Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Postepy Hig Med Dosw (Online) ; 68: 899-911, 2014.
Article in Polish | MEDLINE | ID: mdl-24988610

ABSTRACT

Growth and development in utero is a complex and dynamic process that requires interaction between the mother organism and the fetus. The delivery of macro--and micronutrients, oxygen and endocrine signals has crucial importance for providing a high level of proliferation, growth and differentiation of cells, and a disruption in food intake not only has an influence on the growth of the fetus, but also has negative consequences for the offspring's health in the future. Diseases that traditionally are linked to inappropriate life style of adults, such as type 2 diabetes, obesity, and arterial hypertension, can be "programmed" in the early stage of life and the disturbed growth of the fetus leads to the symptoms of the metabolic syndrome. The structural changes of some organs, such as the brain, pancreas and kidney, modifications of the signaling and metabolic pathways in skeletal muscles and in fatty tissue, epigenetic mechanisms and mitochondrial dysfunction are the basis of the metabolic disruptions. The programming of the metabolic disturbances is connected with the disruption in the intrauterine environment experienced in the early and late gestation period. It causes the changes in deposition of triglycerides, activation of the hormonal "stress axis" and disturbances in the offspring's glucose tolerance. The present review summarizes experimental results that led to the identification of the above-mentioned links and it underlines the role of animal models in the studies of this important concept.


Subject(s)
Disease Models, Animal , Fetal Diseases/genetics , Fetal Diseases/metabolism , Metabolic Diseases/embryology , Metabolic Diseases/metabolism , Pregnancy Complications/metabolism , Prenatal Exposure Delayed Effects/metabolism , Anemia/metabolism , Animals , Brain/embryology , Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/metabolism , Diabetes, Gestational/metabolism , Epigenesis, Genetic , Female , Hypertension/embryology , Hypertension/metabolism , Kidney/embryology , Metabolic Diseases/genetics , Metabolic Syndrome/embryology , Metabolic Syndrome/metabolism , Obesity/embryology , Obesity/metabolism , Pancreas/embryology , Pregnancy
2.
Diabetologia ; 56(1): 194-203, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23064288

ABSTRACT

AIMS/HYPOTHESIS: We used the GK/Par rat, a spontaneous model of type 2 diabetes with early defective beta cell neogenesis, to determine whether the development of GK/Par offspring in a non-diabetic intrauterine/postnatal environment would prevent the alteration of fetal beta cell mass (BCM) and ultimately decrease the risk of diabetes later in adult life. METHODS: We used an embryo-transfer approach, with fertilised GK/Par ovocytes (oGK) being transferred into pregnant Wistar (W) or GK/Par females (pW and pGK). Offspring were phenotyped at fetal age E18.5 and at 10 weeks of age, for BCM, expression of genes of pancreatic progenitor cell regulators (Igf2, Igf1r, Sox9, Pdx1 and Ngn3), glucose tolerance and insulin secretion. RESULTS: (1) Alterations in neogenesis markers/regulators and BCM were as severe in the oGK/pW E18.5 fetuses as in the oGK/pGK group. (2) Adult offspring from oGK transfers into GK (oGK/pGK/sGK) had the expected diabetic phenotype compared with unmanipulated GK rats. (3) Adult offspring from oGK reared in pW mothers and milked by GK foster mothers had reduced BCM, basal hyperglycaemia, glucose intolerance and low insulin, to an extent similar to that of oGK/pGK/sGK offspring. (4) In adult offspring from oGK transferred into pW mothers and milked by their W mothers (oGK/pW/sW), the phenotype was similar to that in oGK/pGK/sGK or oGK/pW/sGK offspring. CONCLUSIONS/INTERPRETATION: These data support the conclusion that early BCM alteration and subsequent diabetes risk in the GK/Par model are not removed despite normalisation of the prenatal and postnatal environments, either isolated or combined.


Subject(s)
Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Insulin-Secreting Cells/pathology , Lactation , Pancreas/embryology , Pancreas/pathology , Animals , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Embryo Transfer , Female , Fetal Development , Glucose Intolerance/embryology , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Insulin/metabolism , Insulin Secretion , Insulin-Like Growth Factor II/metabolism , Insulin-Secreting Cells/metabolism , Male , Pancreas/metabolism , Pregnancy , Pregnancy in Diabetics/physiopathology , Rats , Rats, Inbred Strains , Rats, Wistar , Receptor, IGF Type 1/metabolism , SOX9 Transcription Factor/metabolism
3.
Clin Obstet Gynecol ; 56(3): 591-601, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23703224

ABSTRACT

Altered maternal nutrition and metabolism, restricted utero-placental blood flow, and other perturbations in the maternal compartment may disturb critical periods of fetal development resulting in increased susceptibility to develop disease in childhood and adult life. In response to these perturbations, changes in placental structure and function occur, which influence the supply of nutrients, oxygen, and methyl donors and alter the secretion of hormones and other signaling molecules into the fetal circulation. Thus, the placenta plays a critical role in modulating maternal-fetal resource allocation, thereby affecting fetal growth and the long-term health of the offspring.


Subject(s)
Fetal Development/physiology , Fetal Nutrition Disorders/metabolism , Maternal-Fetal Exchange/physiology , Placenta/physiopathology , Placental Insufficiency/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Cardiovascular Diseases/embryology , Diabetes Mellitus, Type 2/embryology , Female , Humans , Maternal Nutritional Physiological Phenomena , Placenta/pathology , Placental Circulation , Pregnancy , Prenatal Nutritional Physiological Phenomena , Signal Transduction , Trophoblasts/metabolism
4.
Clin Obstet Gynecol ; 56(3): 511-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23787713

ABSTRACT

There is a new "developmental" model for the origins of a wide range of chronic diseases. Under this model the causes to be identified are linked to normal variations in fetoplacental development. These variations are thought to lead to variations in the supply of nutrients to the baby that permanently alter gene expression, a process known as "programming." According to the developmental model variations in the processes of development program the function of a few key systems that are linked to disease, including the immune system, antioxidant defenses, inflammatory responses, and the number and quality of stem cells.


Subject(s)
Coronary Disease/genetics , Diabetes Mellitus, Type 2/genetics , Fetal Development/genetics , Prenatal Exposure Delayed Effects/genetics , Birth Weight , Coronary Disease/embryology , Diabetes Mellitus, Type 2/embryology , Epigenesis, Genetic , Female , Fetal Development/physiology , Humans , Maternal Nutritional Physiological Phenomena/genetics , Maternal Nutritional Physiological Phenomena/physiology , Placental Insufficiency , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Nutritional Physiological Phenomena/genetics , Prenatal Nutritional Physiological Phenomena/physiology
5.
Clin Obstet Gynecol ; 56(3): 520-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23820120

ABSTRACT

Intrauterine growth restriction (IUGR) impairs insulin secretion in humans and in animal models of IUGR. Several underlying mechanisms have been implicated, including decreased expression of molecular regulators of ß-cell mass and function, in some cases shown to be due to epigenetic changes initiated by an adverse fetal environment. Alterations in cell cycle progression contribute to loss of ß-cell mass, whereas decreased islet vascularity and mitochondrial dysfunction impair ß-cell function in IUGR rodents. Animal models of IUGR sharing similar insulin secretion outcomes as the IUGR human are allowing underlying mechanisms to be identified. This review will focus on models of uteroplacental insufficiency.


Subject(s)
Fetal Development/physiology , Fetal Growth Retardation/genetics , Fetus/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic , Female , Fetal Growth Retardation/physiopathology , Fetus/physiopathology , Humans , Insulin Secretion , Insulin-Secreting Cells/physiology , Pregnancy
6.
Clin Obstet Gynecol ; 56(3): 633-41, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23820121

ABSTRACT

A greater proportion of women of reproductive age are now overweight or obese. Gestational diabetes mellitus and maternal obesity are associated with long-term adverse consequences in the offspring and subsequent generations, and are important drivers of the escalating global burden of diabetes and cardiovascular disease. We review the evidence linking gestational diabetes mellitus and maternal obesity with a greater risk of metabolic compromise in the offspring. We use an evolutionary perspective to elucidate the origins of gestational diabetes. Focusing efforts on maternal health is an important approach to combating the growing burden of diabetes and other noncommunicable diseases.


Subject(s)
Cardiovascular Diseases/embryology , Diabetes Mellitus, Type 2/embryology , Diabetes, Gestational/metabolism , Hyperglycemia/metabolism , Obesity/metabolism , Biological Evolution , Birth Weight , Cardiovascular Diseases/genetics , Diabetes Mellitus, Type 2/genetics , Female , Humans , Infant, Newborn , Pregnancy , Pregnancy Complications/metabolism , Prenatal Exposure Delayed Effects , Risk Factors
7.
Klin Med (Mosk) ; 90(11): 30-4, 2012.
Article in Russian | MEDLINE | ID: mdl-23516850

ABSTRACT

The aim of this work was to estimate the relative risk (RR) of type 2 diabetes mellitus (DM2) and cardiovascular diseases, total and cardiovascular mortality in patients with disturbances of carbohydrate metabolism revealed in the prospective study carried out in 2009 that included patients found to have disturbances of carbohydrate metabolism in 2006. We analysed the 3-year risk of development of type 2 diabetes mellitus, total and cardiovascular mortality. RR of DM2 was significantly increased in association with practically all early disturbances of carbohydrate metabolism. The most unfavourable combination is fasting glycemia and impaired glucose tolerance. Within 3 years after its determination, 33.3% of the patients developed DM2 while RR of DM2 increased 11-fold. Newly diagnosed DM2 increased RR of total mortality by 2.3 times. Fasting glycemia during 3 years increased RR of cardiovascular mortality by 3.2 times. Results of the study suggest the necessity of not only timely diagnosis of fasting glycemia and impaired glucose tolerance but also further monitoring and correction of carbohydrate metabolism in patients with this pathology as well as of the elaboration and implementation of a comprehensive program for the screening of disturbed carbohydrate metabolism in high-risk groups.


Subject(s)
Carbohydrate Metabolism , Cardiovascular Diseases/etiology , Diabetes Mellitus, Type 2/etiology , Acute Disease , Adult , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/metabolism , Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Incidence , Male , Middle Aged , Retrospective Studies , Risk Factors , Russia/epidemiology , Time Factors
8.
Sci Rep ; 11(1): 11866, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34088949

ABSTRACT

Meal ingestion elicits a variety of neuronal, physiological and hormonal responses that differ in healthy, obese or diabetic individuals. The mixed meal tolerance test (MMTT) is a well-established method to evaluate pancreatic ß-cell reserve and glucose homeostasis in both preclinical and clinical research in response to calorically defined meal. Nonhuman primates (NHPs) are highly valuable for diabetic research as they can naturally develop type 2 diabetes mellitus (T2DM) in a way similar to the onset and progression of human T2DM. The purpose of this study was to investigate the reproducibility and effects of a MMTT containing acetaminophen on plasma glucose, insulin, C-peptide, incretin hormones, lipids, acetaminophen appearance (a surrogate marker for gastric emptying) in 16 conscious obese cynomolgus monkeys (Macaca fascicularis). Plasma insulin, C-peptide, TG, aGLP-1, tGIP, PYY and acetaminophen significantly increased after meal/acetaminophen administration. A subsequent study in 6 animals showed that the changes of plasma glucose, insulin, C-peptide, lipids and acetaminophen were reproducible. There were no significant differences in responses to the MMTT among the obese NHPs with (n = 11) or without (n = 5) hyperglycemia. Our results demonstrate that mixed meal administration induces significant secretion of several incretins which are critical for maintaining glucose homeostasis. In addition, the responses to the MMTTs are reproducible in NHPs, which is important when the MMTT is used for evaluating post-meal glucose homeostasis in research.


Subject(s)
Animal Feed , Blood Glucose/metabolism , Gastric Emptying , Glucose Tolerance Test , Insulin-Secreting Cells/metabolism , Lipids/chemistry , Acetaminophen , Animals , C-Peptide/blood , Diabetes Mellitus, Type 2/embryology , Gastric Inhibitory Polypeptide/blood , Gastrointestinal Hormones , Glucose , Homeostasis , Incretins/pharmacology , Insulin/metabolism , Macaca fascicularis , Male , Reproducibility of Results
9.
J Intern Med ; 264(3): 205-23, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18452519

ABSTRACT

Several researchers have argued that observed associations between birth weight and cardiovascular diseases, and type 2 diabetes, may be confounded by familial (shared environmental and genetic) factors. However, most studies have found that shared environmental factors, including socio-economic factors, do not influence the foetal origins of adult diseases. Results from two twin studies suggest that genetic factors may be of importance for the association between birth weight and risks of coronary heart disease, but findings from intergenerational studies are not consistent with genetic confounding. More studies have assessed the importance of genetic factors with respect to risk factors of coronary heart, including raised blood pressure and lipid levels. Recent findings suggest that the association between birth weight and hypertension is independent of genetic factors. In contrast, recent twin and intergenerational studies favour the hypothesis that the association between birth weight and risk of type 2 diabetes is confounded by genetic factors.


Subject(s)
Cardiovascular Diseases/embryology , Diabetes Mellitus, Type 2/embryology , Birth Weight/genetics , Cardiovascular Diseases/genetics , Confounding Factors, Epidemiologic , Diabetes Mellitus, Type 2/genetics , Environment , Female , Genetic Predisposition to Disease , Humans , Pregnancy , Prenatal Exposure Delayed Effects , Risk Factors , Socioeconomic Factors
10.
Toxicol Pathol ; 36(4): 529-51, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18467681

ABSTRACT

Type 2 diabetes mellitus (TTDM) is characterized by progressive loss of glucose control through multifactorial mechanisms. The search for an understanding of TTDM has relied on animal models since the realization of the importance of the pancreas in controlling plasma glucose concentration. Rodent models of TTDM are developed to express hyperglycemia and not islet degeneration per se. Degeneration of the islets of Langerhans with beta-cell loss is secondary to insulin resistance and is regarded as the more important lesion. Despite this, differences between models are seen in the development and progression of islet degeneration. Assessing the differences between the models is important to appreciate the various aspects of TTDM and understand their advantages as well as their deficiencies. Relevant animal models of TTDM provide opportunities to investigate important physiological and cell biological processes that may ultimately lead to development of targeted therapies. This article reviews the importance, advantages, and limitations of rodent models of TTDM in relation to the histopathological changes that characterize islet degeneration. Pathophysiological mechanisms that contribute to islet degeneration are also discussed and are placed into the context of changes in islet histological appearances.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Islets of Langerhans/pathology , Rodentia , Animals , Diabetes Mellitus, Experimental/embryology , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression , Humans , Insulin Resistance , Islets of Langerhans/embryology , Streptozocin , Transgenes
11.
J Clin Invest ; 101(4): 899-904, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9466985

ABSTRACT

The Goto-Kakisaki (GK) rat is a genetic model of non-insulin-dependent diabetes. At 21.5 d of age we found that GK fetuses had an increased plasma glucose concentration, a decreased plasma insulin level, and a reduced pancreatic beta cell mass. To investigate the beta cell function during fetal life we used a hyperglycemic clamp protocol applied to the mothers, which allowed us to obtain a steady-state hyperglycemia in the corresponding fetuses. At variance, with Wistar (W) fetuses, plasma insulin concentration in GK fetuses did not rise in response to hyperglycemia. In contrast, GK fetal pancreas released insulin in response to glucose in vitro to the same extent as W fetal pancreas. Such a discrepancy between the in vivo and in vitro results suggests that the lack of pancreatic reactivity to glucose as seen in vivo is extrinsic to the fetal GK beta cell. Finally, the importance of gestational hyperglycemia was investigated by performing crosses between GK and W rats. Fetuses issued from crosses between W mother and GK father or GK mother and W father had a beta cell mass close to normal values and were still able to increase their plasma insulin levels in response to hyperglycemia in vivo. Our data suggest that hyperglycemia in utero does not influence the severity of the decrease of the beta cell mass or the lack of the insulin secretory response to glucose in the fetal GK rat. Moreover they indicate that conjunction of GK genes originating from both parents is necessary in order for these defects to be fully expressed.


Subject(s)
Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/genetics , Islets of Langerhans/embryology , Pancreas/embryology , Pregnancy in Diabetics , Animals , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Disease Models, Animal , Female , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/physiopathology , Male , Organ Size , Pancreas/cytology , Pancreas/metabolism , Pancreas/physiopathology , Pregnancy , Pregnancy in Diabetics/blood , Rats , Rats, Wistar
12.
Diabetes ; 54(3): 702-11, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15734846

ABSTRACT

Low birth weight (LBW) is an important risk factor for type 2 diabetes. We have developed a mouse model of LBW resulting from undernutrition during pregnancy. Restriction of maternal food intake from day 12.5 to 18.5 of pregnancy results in a 23% decrease in birth weight (P < 0.001), with normalization after birth. However, offspring of undernutrition pregnancies develop progressive, severe glucose intolerance by 6 months. To identify early defects that are responsible for this phenotype, we analyzed mice of undernutrition pregnancies at age 2 months, before the onset of glucose intolerance. Fed insulin levels were 1.7-fold higher in mice of undernutrition pregnancies (P = 0.01 vs. controls). However, insulin sensitivity was normal in mice of undernutrition pregnancies, with normal insulin tolerance, insulin-stimulated glucose disposal, and isolated muscle and adipose glucose uptake. Although insulin clearance was mildly impaired in mice of undernutrition pregnancies, the major metabolic phenotype in young mice of undernutrition pregnancies was dysregulation of insulin secretion. Despite normal beta-cell mass, islets from normoglycemic mice of undernutrition pregnancies showed basal hypersecretion of insulin, complete lack of responsiveness to glucose, and a 2.5-fold increase in hexokinase activity. Taken together, these data suggest that, at least in mice, primary beta-cell dysfunction may play a significant role in the pathogenesis of LBW-associated type 2 diabetes.


Subject(s)
Birth Weight , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Islets of Langerhans/physiopathology , Adipocytes/metabolism , Aging , Animal Nutritional Physiological Phenomena , Animals , Diabetes Mellitus, Type 2/embryology , Female , Glucose/metabolism , Humans , Infant, Low Birth Weight , Infant, Newborn , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Inbred ICR , Muscle, Skeletal/metabolism , Pregnancy , Risk Factors
13.
Free Radic Biol Med ; 40(6): 917-22, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16540386

ABSTRACT

The "thrifty phenotype" hypothesis proposes that the fetus adapts to an adverse intrauterine milieu by optimizing the use of a reduced nutrient supply to ensure survival, but, by favoring the development of certain organs over that of others, this leads to persistent alterations in the growth and function of developing tissues. This concept has been somewhat controversial; however, recent epidemiological, clinical, and animal studies provide support for the developmental origins of disease hypothesis. Underlying mechanisms include reprogramming of the hypothalamic-pituitary-adrenal axis, islet development, and insulin signaling pathways. Emerging data suggest that oxidative stress and mitochondrial dysfunction may also play critical roles in the pathogenesis of type 2 diabetes in individuals who were growth retarded at birth.


Subject(s)
Diabetes Mellitus, Type 2/embryology , Oxidative Stress/physiology , Animals , Disease Models, Animal , Fetal Growth Retardation/physiopathology , Humans , Mitochondria/physiology
14.
Physiol Behav ; 88(3): 234-43, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16782139

ABSTRACT

Fetal programming is gaining momentum as a highly documented phenomenon which links poor early growth to adult disease. It is backed up by large cohorts in epidemiological studies worldwide and has been tested in various animal models. The root causes of programming link closely with maternal condition during pregnancy, and therefore the fetal environment. Suboptimal fetal environments due to poor or inadequate nutrition, infection, anemia, hypertension, inflammation, gestational diabetes or hypoxia in the mother expose the fetus to hormonal, growth factor, cytokine or adipokine cues. These in turn act to alter metabolic, immune system, vascular, hemodynamics, renal, growth and mitochondrial parameters respectively and most evidently in the later stages of life where they impact on the individual as poor glucose homeostasis, insulin resistance, type 2 diabetes, hypertension, cardiovascular disease, obesity and heart disease. These events are compounded by over-nutrition or lifestyle choices which are in conflict with the programming of the fetus. We and others have utilised various species to test the early life programming hypothesis and to identify key molecular mechanisms. With parallel studies of human cohorts, these molecular markers can be validated as realistic targets for intervention.


Subject(s)
Diabetes Mellitus, Type 2/etiology , Fetal Development/physiology , Metabolic Syndrome/etiology , Obesity/etiology , Prenatal Exposure Delayed Effects , Stress, Physiological/complications , Adaptation, Physiological , Animals , Diabetes Mellitus, Type 2/embryology , Female , Fetal Growth Retardation/physiopathology , Humans , Metabolic Syndrome/embryology , Models, Animal , Obesity/embryology , Phenotype , Pregnancy , Pregnancy Complications , Prenatal Nutritional Physiological Phenomena
15.
Mol Cell Endocrinol ; 435: 78-84, 2016 11 05.
Article in English | MEDLINE | ID: mdl-26820125

ABSTRACT

Placental insufficiency leads to intrauterine growth restriction (IUGR) and a lifelong risk of developing type 2 diabetes. Impaired islet development in the growth restricted fetus, including decreased ß-cell replication, mass, and insulin secretion, is strongly implicated in the pathogenesis of later life type 2 diabetes. Currently, standard medical management of a woman with a pregnancy complicated by placental insufficiency and fetal IUGR is increased fetal surveillance and indicated preterm delivery. This leads to the dual complications of IUGR and preterm birth - both of which may increase the lifelong risk for type 2 diabetes. In order to develop therapeutic interventions in IUGR pregnancies complicated by placental insufficiency and decrease the risk of later development of type 2 diabetes in the offspring, the mechanisms responsible for impaired islet development in these cases must be determined. This review focuses on current investigations testing the hypothesis that decreased nutrient supply to the IUGR fetus inhibits an intra-islet hepatocyte growth factor - vascular endothelial growth factor A (HGF - VEGFA) feed forward signaling pathway and that this is responsible for developmental islet defects.


Subject(s)
Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/epidemiology , Fetal Growth Retardation/physiopathology , Hepatocyte Growth Factor/metabolism , Islets of Langerhans/metabolism , Vascular Endothelial Growth Factor A/metabolism , Female , Humans , Infant, Newborn , Islets of Langerhans/pathology , Pregnancy , Prevalence , Risk Factors , Signal Transduction
16.
Diabetes ; 51(2): 392-7, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11812746

ABSTRACT

At late fetal age (21.5 days postcoitum [dpc]), GK rats present a severely reduced beta-cell mass compared with Wistar rats. This anomaly largely antedates the onset of hyperglycemia in GK rats. Thus, the beta-cell mass deficit could represent the primary defect leading to type 2 diabetes in the adult. The aim of this work was to investigate, in GK fetuses at the end of fetal age (21.5 dpc), whether impaired availability of growth factors such as insulin, growth hormone, and IGFs and their IGF binding proteins (IGFBPs) could be instrumental in this anomaly. Although it confirms that GK fetuses are hypoinsulinemic despite enhanced plasma glucose level due to maternal hyperglycemia, the present study shows for the first time that IGF-2 expression in the liver and pancreas and IGF-2 serum levels are decreased in GK fetuses. Serum level as well as liver and pancreatic mRNA expression of IGFBP-2 were found to be normal in GK fetuses, whereas serum level and liver mRNA expression of IGFBP-1 were increased. Finally, we found that the maximal beta-cell mitogenic response to IGFs in vitro is kept intact, therefore suggesting that the direct biological action of IGFs on fetal GK beta-cells is not grossly impaired. In conclusion, in GK fetuses at 21.5 dpc, the defective IGF-2 production appears to be an early landmark in the pathological sequence leading to retardation of beta-cell growth in the fetal GK rat.


Subject(s)
Diabetes Mellitus, Type 2/embryology , Insulin-Like Growth Factor II/metabolism , Animals , Disease Models, Animal , Fetal Blood/metabolism , Fetus/physiology , Gestational Age , In Vitro Techniques , Insulin-Like Growth Factor Binding Proteins/blood , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/physiology , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/physiology , Liver/metabolism , Mitosis/physiology , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Rats, Wistar
17.
Nutrients ; 7(9): 8090-111, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26402696

ABSTRACT

The global pandemic of obesity and type 2 diabetes is often causally linked to changes in diet and lifestyle; namely increased intake of calorically dense foods and concomitant reductions in physical activity. Epidemiological studies in humans and controlled animal intervention studies have now shown that nutritional programming in early periods of life is a phenomenon that affects metabolic and physiological functions throughout life. This link is conceptualised as the developmental programming hypothesis whereby environmental influences during critical periods of developmental plasticity can elicit lifelong effects on the health and well-being of the offspring. The mechanisms by which early environmental insults can have long-term effects on offspring remain poorly defined. However there is evidence from intervention studies which indicate altered wiring of the hypothalamic circuits that regulate energy balance and epigenetic effects including altered DNA methylation of key adipokines including leptin. Studies that elucidate the mechanisms behind these associations will have a positive impact on the health of future populations and adopting a life course perspective will allow identification of phenotype and markers of risk earlier, with the possibility of nutritional and other lifestyle interventions that have obvious implications for prevention of non-communicable diseases.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Energy Metabolism , Infant Nutritional Physiological Phenomena , Maternal Nutritional Physiological Phenomena , Obesity/metabolism , Prenatal Exposure Delayed Effects , Age Factors , Animals , Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Epigenesis, Genetic , Female , Gene-Environment Interaction , Genetic Predisposition to Disease , Humans , Infant , Infant, Newborn , Nutritional Status , Obesity/embryology , Obesity/epidemiology , Obesity/genetics , Phenotype , Pregnancy , Prognosis , Risk Assessment , Risk Factors
18.
J Endocrinol ; 224(1): 1-15, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25326603

ABSTRACT

Cardiomyopathies-associated metabolic pathologies (e.g., type 2 diabetes and insulin resistance) are a leading cause of mortality. It is known that the association between these pathologies works in both directions, for which heart failure can lead to metabolic derangements such as insulin resistance. This intricate crosstalk exemplifies the importance of a fine coordination between one of the most energy-demanding organs and an equilibrated carbohydrate metabolism. In this light, to assist in the understanding of the role of insulin-regulated glucose transporters (GLUTs) and the development of cardiomyopathies, we have developed a model for glut12 deficiency in zebrafish. GLUT12 is a novel insulin-regulated GLUT expressed in the main insulin-sensitive tissues, such as cardiac muscle, skeletal muscle, and adipose tissue. In this study, we show that glut12 knockdown impacts the development of the embryonic heart resulting in abnormal valve formation. Moreover, glut12-deficient embryos also exhibited poor glycemic control. Glucose measurements showed that these larvae were hyperglycemic and resistant to insulin administration. Transcriptome analysis demonstrated that a number of genes known to be important in cardiac development and function as well as metabolic mediators were dysregulated in these larvae. These results indicate that glut12 is an essential GLUT in the heart where the reduction in glucose uptake due to glut12 deficiency leads to heart failure presumably due to the lack of glucose as energy substrate. In addition, the diabetic phenotype displayed by these larvae after glut12 abrogation highlights the importance of this GLUT during early developmental stages.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetic Cardiomyopathies/genetics , Disease Models, Animal , Glucose Transport Proteins, Facilitative/genetics , Heart Failure/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Animals, Genetically Modified , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/embryology , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/embryology , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Glucose Transport Proteins, Facilitative/deficiency , Heart Failure/pathology , Insulin/pharmacology , Metformin/pharmacology , Phenotype , Zebrafish/genetics , Zebrafish Proteins/deficiency
19.
Endocrinology ; 144(5): 1703-11, 2003 May.
Article in English | MEDLINE | ID: mdl-12697674

ABSTRACT

GLUT8 is a novel glucose transporter protein that is widely distributed in tissues including liver, a central organ of regulation of glucose homeostasis. The purpose of the current study was to investigate expression and regulation of hepatic GLUT8 mRNA and protein. Therefore, Northern and immunoblot analysis, semiquantitative RT-PCR, and immunofluorescence microscopy were performed using mouse livers at different stages of embryonic and postnatal development and in type 1 (streprozotocin treated) and type 2 (GLUT4 heterozygous) diabetes. GLUT8 mRNA and protein expression in embryonic liver was differentially regulated depending on the prenatal and postnatal developmental stage of the mice. Immunofluorescence microscopy of liver from wild-type mice demonstrated the highest levels of GLUT8 protein in perivenous hepatocytes pointing to its role in regulation of glycolytic flux. In diabetic scenarios, GLUT8 mRNA levels were correlated with circulating insulin; specifically, GLUT8 mRNA decreased in a type 1 diabetes model and increased in a type 2 diabetes model, suggesting a regulatory role for insulin in GLUT8 mRNA expression. While up-regulation of GLUT8 protein occurred in both models of diabetes, only in streptozotocin diabetic livers was GLUT8 zonation altered. These data demonstrate that GLUT8 mRNA and protein are differentially regulated in liver in response to physiologic and pathologic (diabetes) milieu and suggests that GLUT8 is intimately linked to glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Liver/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Animals , Animals, Newborn/metabolism , Diabetes Mellitus, Experimental/embryology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/embryology , Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/genetics , Female , Fetus/metabolism , Gene Deletion , Glucose Transport Proteins, Facilitative , Glucose Transporter Type 4 , Heterozygote , Immunologic Techniques , Mice , Mice, Inbred CBA , Monosaccharide Transport Proteins/genetics , RNA, Messenger/metabolism , Reference Values
20.
Int J Epidemiol ; 33(5): 948-53; discussion 953-4, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15166205

ABSTRACT

BACKGROUND: To investigate the association between low birthweight and diabetes in a population-based Swedish twin sample. Method A cohort of 11 162 same-sexed Swedish twins born between 1906 and 1958 was used in order to investigate the risk of developing Type 2 diabetes between and within twin pairs by utilizing random effects linear models. RESULTS: Between pairs there was a significant increase in risk of developing Type 2 diabetes for a 1-kg increase in their mean birthweight (odds ratio [OR] = 2.13; P < 0.01), adjusted for age, sex, body mass index (BMI), and smoking status. The corresponding risk within pair was 2.03 (P = 0.07) for monozygotic twins and 1.15 (P = 0.71) for dizygotic twins. The test of the heterogeneity of the within and between effects showed no significant difference between the estimates. CONCLUSIONS: The study suggests that reduced fetal growth increase the risk of Type 2 diabetes due to an in utero programming effect possibly caused by intrauterine malnutrition. However, it does not exclude the possibility of a common genetic mechanism.


Subject(s)
Diabetes Mellitus, Type 2/embryology , Diseases in Twins/embryology , Infant, Low Birth Weight , Adult , Aged , Birth Weight , Cohort Studies , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/etiology , Diseases in Twins/epidemiology , Diseases in Twins/etiology , Female , Humans , Infant, Newborn , Linear Models , Male , Middle Aged , Pregnancy , Prenatal Exposure Delayed Effects , Risk Assessment/methods , Sweden/epidemiology , Twins, Dizygotic , Twins, Monozygotic
SELECTION OF CITATIONS
SEARCH DETAIL