJ JR AND KAESTNER KH. 2007. Expansion of adult beta-cell mass in response to increased metabolic demand is dependent on HNF-4alpha. Genes Dev 21: 756-769.
  • HAMI J, SADR-NABAVI A, SANKIAN M, BALALI-MOOD M AND HAGHIR H. 2013. The effects of maternal diabetes on expression of insulin-like growth factor-1 and insulin receptors in male developing rat hippocampus. Brain Struct Funct 218: 73-84.
  • HAWKES CA, GENTLEMAN SM, NICOLL JA AND CARARE RO. 2015. Prenatal high-fat diet alters the cerebrovasculature and clearance of beta-amyloid in adult offspring. J Pathol 235: 619-631.
  • HOTAMISLIGIL GS. 2008. Inflammation and endoplasmic reticulum stress in obesity and diabetes. Int J Obes (Lond) 32(Suppl 7): S52-54.
  • IWASHYNA TJ, ELY EW, SMITH DM AND LANGA KM. 2010. Long-term cognitive impairment and functional disability among survivors of severe sepsis. JAMA 304: 1787-1794.
  • JAWERBAUM A, CATAFAU JR, GONZALES ET, RODRIGUEZ RR, GELPI E, GOMEZ G, GIMENO AL AND GIMENO MA. 1993. Eicosanoid production by uterine strips and by embryos obtained from diabetic pregnant rats. Prostaglandins 45: 487-495.
  • JING YH, SONG YF, YAO YM, YIN J, WANG DG AND GAO LP. 2014. Retardation of fetal dendritic development induced by gestational hyperglycemia is associated with brain insulin/IGF-I signals. Int J Dev Neurosci 37: 15-20.
  • JUNOD A, LAMBERT AE, STAUFFACHER W AND RENOLD AE. 1969. Diabetogenic action of streptozotocin: relationship of dose to metabolic response. J Clin Invest 48: 2129-2139.
  • KARNIK SK, CHEN H, MCLEAN GW, HEIT JJ, GU X, ZHANG AY, FONTAINE M, YEN MH AND KIM SK. 2007. Menin controls growth of pancreatic beta-cells in pregnant mice and promotes gestational diabetes mellitus. Science 318: 806-809.
  • KELSTRUP L, CLAUSEN TD, MATHIESEN ER, HANSEN T AND DAMM P. 2012. Low-grade inflammation in young adults exposed to intrauterine hyperglycemia. Diabetes Res Clin Pract 97: 322-330.
  • KIM H ET AL. 2010. Serotonin regulates pancreatic beta cell mass during pregnancy. Nat Med 16: 804-808.
  • KIM SY, ENGLAND L, SAPPENFIELD W, WILSON HG, BISH CL, SALIHU HM AND SHARMA AJ. 2012a. Racial/ethnic differences in the percentage of gestational diabetes mellitus cases attributable to overweight and obesity, Florida, 2004-2007. Prev Chronic Dis 9: E88.
  • KIM SY, SHARMA AJ AND CALLAGHAN WM. 2012b. Gestational diabetes and childhood obesity: what is the link? Curr Opin Obstet Gynecol 24: 376-381.
  • KINNEY BA, RABE MB, JENSEN RA AND STEGER RW. 2003. Maternal hyperglycemia leads to gender-dependent deficits in learning and memory in offspring. Exp Biol Med (Maywood) 228: 152-159.
  • KOVACS P AND HAJNAL A. 2009. In vivo electrophysiological effects of insulin in the rat brain. Neuropeptides 43: 283-293.
  • LAMBIN S, VAN BREE R, CALUWAERTS S, VERCRUYSSE L, VERGOTE I AND VERHAEGHE J. 2007. Adipose tissue in offspring of Lepr(db/+) mice: early-life environment vs. genotype. Am J Physiol Endocrinol Metab 292: E262-271.
  • LANDON MB ET AL. 2009. A multicenter, randomized trial of treatment for mild gestational diabetes. N Engl J Med 361: 1339-1348.
  • LEE CC, KUO YM, HUANG CC AND HSU KS. 2009. Insulin rescues amyloid beta-induced impairment of hippocampal long-term potentiation. Neurobiol Aging 30: 377-387.
  • LENZEN S AND PANTEN U. 1988. Alloxan: history and mechanism of action. Diabetologia 31: 337-342.
  • LINDER K, SCHLEGER F, KIEFER-SCHMIDT I, FRITSCHE L, KUMMEL S, HENI M, WEISS M, HARING HU, PREISSL H AND FRITSCHE A. 2015. Gestational Diabetes Impairs Human Fetal Postprandial Brain Activity. J Clin Endocrinol Metab 100: 4029-4036.
  • LIU TW, HEDEN TD, MATTHEW MORRIS E, FRITSCHE KL, VIEIRA-POTTER VJ AND THYFAULT JP. 2015. High-Fat Diet Alters Serum Fatty Acid Profiles in Obesity Prone Rats: Implications for In Vitro Studies. Lipids 50: 997-1008.
  • LOURENCO MV ET AL. 2013. TNF-alpha mediates PKR-dependent memory impairment and brain IRS-1 inhibition induced by Alzheimer’s beta-amyloid oligomers in mice and monkeys. Cell Metab 18: 831-843.
  • LOWE LP, METZGER BE, LOWE WL JR, DYER AR, MCDADE TW, MCINTYRE HD AND GROUP HSCR. 2010. Inflammatory mediators and glucose in pregnancy: results from a subset of the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study. J Clin Endocrinol Metab 95: 5427-5434.
  • MA QL ET AL. 2009. Beta-amyloid oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling: suppression by omega-3 fatty acids and curcumin. J Neurosci 29: 9078-9089.
  • MARKOWITZ J AND SOSKIN S. 1927. Pancreatic diabetes and pregnancy. Am J Physiol 79: 553-558.
  • MARTIN SA, JAMESON CH, ALLAN SM AND LAWRENCE CB. 2014. Maternal high-fat diet worsens memory deficits in the triple-transgenic (3xTgAD) mouse model of Alzheimer’s disease. PLoS One 9: e99226.
  • MELO AM, BENATTI RO, IGNACIO-SOUZA LM, OKINO C, TORSONI AS, MILANSKI M, VELLOSO LA AND TORSONI MA. 2014. Hypothalamic endoplasmic reticulum stress and insulin resistance in offspring of mice dams fed high-fat diet during pregnancy and lactation. Metabolism 63: 682-692.
  • NEVES FS ET AL. 2016. Brain-Defective Insulin Signaling Is Associated to Late Cognitive Impairment in Post-Septic Mice. Mol Neurobiol.
  • NGUYEN HT, BHATTARAI JP, PARK SJ, LEE JC, CHO DH AND HAN SK. 2015. Enhanced GABA action on the substantia gelatinosa neurons of the medullary dorsal horn in the offspring of streptozotocin-injected mice. J Diabetes Complications 29: 629-636.
  • NIZARI S, CARARE RO AND HAWKES CA. 2016. Increased Abeta pathology in aged Tg2576 mice born to mothers fed a high fat diet. Sci Rep 6: 21981.
  • OTT V, BENEDICT C, SCHULTES B, BORN J AND HALLSCHMID M. 2012. Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism. Diabetes Obes Metab 14: 214-221.
  • PANDHARIPANDE PP ET AL. 2013. Long-term cognitive impairment after critical illness. N Engl J Med 369: 1306-1316.
  • PASEK RC AND GANNON M. 2013. Advancements and challenges in generating accurate animal models of gestational diabetes mellitus. Am J Physiol Endocrinol Metab 305: E1327-1338.
  • PLAGEMANN A, HARDER T, LINDNER R, MELCHIOR K, RAKE A, RITTEL F, ROHDE W AND DORNER G. 1998. Alterations of hypothalamic catecholamines in the newborn offspring of gestational diabetic mother rats. Brain Res Dev Brain Res 109: 201-209.
  • PLANK JL, FRIST AY, LEGRONE AW, MAGNUSON MA AND LABOSKY PA. 2011. Loss of Foxd3 results in decreased beta-cell proliferation and glucose intolerance during pregnancy. Endocrinology 152: 4589-4600.
  • PLUM L, SCHUBERT M AND BRUNING JC. 2005. The role of insulin receptor signaling in the brain. Trends Endocrinol Metab 16: 59-65.
  • POSTON L. 2010. Developmental programming and diabetes - The human experience and insight from animal models. Best Pract Res Clin Endocrinol Metab 24: 541-552.
  • POSTON L. 2011. Intergenerational transmission of insulin resistance and type 2 diabetes. Prog Biophys Mol Biol 106: 315-322.
  • RAMANATHAN M, JAISWAL AK AND BHATTACHARYA SK. 2000. Hyperglycaemia in pregnancy: effects on the offspring behaviour with special reference to anxiety paradigms. Indian J Exp Biol 38: 231-236.
  • REISINGER S, KHAN D, KONG E, BERGER A, POLLAK A AND POLLAK DD. 2015. The poly(I:C)-induced maternal immune activation model in preclinical neuropsychiatric drug discovery. Pharmacol Ther 149: 213-226.
  • RENAULT KM, CARLSEN EM, NORGAARD K, NILAS L, PRYDS O, SECHER NJ, OLSEN SF AND HALLDORSSON TI. 2015. Intake of Sweets, Snacks and Soft Drinks Predicts Weight Gain in Obese Pregnant Women: Detailed Analysis of the Results of a Randomised Controlled Trial. PLoS One 10: e0133041.
  • RIGGINS T, BAUER PJ, GEORGIEFF MK AND NELSON CA. 2010. Declarative memory performance in infants of diabetic mothers. Adv Child Dev Behav 38: 73-110.
  • SALKOVIC-PETRISIC M AND HOYER S. 2007. Central insulin resistance as a trigger for sporadic Alzheimer-like pathology: an experimental approach. J Neural Transm Suppl: 217-233.
  • SAVITZ DA, JANEVIC TM, ENGEL SM, KAUFMAN JS AND HERRING AH. 2008. Ethnicity and gestational diabetes in New York City, 1995-2003. BJOG 115: 969-978.
  • SEMMLER A ET AL. 2013. Persistent cognitive impairment, hippocampal atrophy and EEG changes in sepsis survivors. J Neurol Neurosurg Psychiatry 84: 62-69.
  • SILVERMAN BL, METZGER BE, CHO NH AND LOEB CA. 1995. Impaired glucose tolerance in adolescent offspring of diabetic mothers. Relationship to fetal hyperinsulinism. Diabetes Care 18: 611-617.
  • SRINIVASAN M, KATEWA SD, PALANIYAPPAN A, PANDYA JD AND PATEL MS. 2006. Maternal high-fat diet consumption results in fetal malprogramming predisposing to the onset of metabolic syndrome-like phenotype in adulthood. Am J Physiol Endocrinol Metab 291: E792-799.
  • STECULORUM SM AND BOURET SG. 2011. Maternal diabetes compromises the organization of hypothalamic feeding circuits and impairs leptin sensitivity in offspring. Endocrinology 152: 4171-4179.
  • TAKEDA S, SATO N, UCHIO-YAMADA K, SAWADA K, KUNIEDA T, TAKEUCHI D, KURINAMI H, SHINOHARA M, RAKUGI H AND MORISHITA R. 2010. Diabetes-accelerated memory dysfunction via cerebrovascular inflammation and Abeta deposition in an Alzheimer mouse model with diabetes. Proc Natl Acad Sci U S A 107: 7036-7041.
  • TANG X, QIN Q, XIE X AND HE P. 2015. Protective effect of sRAGE on fetal development in pregnant rats with gestational diabetes mellitus. Cell Biochem Biophys 71: 549-556.
  • TAYLOR PD ET AL. 2005. Impaired glucose homeostasis and mitochondrial abnormalities in offspring of rats fed a fat-rich diet in pregnancy. Am J Physiol Regul Integr Comp Physiol 288: R134-139.
  • THALER JP ET AL. 2012. Obesity is associated with hypothalamic injury in rodents and humans. J Clin Invest 122: 153-162.
  • TORRES-ESPINOLA FJ ET AL. 2015. Maternal Obesity, Overweight and Gestational Diabetes Affect the Offspring Neurodevelopment at 6 and 18 Months of Age--A Follow Up from the PREOBE Cohort. PLoS One 10: e0133010.
  • VAN LIESHOUT RJ AND VORUGANTI LP. 2008. Diabetes mellitus during pregnancy and increased risk of schizophrenia in offspring: a review of the evidence and putative mechanisms. J Psychiatry Neurosci 33: 395-404.
  • WAN Q, XIONG ZG, MAN HY, ACKERLEY CA, BRAUNTON J, LU WY, BECKER LE, MACDONALD JF AND WANG YT. 1997. Recruitment of functional GABA(A) receptors to postsynaptic domains by insulin. Nature 388: 686-690.
  • WEISSMANN L ET AL. 2014. IKKepsilon is key to induction of insulin resistance in the hypothalamus, and its inhibition reverses obesity. Diabetes 63: 3334-3345.
  • WOJCIK M, MAC-MARCJANEK K, NADEL I, WOZNIAK L AND CYPRYK K. 2015. Gestational diabetes mellitus is associated with increased leukocyte peroxisome proliferator-activated receptor gamma expression. Arch Med Sci 11: 779-787.
  • WOODS SC, LOTTER EC, MCKAY LD AND PORTE D JR. 1979. Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature 282: 503-505.
  • WREN C, BIRRELL G AND HAWTHORNE G. 2003. Cardiovascular malformations in infants of diabetic mothers. Heart 89: 1217-1220.
  • XIANG AH, WANG X, MARTINEZ MP, WALTHALL JC, CURRY ES, PAGE K, BUCHANAN TA, COLEMAN KJ AND GETAHUN D. 2015. Association of maternal diabetes with autism in offspring. JAMA 313: 1425-1434.
  • YESSOUFOU A AND MOUTAIROU K. 2011. Maternal diabetes in pregnancy: early and long-term outcomes on the offspring and the concept of “metabolic memory”. Exp Diabetes Res 2011: 218598.
  • ZHANG H, ZHANG J, POPE CF, CRAWFORD LA, VASAVADA RC, JAGASIA SM AND GANNON M. 2010. Gestational diabetes mellitus resulting from impaired beta-cell compensation in the absence of FoxM1, a novel downstream effector of placental lactogen. Diabetes 59: 143-152.
  • ZHANG TY AND MEANEY MJ. 2010. Epigenetics and the environmental regulation of the genome and its function. Annu Rev Psychol 61: 439-466, C431-433.
  • ZHAO WQ AND ALKON DL. 2001. Role of insulin and insulin receptor in learning and memory. Mol Cell Endocrinol 177: 125-134.
  • ZHAO WQ, CHEN H, QUON MJ AND ALKON DL. 2004. Insulin and the insulin receptor in experimental models of learning and memory. Eur J Pharmacol 490: 71-81.
  • **
    Contribution to the centenary of the Brazilian Academy of Sciences.

Publication Dates

  • Publication in this collection
    14 Aug 2017
  • Date of issue
    Aug 2018

History

  • Received
    11 Apr 2017
  • Accepted
    30 May 2017
Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100 - Rio de Janeiro - RJ - Brazil
E-mail: aabc@abc.org.br