Topic Highlight Open Access
Copyright ©2011 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Diabetes. Nov 15, 2011; 2(11): 196-203
Published online Nov 15, 2011. doi: 10.4239/wjd.v2.i11.196
Consequences of gestational and pregestational diabetes on placental function and birth weight
Anne Vambergue, EA 4489 “Perinatal Environment and Fetal Growth”, Department of Diabetology, Huriez Hospital, 59800 CHRU Lille, France
Isabelle Fajardy, EA 4489 “Perinatal Environment and Fetal Growth”, Biology and Pathology Center, 59800 CHRU Lille, France
Author contributions: Vambergue A and Fajardy I designed and wrote Part I, II, IV, V and part III respectively; all authors approved the final version of the topics (manuscript) to be published.
Correspondence to: Anne Vambergue, Professor, EA 4489 “Perinatal Environment and Fetal Growth”, Department of Diabetology, Huriez Hospital, 59800 CHRU Lille, France. anne.vambergue@chru-lille.fr
Telephone: +33-320-446816 Fax: +33-320-444184
Received: March 18, 2011
Revised: October 19, 2011
Accepted: October 26, 2011
Published online: November 15, 2011

Abstract

Maternal diabetes constitutes an unfavorable environment for embryonic and fetoplacental development. Despite current treatments, pregnant women with pregestational diabetes are at increased risk for congenital malformations, materno-fetal complications, placental abnormalities and intrauterine malprogramming. The complications during pregnancy concern the mother (gravidic hypertension and/or preeclampsia, cesarean section) and the fetus (macrosomia or intrauterine growth restriction, shoulder dystocia, hypoglycemia and respiratory distress). The fetoplacental impairment and intrauterine programming of diseases in the offspring’s later life induced by gestational diabetes are similar to those induced by type 1 and type 2 diabetes mellitus. Despite the existence of several developmental and morphological differences in the placenta from rodents and women, there are similarities in the alterations induced by maternal diabetes in the placenta from diabetic patients and diabetic experimental models. From both human and rodent diabetic experimental models, it has been suggested that the placenta is a compromised target that largely suffers the impact of maternal diabetes. Depending on the maternal metabolic and proinflammatory derangements, macrosomia is explained by an excessive availability of nutrients and an increase in fetal insulin release, a phenotype related to the programming of glucose intolerance. The degree of fetal damage and placental dysfunction and the availability and utilisation of fetal substrates can lead to the induction of macrosomia or intrauterine growth restriction. In maternal diabetes, both the maternal environment and the genetic background are important in the complex and multifactorial processes that induce damage to the embryo, the placenta, the fetus and the offspring. Nevertheless, further research is needed to better understand the mechanisms that govern the early embryo development, the induction of congenital anomalies and fetal overgrowth in maternal diabetes.

Key Words: Maternal diabetes, Placental function, Birth weight, Macrosomia, Intrauterine growth retardation



INTRODUCTION

Diabetes in pregnant women is associated with an increased risk of maternal and neonatal morbidity and remains a significant medical challenge.

Diabetes during pregnancy may be divided into clinical diabetes (women with previously diagnosed with type 1 or type 2 diabetes) and gestational diabetes. The American Diabetes Association defines gestational diabetes as “any degree of glucose intolerance with onset or first recognition during pregnancy”, but provides diagnostic thresholds for fasting and post-glucose loading values. The International Association of Diabetes in Pregnancy Study Groups recently published a consensus derived from the Hyperglycemia Adverse Pregnancy Outcome study data, suggested that all pregnant women without known diabetes should have a 75 g oral glucose tolerance test at 24-28 wk of gestation[1]. Gestational diabetes would be diagnosed if one or more values met or exceeded the following levels of glucose: fasting 5.1 mmol/L, 1 h post glucose 10.0 mmol/L and 2 h post glucose 8.5 mmol/L.

While diabetes in pregnancy is associated with increased obstetric risk compared with normal pregnancy, the overall contribution of diabetes to most obstetric and neonatal complications on a population basis is low, with the largest impact being on shoulder dystocia. Except malformations, which are likely to have resulted from preconceptional or periconceptional hyperglycemia, improvements in obstetric practice have led to major reductions in adverse outcomes. Prepregnancy care for women with diabetes was introduced a long time ago and is associated with improved pregnancy outcomes. However, overall pregnancy outcomes remain very poor for women with diabetes with only a third receiving prepregnancy care. The importance of other metabolic factors, such as obesity and hypertriglyceridemia, in pregnancy are also now increasingly being recognized.

As well as its effects on perinatal outcomes, the intrauterine environment is a key determinant of child and adult health, particularly of conditions associated with metabolic disturbances[2-4]. Aberrant fetal growth has been linked to the development of metabolic diseases in later life, with many authors describing J or U-shaped curves linking both low and high birth weights with these conditions[5,6].

The diabetic intrauterine environment affects the offspring of women with all types of diabetes mellitus (DM). Therefore, type 2 diabetes is very often associated with obesity. It is possible that changes in placental structure and function may exist in obesity independently of diabetes. But, at this time, we do not have sufficient data in the literature to distinguish between what is related to obesity or to diabetes. The offspring of women with diabetes during pregnancy is at higher risk of developing hypertension and other cardiovascular disease. The intrauterine environment represents a vicious cycle with the offspring being at risk of developing gestational diabetes or diabetes at a young age. More recently, it has been evocated that environmental signals can alter the epigenetic state of specific genes and modulate their activity. It is possible that modulation of epigenetic states provides a plausible mechanism by which maternal diabetes can mediate known long-term effects on risk for type 2 diabetes for the offspring, but we need more information to understand the concept of programming in maternal diabetes.

PLACENTA IN DIABETES

The placenta is located at the interface between the maternal and fetal circulation with fundamental functions for pregnancy. It has been postulated that the diabetic environment may have profound effects on placental development and function. It is very important to be precise that these specific effects will depend on the time period in gestation[7]. Maternal and fetal hyperglycemias are likely to have an impact on the production of various placental proteins. These maternal and fetal hyperglycemiae also affect placental metabolism, growth and development.

Despite the improvement in maternal glycemic control, structural and functional changes of the diabetic placenta at term may occur independently of the type of diabetes. In the case of diabetes, the surface area is particularly increased in the periphery of the villous tree. The diffusion distance between the maternal and fetal systemic circulations is increased due to a thickening of the trophoblastic basement membrane with higher amounts of collagen, predominantly type IV[8].

In type 1 diabetes and in gestational DM (GDM), the villous stroma is slightly edematous with an over-representation of Hofbauer cells which are the placental resident macrophages[9]. The increased number of these cells contributes to a higher release of placental cytokines such as leptin, tumor necrosis factor-α (TNF-α) and interleukins, and subsequently modifies placental metabolic and endocrine functions[10]. Enlargement of the capillary surface area with capillary proliferation and penetration of newly formed vessels has been also described in maternal diabetes[11]. So the result is a hyper-vascularization and an increased surface of exchange that could contribute to oxygen diffusion across the placenta to compensate for the impaired maternal-fetal transfer of diffusion-limited substances.

Placental weight tends to be heavier in diabetes, similar to fetal weight, but the weight gain is more pronounced in the placenta than in the fetus, as is reflected in a higher placental-to-fetal weight ratio than in normal gestation. Placentomegaly is correlated with fetal macrosomia confirming the close correlation of placental weight with that of the offspring[12]. Actually, it is not possible to determine if placental overweight is the cause or the consequence of fetal overweight.

Modification in placental transport

Despite altered expression of placental glucose transporters, an unchanged transplacental glucose transport in GDM has been demonstrated. Taricco et al[13] showed that there was an unchanged concentration difference for glucose in umbilical arteries and veins in GDM. The higher flux results from the steeper maternal-to-fetal concentration gradient as the major reason for increased glucose across the placenta in diabetes.

Amino acid transport may be also altered in diabetes[14]. Changes in placental amino acid transporters are not associated with maternal diabetes, but rather with elevated fetal weight[15]. We do not know if increased nutrient transport will stimulate fetal growth or serve to cover the increased fetal nutrient in case of overgrowth of the fetus.

Modification in materno-placental oxygen supply

The placental structure is altered in pregestational and gestational diabetes. The surface and exchange areas are enlarged as a result of hyperproliferation and hypervascularization. The underlying mechanisms are unclear but we can not exclude the role of the maternal hyperglycemia associated with other maternal factors[16].

In diabetes, it has been shown that the maternal-placental oxygen supply is reduced[17]. In addition to impaired oxygen supply, fetal oxygen demand is increased. This phenomenon could be explained by aerobic metabolism which is stimulated by fetal hyperinsulinemia. The resulting low fetal oxygen levels upregulate the transcription synthesis of proangiogenic factors such as leptin, vascular endothelial growth factor (VEGF) or fibroblast growth factor 2 (FGF2)[18,19]. In excess, these factors promote placental endothelial cell proliferation. In GDM and in type 1 DM (T1DM), alterations in fetal levels of proangiogenic [VEGF, FGF2, leptin, OGF1, insulin-like growth factor (IGF)2, hypoxia] or anti-angiogenic (TNF-α) factors have been reported. Both types of diabetes are characterized by enhanced vascularisation.

The diabetic milieu will have an influence on placental development and function especially during the first trimester. In this period, the placental structures are formed and the placenta is more sensitive to modifications. The hyperglycemia can induce a reduction of trophoblast proliferation which delays placental growth and development, especially in the first gestational weeks[20]. This mechanism could explain the higher incidence of spontaneous abortion, pre-eclampsia and intrauterine growth restriction associated with diabetes, and suggest impaired trophoblast invasion[21].

More recently, it has been shown that placental expression and activity of the matrix metalloproteinases (MMPs) as MMP14 and MMP15 are elevated in diabetes, especially in type 1 diabetes induced by maternal hyperinsulinemia and TNF-α[21]. MMP14 and MMP15 are proteases which are involved in tissue remodeling processes associated with invasion, angiogenesis and proliferation. The active form of placental MMP14 is elevated in diabetes. It is possible that hypoxic situations in the villous placental structure may be implicated as a cause of increased MMP14 activity.

SUITABLE ANIMAL MODELS ARE NEEDED TO STUDY DIABETES DURING PREGNANCY

With regard to diabetes in pregnancy, experimental findings from animal models and particularly rodents were developed with the purpose of enhancing understanding of pathophysiological mechanisms and further finding treatment strategies to maintain the closest to normal metabolic intrauterine milieu, improving perinatal development by preventing fetal growth restriction or macrosomia. Resulting diabetes in various animal species, including sheep, pig and rabbit have been described in many reviews[22-25]. However, the most often used experimental models are rodents (Wistar rat) because of their convenient maintenance, short length of pregnancy and multiparity, thus enabling studies on multiple fetuses and generations. The aim of type 1 and type 2 animal models is to reproduce metabolic maternal and fetal effects similar to pregestational and GDM. But which is the most suitable model? It has been shown that children born from either pre-existing T1DM, type 2 DM (T2DM) and GDM are equally exposed to impaired glucose intolerance[26]. How to reproduce the more accurately metabolic maternal end neonatal effects of diabetes? What is the most efficient tool? The efficiency of an experimental model is based on the observation of a stable hyperglycemia throughout gestation.

We will first distinguish models of inducing maternal diabetes using chemical agents able to damage the maternal β cell with short term consequences in fetuses. Secondly, long term induction of metabolic diseases by nutritional changes occurring before and/or during pregnancy named fetal programming models. We will then expose genetically predetermined diabetes animal models.

Chemical models of diabetes during pregnancy

Chemical methods used to induce damage to the pancreatic β-cells are obtained through the administration of drugs such as streptozotocin (STZ).

STZ: STZ is a potent alkylating agent able to methylate DNA and is often used to induce diabetes in experimental animals due to its toxic effects on pancreatic β-cells[27]. The nitrosurea moiety of STZ is responsible for its cellular toxicity, which is probably mediated through a decrease in nicotinamide (NAD) levels and the production of intracellular free radicals. Beta cells are particularly sensitive to STZ due to their low level of NAD. Many different approaches have been used regarding the mode of injection (intravenous or intraperitoneal), the doses (30 to 50 mg/kg) and the stage (pre gestational and gestational) leading to severe or mild diabetes and subsequent opposite fetal phenotypes (intra-uterine growth restriction or macrosomic[28,29]). In fact, mildly diabetic dams are hyperglycemic (glycemia between 120 and 300 mg/dL), hypoinsulinemic and give birth to macrosomic fetuses. In contrast, severely diabetic mothers are insulin deficient, hyperglycemic with low body weight and give birth to microsomic and malformed fetuses. The mechanisms are a brutal destruction of the mother and fetus’s pancreas and intrauterine growth restriction is due to lack of insulin[30]. The altered maternal-fetal metabolic fuel relationship resulting from diabetes in pregnancy modulates fetal growth. The increase in fetal glucose and insulin availability with maternal diabetes is strongly associated with the development of fetal macrosomia but severe DM or diabetes of long duration restricts fetal growth. Rat fetal body weight correlates positively with maternal glucose in diabetic rats with a glucose level less than 220 mg/dL but it correlates negatively in rats with a level above 220 mg/dL. Merzouk and Soulimane-Mokhtari[31] verified that mildly hyperglycemic dams have fetuses that are large for gestational age, classified as macrosomic, but this data has been hardly reproduced in other studies[32]. In 2010, other authors observed an increase of placental weight in their mildly diabetic rats showing a compensatory mechanism to assure the maternal fetal exchanges contribution to fetal development[33]. The degree of fetal damage and placental dysfunction and the availability and utilization of fetal substrates can lead to the induction of macrosomia or microsomia. We observed a U-shaped relationship between offspring weight and metabolic changes, like in clinical studies.

STZ + NAD chemical model: NAD is an anti oxidant molecule that protects the β cell destruction induced by STZ. In 1998, Masiello and co-workers developed NAD-STZ diabetes model induced by STZ and partially protected with a suitable dose of NAD[34]. In this NAD-STZ model, the diabetic syndrome shares a number of features with human T2DM such as a stable moderate hyperglycemia, glucose intolerance, an altered insulin secretion and a reduction in pancreatic β cell mass. So a mild diabetic model might be induced by STZ under NAD protection.

Using STZ in experimental models: Chemical models are characterized by a toxic action against β cells leading to a destruction of producing insulin cell without insulin resistance. These models mimic a type 1 diabetic state, despite the lack of immunological and genetic disorders. Thus, we obtain a type 1 maternal diabetes quite different from gestational diabetes which is characterized by a lack of adaptation of β cells to metabolic changes occurring during pregnancy and by an enhanced resistance to insulin.

Nevertheless, some authors have tried to mimic gestational diabetes by varying the doses and the window of injection of STZ (in the neonatal period[35,36], before mating[37] or during pregnancy[38]).

Drawbacks of chemical models: The efficiency of a model is based on the observation of a stable hyperglycemia throughout gestation and depends on multiple parameters such as the dose and mode of injection (venous or peritoneal route)[38]. Moreover, the efficiency varies according to the sex, with a lower metabolic sensibility in females[39]. The authors reported a sexual dimorphism in insulin sensibility with females being less sensitive to insulin than males, leading to a higher susceptibility to the rapid development of a more severe form of diabetes. Females have more islets compared to males: higher insulin rates and lower glucose rates and rat male pancreatic β cells are protected by testosterone against STZ induced apoptosis[40]. In light of these data, it is clear that we have to take into account these variations in experimental model settings.

Dietary interventions

Exposure to an adverse environment in utero programs the physiology and metabolism of the offspring permanently with long term consequences for health[41]. This is the basis of the thrifty phenotype hypothesis driven from epidemiological studies showing a strong statistical link between birth weight, further metabolic syndrome and maternal nutrition in a context of type 2 diabetes[42]. Nutrition is a key environmental factor and it has been shown that inappropriate nutrition in utero has consequences into adulthood[43]. We speak about nutritional programming as the effect that occurred long after the stressor has been removed. Beta cell development is irreversibly damaged by inadequate nutrition during critical periods of fetal development[44]. This is a fetus organogenesis adaptation to the fetal-placenta unit environment.

Others studies suggest that the post natal period, when catch up growth occurs, may be more important than the in utero period[45]. The two arguments are now taken into account to suggest that a combination of intrauterine deprivation followed by accelerated post natal growth induce the highest risk of further metabolic disease. In other words, increased risk of disease arises if there is an imbalance between pre natal and post natal nutritional uptake. This is the basis of two steps nutritional models.

We will consider four main nutritional models: high fat, low protein, high carbohydrate and two steps nutritional programming model.

High fat diet model: In humans, consuming a high fat diet model (HFD) causes an increase in body fat deposition and a decrease in insulin sensibility which leads to insulinoresistance and T2DM.

HFD diet is more representative of the eating habits of the current society in both the developing and westernized world. Diets rich in saturated fats before and during pregnancy may result in pathological manifestations in rodents similar to the human condition of GDM.

Ozaki et al[46] have shown that a diet that includes 20% fat during pregnancy changes blood pressure in the offspring. This is explained by alterations in the fatty acid profiles of the membrane of the aorta. Fatty acid oxidation inhibits both glucose oxidation and its ability to enter cells. HFD increases insulin resistance in rats by secretion of cytokines and TNF in several tissues[47].

Rats fed with HFD develop obesity, hyperinsulinemia and insulin resistance but not frank hyperglycemia and diabetes. HFD impairs the glucose signaling system of the β cell and the capacity of insulin secretion and leads to a reduction of β cell mass and an increased apoptosis[48].

Otherwise, by increasing the fat component in the diet, the levels of the other macronutrients would be also affected, leading to nutrient deficiencies.

Low protein model: Low protein diet exposure during the first 3 or 6 wk of life in rats have consequences on growth and insulin secretory response[49]. During pregnancy, these animals are unable to match or adapt to insulin request and become glucose intolerant. Authors concluded that temporary protein energy malnutrition in young rats reduces the ability to increase insulin production to meet the needs of pregnancy.

Glucose regulation is perturbed and glucose and other nutrients are transferred to fetuses in increased amounts. This stimulates pancreatic β cell growth and insulin secretion and thus the occurrence of macrosomia in the offspring. The length of diet exposure has an impact on fetus development and physiology. A diet restricted to the first week of gestation leads to hypoglycemic and low weight fetuses with a high risk of further T2DM[50]. Diet during complete gestation leads to hyperglycemic fetuses and permanent β cell alterations such as reducing β cell volume and content and consequently the capacity of insulin secretion[51].

Carbohydrate model: Infusions of glucose during pregnancy in rats lead to a transient hyperglycemia[52]. However, significant effects on fetuses need repeated injections of glucose and the window of injection is important with a positive effect restricted to early pregnancy. This finding matches clinical observations of macrosomia despite a good glycemic control in second trimester. This suggests that metabolic control in early pregnancy is an important determinant for fetal-placental growth throughout gestation.

Two steps programming nutritional models: We consider a first step of restrictive diet (food restriction, FR30) occurring at the first generation during the 1st period of life and gestation, and a second step of high caloric diet submitted to the offspring after birth and weaning. Fetuses of this second generation will develop a metabolic syndrome[53].

Pregnant genetically determined diabetes

Pregnant animals with genetically determined type 1 diabetes: The Non Obese Diabetic (NOD) mice and Bio Breeding (BB) rats develop spontaneous pre-gestational diabetes and thus represent good candidates for type 1 maternal diabetes models. In common with human diabetes, β cells are submitted to an immune attack and animals have to be treated with insulin during gestation. Stopping insulin treatment induces a loss of maternal weight, ketosis, high rate of fetus resorption, lower fetal weight and higher placental weight[54]. Therefore, BB rats are a good model for the study of perinatal morbidity, microsomia and malformations. NOD mice develop a mild form of diabetes with macrosomic fetuses and adiposity. It has been shown that maternal hyperglycemia is not the only causative factor of macrosomia, partially explained by a dysregulation of placental glucose transporters and hexokinase protein production unable to protect the fetus from hyperglycemia and hyperinsulinemia[55].

Pregnant animals with genetically determined type 2-like diabetes: There are models of obesity and diabetes affecting a common pathway, a defect in the leptin receptor (db) and a defect in the leptin gene (ob). The deficiency of leptin has consequences in multiple areas of metabolism, ingestive behavior and reproduction (insulin resistance, hyperphagia and infertility). Most of these experimental models at the homozygous state are infertile. Therefore, mating heterozygotes animals is necessary to inbreeding. Three main models exist: C57 BIKS Lepr db +and C57Bl/6j mice and Goto-Kakizaki rats. Heterozygous Lepr db+ mice are normoglycemic before pregnancy and present significant glucose intolerance restricted to the gestation period and associated with fetal macrosomia[56]. After delivery, glycemia reverts to a normal state, making Lepr db mice a good model for GDM investigation. C57BL/6j mice develop diabetes only after a high-fat and sucrose-rich diet. This model induced diabetes and obesity with hyperinsulinemia and hyperlipidemia, and females return to normal weight and glucose tolerance after gestation[57]. Goto Kakizaki (GK) is a rodent model of non obese type 2 diabetes that was produced by selective breeding of individuals with mild glucose intolerance from a non diabetic Wistar rat colony. A stable and heritable DM is obtained by selection of a diabetic line isolated by repeated breeding of normal animals[58]. These rats are not obese and not hyperinsulinemic. Offspring of GK females is exposed in utero to mild diabetes throughout gestation. Fetuses present a reduced β-cell mass associated with a lack of pancreatic reactivity to glucose. Thus, maternal mild hyperglycemia might contribute to endocrine pancreas defects in the first offspring generation.

ROLE OF THE INSULIN/INSULIN-LIKE GROWTH FACTORS SYSTEM

The insulin/IGF system is implicated in the regulation of fetal and placental growth and development. The feto-placental expression of insulin, IGF1, IGF2 and their receptors is regulated in a tissue-specific manner and can be affected by nutritional and endocrine conditions[59]. Hiden et al[60] have demonstrated that there is a spatio-temporal change in placental insulin receptor (IR) expression, suggesting a shift in the regulation of placental insulin effects from mother to fetus. In the first trimester, IR is predominantly expressed on the syncytiotrophoblast facing the maternal circulation, whereas at term, the placental endothelial cells facing the fetal circulation are the main expression site. The placental IGF1 receptor (IGF1R) is mainly expressed on the basal membrane of the syncytiotrophoblast. Hence, it is predominantly accessible for fetal IGF1 and IGF2[61]. IGF2 overexpression enhanced fetal growth, whereas targeted disruption of the fetal IGF1, IGF2 or IGF1R genes in mice resulted in retardation of fetal growth.

The IGF-binding proteins (IGFBPs) are important players in the IGF system. They are key modulators of the ligand-receptor interaction. In humans, the most prevalent IGFBPs in fetal plasma and tissue are the IGFBPs 1-4. The serum of pregnant women contains the placenta-derived IGFBP3 protease. It cleaves IGFBP3 into smaller fragments with lower affinities for IGFs. Decidual cells of the basal plate region express mRNA of all six IGFBPs in the second and third trimester, with IGFBP1 being the most abundant. In the placenta, IGFBP3 is expressed in the extravillous cytiotrophoblasts. Fetal cord blood data suggest that these binding proteins may be dysregulated by diabetes during pregnancy[62]. IGFBP3 mRNA is increased in maternal T1DM[63]. In the cord blood, increased IGFBP3 levels correlate with IGF1 levels and the incidence of macrosomia. This is consistent with the observation that IGF1 and IGFBP3 levels directly correlate with birth weight in diseased states[64].

The endocrine interaction between mother, fetus and placenta is exemplified by the effect of maternal and fetal insulin on the placenta. Maternal insulin affects placental development via receptors expressed on the microvillous membrane of the syncytiotrophoblast[21]. Fetal insulin affects gene expression in endothelial cells from placental arteries and veins, which will affect placental development. The spatio-temporal change of IR expression in the placenta allows a shift in the control of insulin regulation from the mother to the fetus. In the first trimester, maternal insulin influences the placenta by interaction with trophoblast IRs. This may in turn affect the mother by secretion of other factors as cytokines and hormones. Later, the fetus takes over control of insulin-dependent placental processes by fetal insulin interacting with placental endothelial cells.

In addition, in the first trimester, IGF1 and IGF2 produced by trophoblasts stimulate various processes that are involved in trophoblast invasion into the maternal uterus such as invasiveness, migration, MMP2 production, proliferation and MT1-MMP expression. Lower maternal IGF1 levels in T1DM may thus contribute to impaired trophoblast invasion. Hence, in GDM, transplacental amino acid transport and fetal growth may be promoted by the diabetes-associated increase in maternal concentrations of growth factors. TNF-α inhibits trophoblast invasion, whereas VEGF, leptin, insulin, IGF1 and IGF2 promote trophoblastic invasion. Changes can also be seen in the fetal circulation. However, the consequences of these changes for the fetus remain unclear.

ROLE OF LEPTIN

Maternal and fetal hyperleptinemia are well-established in diabetes and obesity. The extensive cross-talk between insulin and leptin signaling cascades may represent a major factor to the diabetes-induced placental changes. In humans, leptin levels correlate with adiposity. This hormone has different functions such as stimulation of angiogenesis, regulation of hematopoiesis and inflammatory response[65]. During gestation, maternal leptin concentration rises by 30% and the placenta becomes the primary leptin source. The leptin receptor is expressed in the syncytiotrophoblast. Hauguel-de Mouzon et al[65] have shown that leptin induces hCG production, enhances mitogenesis, stimulates amino acid uptake and increases the synthesis of extracellular matrix proteins and metalloproteinases. So leptin plays a role in the regulation of placental growth. However, hyperleptinemia contributes to other placental modifications in the case of diabetes as basement membrane thickening owing to its ability to alter collagen synthesis[66].

Hiden et al[67] have proposed a hypothetical model for diabetes-induced alterations in human placenta. Elevated maternal TNF-α and reduced IGF1 levels in T1DM may inhibit placental invasion, paralleling a higher incidence of early pregnancy loss in diabetes. Maternal hyperglycemia induces thickening of the placental basement membrane, hence reducing oxygen transport. Increased levels of placental leptin may even further contribute to the excessive extracellular matrix synthesis. Different factors elevated in the placenta (IGF2, leptin), maternal (insulin, VEGF) or fetal (insulin, IGF1, IGF2, leptin) circulations in diabetes promote proliferation and placental growth. Placental hypervascularization may be supported by elevated levels of placental IGF2 and leptin, increased fetal IGF1, IGF2, leptin, FGF2, and reduced TNF-α, as well as fetal hypoxia. These modifications in the feto-placental compartment are characteristic of GDM, overt diabetes or both[67].

CONCLUSION

Placental structure and function can be changed as a result of maternal diabetes. The nature and extent of these changes depend on the type of diabetes and on the gestational period. For a complex disease syndrome, no animal model can be expected to serve all needs of research. Although each animal model has limitations and strengths, used together in a complementary fashion, they are essential for research on the metabolic syndrome and for rapid progress in understanding the etiology and pathogenesis towards a cure. Animal models have shown convincingly that diabetes may be transmitted by intrauterine exposure to maternal hyperglycemia. Intrauterine exposure to mild hyperglycemia is associated with normal weight or macrosomic newborns and IGT at adult age, related to a deficient insulin secretion. In contrast, a newborn offspring of severely hyperglycemic mothers is microsomic and displays, at adult age, a decreased insulin action. In addition, long-term and persistent effects of gestational diabetes on glucose homeostasis in the offspring may be transmitted through generations. These data support the concept of programming of physiological metabolism in offspring by manipulating maternal nutrition. It is known that hyperglycemia is not the only causal factor. Maternal and fetal concentrations of several growth factors, hormones and cytokines are altered in diabetes and may affect the placenta and the fetal development. It is thus necessary to identify the specific biological effects and the mechanisms underlying them.

Footnotes

Peer reviewers: David J Hill, Professor, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A4V2, Canada; KVS Hari Kumar, Dr., Department of Endocrinology, Command Hospital, Central Command, Lucknow Cantt, Lucknow 226002, India

S- Editor Wu X L- Editor Roemmele A E- Editor Zheng XM

References
1.  Metzger BE, Gabbe SG, Persson B, Buchanan TA, Catalano PA, Damm P, Dyer AR, Leiva A, Hod M, Kitzmiler JL. International association of diabetes and pregnancy study groups recommendations on the diagnosis and classification of hyperglycemia in pregnancy. Diabetes Care. 2010;33:676-682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1074]  [Cited by in F6Publishing: 1078]  [Article Influence: 77.0]  [Reference Citation Analysis (1)]
2.  Boney CM, Verma A, Tucker R, Vohr BR. Metabolic syndrome in childhood: association with birth weight, maternal obesity, and gestational diabetes mellitus. Pediatrics. 2005;115:e290-e296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1590]  [Cited by in F6Publishing: 1535]  [Article Influence: 80.8]  [Reference Citation Analysis (0)]
3.  Catalano PM. Obesity and pregnancy--the propagation of a viscous cycle? J Clin Endocrinol Metab. 2003;88:3505-3506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 244]  [Cited by in F6Publishing: 255]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
4.  Simmons R. Developmental origins of adult metabolic disease. Endocrinol Metab Clin North Am. 2006;35:193-204, viii.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Grattan DR. Fetal programming from maternal obesity: eating too much for two? Endocrinology. 2008;149:5345-5347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 30]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
6.  McCance DR, Pettitt DJ, Hanson RL, Jacobsson LT, Knowler WC, Bennett PH. Birth weight and non-insulin dependent diabetes: thrifty genotype, thrifty phenotype, or surviving small baby genotype? BMJ. 1994;308:942-945.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Desoye G, Hauguel-de Mouzon S. The human placenta in gestational diabetes mellitus. The insulin and cytokine network. Diabetes Care. 2007;30 Suppl 2:S120-S126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 230]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
8.  Leushner JR, Tevaarwerk GJ, Clarson CL, Harding PG, Chance GW, Haust MD. Analysis of the collagens of diabetic placental villi. Cell Mol Biol. 1986;32:27-35.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Sutton LN, Mason DY, Redman CW. Isolation and characterization of human fetal macrophages from placenta. Clin Exp Immunol. 1989;78:437-443.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Varastehpour A, Radaelli T, Minium J, Ortega H, Herrera E, Catalano P, Hauguel-de Mouzon S. Activation of phospholipase A2 is associated with generation of placental lipid signals and fetal obesity. J Clin Endocrinol Metab. 2006;91:248-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 55]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
11.  Sherer DM, Divon MY. Prenatal ultrasonographic assessment of the ductus arteriosus: a review. Obstet Gynecol. 1996;87:630-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
12.  Taricco E, Radaelli T, Nobile de Santis MS, Cetin I. Foetal and placental weights in relation to maternal characteristics in gestational diabetes. Placenta. 2003;24:343-347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 120]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
13.  Taricco E, Radaelli T, Rossi G, Nobile de Santis MS, Bulfamante GP, Avagliano L, Cetin I. Effects of gestational diabetes on fetal oxygen and glucose levels in vivo. BJOG. 2009;116:1729-1735.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 94]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
14.  Jansson T, Ekstrand Y, Björn C, Wennergren M, Powell TL. Alterations in the activity of placental amino acid transporters in pregnancies complicated by diabetes. Diabetes. 2002;51:2214-2219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 159]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
15.  Kuruvilla AG, D'Souza SW, Glazier JD, Mahendran D, Maresh MJ, Sibley CP. Altered activity of the system A amino acid transporter in microvillous membrane vesicles from placentas of macrosomic babies born to diabetic women. J Clin Invest. 1994;94:689-695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 75]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
16.  Mandl M, Haas J, Bischof P, Nöhammer G, Desoye G. Serum-dependent effects of IGF-I and insulin on proliferation and invasion of human first trimester trophoblast cell models. Histochem Cell Biol. 2002;117:391-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 53]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
17.  Madsen H, Ditzel J. Blood-oxygen transport in first trimester of diabetic pregnancy. Acta Obstet Gynecol Scand. 1984;63:317-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
18.  Jośko J, Mazurek M. Transcription factors having impact on vascular endothelial growth factor (VEGF) gene expression in angiogenesis. Med Sci Monit. 2004;10:RA89-RA98.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Black SM, DeVol JM, Wedgwood S. Regulation of fibroblast growth factor-2 expression in pulmonary arterial smooth muscle cells involves increased reactive oxygen species generation. Am J Physiol Cell Physiol. 2008;294:C345-C354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 62]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
20.  Weiss U, Cervar M, Puerstner P, Schmut O, Haas J, Mauschitz R, Arikan G, Desoye G. Hyperglycaemia in vitro alters the proliferation and mitochondrial activity of the choriocarcinoma cell lines BeWo, JAR and JEG-3 as models for human first-trimester trophoblast. Diabetologia. 2001;44:209-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 57]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
21.  Hiden U, Glitzner E, Ivanisevic M, Djelmis J, Wadsack C, Lang U, Desoye G. MT1-MMP expression in first-trimester placental tissue is upregulated in type 1 diabetes as a result of elevated insulin and tumor necrosis factor-alpha levels. Diabetes. 2008;57:150-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 39]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
22.  Baird JD, Aerts L. Research priorities in diabetic pregnancy today: the role of animal models. Biol Neonate. 1987;51:119-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
23.  Susa JB, Boylan JM, Sehgal P, Schwartz R. Impaired insulin secretion after intravenous glucose in neonatal rhesus monkeys that had been chronically hyperinsulinemic in utero. Proc Soc Exp Biol Med. 1992;199:327-331.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Jovanovic L, Kitzmiller JL, Peterson CM. Randomized trial of human versus animal species insulin in diabetic pregnant women: improved glycemic control, not fewer antibodies to insulin, influences birthweight. Am J Obstet Gynecol. 1992;167:1325-1330.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  McNeill JH. Experimental Models of Diabetes. N Engl J Med. 2000;342:63-64.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Burguet A. Long-term outcome in children of mothers with gestational diabetes. Diabetes Metab. 2010;36:682-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
27.  Sybulski S, Maughan GB. Use of streptozotocin as diabetic agent in pregnant rats. Endocrinology. 1971;89:1537-1540.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Aerts L, Van Assche FA. Endocrine pancreas in the offspring of rats with experimentally induced diabetes. J Endocrinol. 1981;88:81-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
29.  Aerts L, Holemans K, Van Assche FA. Impaired insulin response and action in offspring of severely diabetes rats. Frontiers in Diabetes Research. Lessons from Animal Diabetes III. London: Slmith Godon 1990; 561-566.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Schaffer SW, Mozaffari MS. The neonatal STZ model of diabetes. Experimental models of diabetes. Boca Raton, FL: CRC Press 1999; 231-255.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Soulimane-Mokhtari NA, Guermouche B, Yessoufou A, Saker M, Moutairou K, Hichami A, Merzouk H, Khan NA. Modulation of lipid metabolism by n-3 polyunsaturated fatty acids in gestational diabetic rats and their macrosomic offspring. Clin Sci (Lond). 2005;109:287-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
32.  Kiss AC, Lima PH, Sinzato YK, Takaku M, Takeno MA, Rudge MV, Damasceno DC. Animal models for clinical and gestational diabetes: maternal and fetal outcomes. Diabetol Metab Syndr. 2009;1:21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
33.  Bobadilla RA, van Bree R, Vercruysse L, Pijnenborg R, Verhaeghe J. Placental effects of systemic tumour necrosis factor-α in an animal model of gestational diabetes mellitus. Placenta. 2010;31:1057-1063.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
34.  Masiello P, Broca C, Gross R, Roye M, Manteghetti M, Hillaire-Buys D, Novelli M, Ribes G. Experimental NIDDM: development of a new model in adult rats administered streptozotocin and nicotinamide. Diabetes. 1998;47:224-229.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Blondel O, Portha B. Early appearance of in vivo insulin resistance in adult streptozotocin-injected rats. Diabete Metab. 1989;15:382-387.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Tsuji K, Taminato T, Usami M, Ishida H, Kitano N, Fukumoto H, Koh G, Kurose T, Yamada Y, Yano H. Characteristic features of insulin secretion in the streptozotocin-induced NIDDM rat model. Metabolism. 1988;37:1040-1044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 63]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
37.  Caluwaerts S, Holemans K, van Bree R, Verhaeghe J, Van Assche FA. Is low-dose streptozotocin in rats an adequate model for gestational diabetes mellitus? J Soc Gynecol Investig. 2003;10:216-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
38.  López-Soldado I, Herrera E. Different diabetogenic response to moderate doses of streptozotocin in pregnant rats, and its long-term consequences in the offspring. Exp Diabesity Res. 2003;4:107-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
39.  Vital P, Larrieta E, Hiriart M. Sexual dimorphism in insulin sensitivity and susceptibility to develop diabetes in rats. J Endocrinol. 2006;190:425-432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 77]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
40.  Palomar-Morales M, Morimoto S, Mendoza-Rodríguez CA, Cerbón MA. The protective effect of testosterone on streptozotocin-induced apoptosis in beta cells is sex specific. Pancreas. 2010;39:193-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 42]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
41.  Langley-Evans SC, Gardner DS, Jackson AA. Association of disproportionate growth of fetal rats in late gestation with raised systolic blood pressure in later life. J Reprod Fertil. 1996;106:307-312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 117]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
42.  Hales CN, Barker DJ. The thrifty phenotype hypothesis. Br Med Bull. 2001;60:5-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1807]  [Cited by in F6Publishing: 1619]  [Article Influence: 70.4]  [Reference Citation Analysis (0)]
43.  Yajnik C. Interactions of perturbations in intrauterine growth and growth during childhood on the risk of adult-onset disease. Proc Nutr Soc. 2000;59:257-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 169]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
44.  Cerf ME, Chapman CS, Muller CJ, Louw J. Gestational high-fat programming impairs insulin release and reduces Pdx-1 and glucokinase immunoreactivity in neonatal Wistar rats. Metabolism. 2009;58:1787-1792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 31]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
45.  Poston L. Developmental programming and diabetes - The human experience and insight from animal models. Best Pract Res Clin Endocrinol Metab. 2010;24:541-552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 61]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
46.  Ozaki T, Nishina H, Hanson MA, Poston L. Dietary restriction in pregnant rats causes gender-related hypertension and vascular dysfunction in offspring. J Physiol. 2001;530:141-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 224]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
47.  Brown JL, Spicer MT, Spicer LJ. Effect of high-fat diet on body composition and hormone responses to glucose tolerance tests. Endocrine. 2002;19:327-332.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Liang C, DeCourcy K, Prater MR. High-saturated-fat diet induces gestational diabetes and placental vasculopathy in C57BL/6 mice. Metabolism. 2010;59:943-950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 88]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
49.  Jansson N, Pettersson J, Haafiz A, Ericsson A, Palmberg I, Tranberg M, Ganapathy V, Powell TL, Jansson T. Down-regulation of placental transport of amino acids precedes the development of intrauterine growth restriction in rats fed a low protein diet. J Physiol. 2006;576:935-946.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Rees WD, Hay SM, Buchan V, Antipatis C, Palmer RM. The effects of maternal protein restriction on the growth of the rat fetus and its amino acid supply. Br J Nutr. 1999;81:243-250.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Dumortier O, Blondeau B, Duvillié B, Reusens B, Bréant B, Remacle C. Different mechanisms operating during different critical time-windows reduce rat fetal beta cell mass due to a maternal low-protein or low-energy diet. Diabetologia. 2007;50:2495-2503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 73]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
52.  Ericsson A, Säljö K, Sjöstrand E, Jansson N, Prasad PD, Powell TL, Jansson T. Brief hyperglycaemia in the early pregnant rat increases fetal weight at term by stimulating placental growth and affecting placental nutrient transport. J Physiol. 2007;581:1323-1332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 62]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
53.  Breton C, Lukaszewski MA, Risold PY, Enache M, Guillemot J, Rivière G, Delahaye F, Lesage J, Dutriez-Casteloot I, Laborie C. Maternal prenatal undernutrition alters the response of POMC neurons to energy status variation in adult male rat offspring. Am J Physiol Endocrinol Metab. 2009;296:E462-E472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 76]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
54.  Eriksson UJ, Bone AJ, Turnbull DM, Baird JD. Timed interruption of insulin therapy in diabetic BB/E rat pregnancy: effect on maternal metabolism and fetal outcome. Acta Endocrinol (Copenh). 1989;120:800-810.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Bevier WC, Jovanovic-Peterson L, Formby B, Peterson CM. Maternal hyperglycemia is not the only cause of macrosomia: lessons learned from the nonobese diabetic mouse. Am J Perinatol. 1994;11:51-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
56.  Yamashita H, Shao J, Qiao L, Pagliassotti M, Friedman JE. Effect of spontaneous gestational diabetes on fetal and postnatal hepatic insulin resistance in Lepr(db/+) mice. Pediatr Res. 2003;53:411-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 58]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
57.  Ling ZC, Efendic S, Wibom R, Abdel-Halim SM, Ostenson CG, Landau BR, Khan A. Glucose metabolism in Goto-Kakizaki rat islets. Endocrinology. 1998;139:2670-2675.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Galli J, Li LS, Glaser A, Ostenson CG, Jiao H, Fakhrai-Rad H, Jacob HJ, Lander ES, Luthman H. Genetic analysis of non-insulin dependent diabetes mellitus in the GK rat. Nat Genet. 1996;12:31-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 189]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
59.  Gicquel C, Le Bouc Y. Hormonal regulation of fetal growth. Horm Res. 2006;65 Suppl 3:28-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 104]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
60.  Hiden U, Maier A, Bilban M, Ghaffari-Tabrizi N, Wadsack C, Lang I, Dohr G, Desoye G. Insulin control of placental gene expression shifts from mother to foetus over the course of pregnancy. Diabetologia. 2006;49:123-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 76]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
61.  Hayati AR, Cheah FC, Tan AE, Tan GC. Insulin-like growth factor-1 receptor expression in the placentae of diabetic and normal pregnancies. Early Hum Dev. 2007;83:41-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
62.  Yan-Jun L, Tsushima T, Minei S, Sanaka M, Nagashima T, Yanagisawa K, Omori Y. Insulin-like growth factors (IGFs) and IGF-binding proteins (IGFBP-1, -2 and -3) in diabetic pregnancy: relationship to macrosomia. Endocr J. 1996;43:221-231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 21]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
63.  Liu YJ, Tsushima T, Onoda N, Minei S, Sanaka M, Nagashima T, Yanagisawa K, Omori Y. Expression of messenger RNA of insulin-like growth factors (IGFs) and IGF binding proteins (IGFBP1-6) in placenta of normal and diabetic pregnancy. Endocr J. 1996;43 Suppl:S89-S91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
64.  Nelson SM, Freeman DJ, Sattar N, Lindsay RS. Role of adiponectin in matching of fetal and placental weight in mothers with type 1 diabetes. Diabetes Care. 2008;31:1123-1125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 12]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
65.  Hauguel-de Mouzon S, Lepercq J, Catalano P. The known and unknown of leptin in pregnancy. Am J Obstet Gynecol. 2006;194:1537-1545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 165]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
66.  Madani S, De Girolamo S, Muñoz DM, Li RK, Sweeney G. Direct effects of leptin on size and extracellular matrix components of human pediatric ventricular myocytes. Cardiovasc Res. 2006;69:716-725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 83]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
67.  Hiden U, Desoye G. The placenta in diabetes in pregnancy. A practical manual of Diabetes in pregnancy. 1st ed. Oxford: Wiley-Blackwell 2010; 26-33.  [PubMed]  [DOI]  [Cited in This Article: ]