Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 2(11): 100434, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34841287

ABSTRACT

miRNAs have crucial functions in many biological processes and are candidate biomarkers of disease. Here, we show that miR-216a is a conserved, pancreas-specific miRNA with important roles in pancreatic islet and acinar cells. Deletion of miR-216a in mice leads to a reduction in islet size, ß-cell mass, and insulin levels. Single-cell RNA sequencing reveals a subpopulation of ß-cells with upregulated acinar cell markers under a high-fat diet. miR-216a is induced by TGF-ß signaling, and inhibition of miR-216a increases apoptosis and decreases cell proliferation in pancreatic cells. Deletion of miR-216a in the pancreatic cancer-prone mouse line KrasG12D;Ptf1aCreER reduces the propensity of pancreatic cancer precursor lesions. Notably, circulating miR-216a levels are elevated in both mice and humans with pancreatic cancer. Collectively, our study gives insights into how ß-cell mass and acinar cell growth are modulated by a pancreas-specific miRNA and also suggests miR-216a as a potential biomarker for diagnosis of pancreatic diseases.


Subject(s)
Disease Progression , Gene Deletion , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Animals , Apoptosis , Base Sequence , Cell Line, Tumor , Cell Movement , Diet, High-Fat , Humans , Insulin Secretion , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Organ Specificity , Rats
2.
Diabetes ; 70(12): 2771-2784, 2021 12.
Article in English | MEDLINE | ID: mdl-34544729

ABSTRACT

We previously demonstrated that male, but not female, Swiss Webster mice are susceptible to diabetes, with incidence increased by early overnutrition and high-fat diet (HFD). In this study, we investigated how HFD in Swiss Webster males and females during preweaning, peripubertal, and postpubertal periods alters glucose homeostasis and diabetes susceptibility. In males, HFD throughout life resulted in the highest diabetes incidence. Notably, switching to chow postpuberty was protective against diabetes relative to switching to chow at weaning, despite the longer period of HFD exposure. Similarly, HFD throughout life in males resulted in less liver steatosis relative to mice with shorter duration of postpubertal HFD. Thus, HFD timing relative to weaning and puberty, not simply exposure length, contributes to metabolic outcomes. Females were protected from hyperglycemia regardless of length or timing of HFD. However, postpubertal HFD resulted in a high degree of hepatic steatosis and adipose fibrosis, but glucose regulation and insulin sensitivity remained unchanged. Interestingly, peri-insulitis was observed in the majority of females but was not correlated with impaired glucose regulation. Our findings reveal critical periods of HFD-induced glucose dysregulation with striking sex differences in Swiss Webster mice, highlighting the importance of careful consideration of HFD timing relative to critical developmental periods.


Subject(s)
Diet, High-Fat , Glucose/metabolism , Prenatal Nutritional Physiological Phenomena , Animals , Animals, Newborn , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Disease Susceptibility , Fatty Liver/etiology , Fatty Liver/metabolism , Female , Gestational Age , Homeostasis/physiology , Male , Mice , Mice, Inbred Strains , Overnutrition/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Sex Characteristics , Time Factors
3.
Sci Rep ; 11(1): 18394, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526546

ABSTRACT

Although innate immunity is linked to metabolic health, the effect of leptin signaling in cells from the innate immune system on glucose homeostasis has not been thoroughly investigated. We generated two mouse models using Cre-lox methodology to determine the effect of myeloid cell-specific leptin receptor (Lepr) reconstitution and Lepr knockdown on in vivo glucose metabolism. Male mice with myeloid cell-specific Lepr reconstitution (Lyz2Cre+LeprloxTB/loxTB) had better glycemic control as they aged compared to male mice with whole-body transcriptional blockade of Lepr (Lyz2Cre-LeprloxTB/loxTB). In contrast, Lyz2Cre+LeprloxTB/loxTB females only had a trend for diminished hyperglycemia after a prolonged fast. During glucose tolerance tests, Lyz2Cre+LeprloxTB/loxTB males had a mildly improved plasma glucose profile compared to Cre- controls while Lyz2Cre+LeprloxTB/loxTB females had a similar glucose excursion to their Cre- controls. Myeloid cell-specific Lepr knockdown (Lyz2Cre+Leprflox/flox) did not significantly alter body weight, blood glucose, insulin sensitivity, or glucose tolerance in males or females. Expression of the cytokine interleukin 10 (anti-inflammatory) tended to be higher in adipose tissue of male Lyz2Cre+LeprloxTB/loxTB mice (p = 0.0774) while interleukin 6 (pro-inflammatory) was lower in male Lyz2Cre+Leprflox/flox mice (p < 0.05) vs. their respective controls. In conclusion, reconstitution of Lepr in cells of myeloid lineage has beneficial effects on glucose metabolism in male mice.


Subject(s)
Glucose/metabolism , Leptin/metabolism , Myeloid Cells/metabolism , Signal Transduction , Animals , Biomarkers , Blood Glucose/metabolism , Disease Models, Animal , Disease Susceptibility , Energy Metabolism , Gene Knockdown Techniques , Homeostasis , Leptin/genetics , Male , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Mice
4.
Sci Rep ; 11(1): 14032, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34234216

ABSTRACT

Overconsumption of saturated fats promotes obesity and type 2 diabetes. Excess weight gain in early life may be particularly detrimental by promoting earlier diabetes onset and potentially by adversely affecting normal development. In the present study we investigated the effects of dietary fat composition on early overnutrition-induced body weight and glucose regulation in Swiss Webster mice, which show susceptibility to high-fat diet-induced diabetes. We compared glucose homeostasis between a high-fat lard-based (HFL) diet, high in saturated fats, and a high-fat olive oil/fish oil-based (HFO) diet, high in monounsaturated and omega-3 fats. We hypothesized that the healthier fat profile of the latter diet would improve early overnutrition-induced glucose dysregulation. However, early overnutrition HFO pups gained more weight and adiposity and had higher diabetes incidence compared to HFL. In contrast, control pups had less weight gain, adiposity, and lower diabetes incidence. Plasma metabolomics revealed reductions in various phosphatidylcholine species in early overnutrition HFO mice as well as with diabetes. These findings suggest that early overnutrition may negate any beneficial effects of a high-fat diet that favours monounsaturated and omega-3 fats over saturated fats. Thus, quantity, quality, and timing of fat intake throughout life should be considered with respect to metabolic health outcomes.


Subject(s)
Diet, High-Fat , Dietary Fats, Unsaturated/metabolism , Energy Metabolism , Fatty Acids, Omega-3/metabolism , Overnutrition/metabolism , Age Factors , Animals , Biomarkers , Diabetes Mellitus, Experimental , Glucose/metabolism , Hormones/blood , Hormones/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Phosphatidylcholines/blood
5.
Endocr Rev ; 42(1): 1-28, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33150398

ABSTRACT

The discovery of leptin was intrinsically associated with its ability to regulate body weight. However, the effects of leptin are more far-reaching and include profound glucose-lowering and anti-lipogenic effects, independent of leptin's regulation of body weight. Regulation of glucose metabolism by leptin is mediated both centrally and via peripheral tissues and is influenced by the activation status of insulin signaling pathways. Ectopic fat accumulation is diminished by both central and peripheral leptin, an effect that is beneficial in obesity-associated disorders. The magnitude of leptin action depends upon the tissue, sex, and context being examined. Peripheral tissues that are of particular relevance include the endocrine pancreas, liver, skeletal muscle, adipose tissues, immune cells, and the cardiovascular system. As a result of its potent metabolic activity, leptin is used to control hyperglycemia in patients with lipodystrophy and is being explored as an adjunct to insulin in patients with type 1 diabetes. To fully understand the role of leptin in physiology and to maximize its therapeutic potential, the mechanisms of leptin action in these tissues needs to be further explored.


Subject(s)
Glucose/metabolism , Leptin/pharmacology , Lipid Metabolism/drug effects , Animals , Body Weight/drug effects , Humans , Insulin/metabolism , Organ Specificity/drug effects
6.
Sci Rep ; 10(1): 10518, 2020 06 29.
Article in English | MEDLINE | ID: mdl-32601405

ABSTRACT

In vivo genetic manipulation is used to study the impact of gene deletion or re-expression on ß-cell function and organism physiology. Cre-LoxP is a system wherein LoxP sites flanking a gene are recognized by Cre recombinase. Cre transgenic mice are the most prevalent technology used to deliver Cre but many models have caveats of off-target recombination, impaired ß-cell function, and high cost of animal production. Inducible estrogen receptor conjugated Cre models face leaky recombination and confounding effects of tamoxifen. As an alternative, we characterize an adeno associated virus (AAV) with a rat insulin 1 promoter driving Cre recombinase (AAV8 Ins1-Cre) that is economical and rapid to implement, and has limited caveats. Intraperitoneal AAV8 Ins1-Cre produced efficient ß-cell recombination, alongside some hepatic, exocrine pancreas, α-cell, δ-cell, and hypothalamic recombination. Delivery of lower doses via the pancreatic duct retained good rates of ß-cell recombination and limited rates of off-target recombination. Unlike inducible Cre in transgenic mice, AAV8 Ins1-Cre required no tamoxifen and premature recombination was avoided. We demonstrate the utility of this technology by inducing hyperglycemia in inducible insulin knockout mice (Ins1-/-;Ins2f/f). AAV-mediated expression of Cre in ß-cells provides an effective alternative to transgenic approaches for inducible knockout studies.


Subject(s)
Dependovirus , Insulin-Secreting Cells/metabolism , Insulin/genetics , Promoter Regions, Genetic , Recombination, Genetic , Animals , Insulin/metabolism , Integrases , Mice , Mice, Transgenic
7.
Sci Rep ; 9(1): 10829, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31346189

ABSTRACT

The study of primary glucagon-secreting α-cells is hampered by their low abundance and scattered distribution in rodent pancreatic islets. We have designed a double-stranded adeno-associated virus containing a rat proglucagon promoter (700 bp) driving enhanced green fluorescent protein (AAV GCG-EGFP), to specifically identify α-cells. The administration of AAV GCG-EGFP by intraperitoneal or intraductal injection led to EGFP expression selectively in the α-cell population. AAV GCG-EGFP delivery to mice followed by islet isolation, dispersion and separation by FACS for EGFP resulted in an 86% pure population of α-cells. Furthermore, the administration of AAV GCG-EGFP at various doses to adult wild type mice did not significantly alter body weight, blood glucose, plasma insulin or glucagon levels, glucose tolerance or arginine tolerance. In vitro experiments in transgene positive α-cells demonstrated that EGFP expression did not alter the intracellular Ca2+ pattern in response to glucose or adrenaline. This approach may be useful for studying purified primary α-cells and for the in vivo delivery of other genes selectively to α-cells to further probe their function or to manipulate them for therapeutic purposes.


Subject(s)
Dependovirus , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Green Fluorescent Proteins , Animals , Blood Glucose , Body Weight/physiology , Insulin/blood , Islets of Langerhans/metabolism , Mice , Promoter Regions, Genetic , Rats
8.
Sci Rep ; 9(1): 3619, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30842440

ABSTRACT

Childhood obesity and early rapid growth increase the risk for type 2 diabetes. Such early overnutrition can be modeled in mice by reducing litter size. We investigated the effects of early overnutrition and increased dietary fat intake on ß cell function in Swiss Webster mice. On a moderate-fat diet, early overnutrition accelerated weight gain and induced hyperinsulinemia in pups. Early overnutrition males exhibited higher ß cell mass but reduced islet insulin content and Pdx1 expression. Males had a high diabetes incidence that was increased by early overnutrition, characterized by a progressive increase in insulin secretion as well as ß cell death, indicated by histological analysis and increased circulating miR-375 levels. Females maintained normoglycemia throughout life. High-fat diet (HFD) increased diabetes incidence in males, whereas low-fat diet was completely protective. This protective effect was abolished in early overnutrition males transiently exposed to HFD in early life. Although Swiss Webster mice are not known to be diabetes-prone, the high diabetes incidence suggests an underlying genetic susceptibility that can be induced by overnutrition and increased dietary fat intake in early life. Thus, the nutritional environment in early life may impact long-term ß cell function and increase diabetes risk, particularly in genetically susceptible individuals.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diet, High-Fat/adverse effects , Homeodomain Proteins/metabolism , Insulin Resistance , Insulin-Secreting Cells/pathology , Overnutrition/complications , Trans-Activators/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Female , Homeodomain Proteins/genetics , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Trans-Activators/genetics , Weight Gain
9.
Sci Rep ; 9(1): 3307, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30824713

ABSTRACT

The relative contribution of peripheral and central leptin signalling to the regulation of metabolism and the mechanisms through which leptin affects glucose homeostasis have not been fully elucidated. We generated complementary lines of mice with either leptin receptor (Lepr) knockdown or reconstitution in adipose tissues using Cre-lox methodology. Lepr knockdown mice were modestly lighter and had lower plasma insulin concentrations following an oral glucose challenge compared to controls, despite similar insulin sensitivity. We rendered male mice diabetic using streptozotocin (STZ) and found that upon prolonged leptin therapy, Lepr knockdown mice had an accelerated decrease in blood glucose compared to controls that was associated with higher plasma concentrations of leptin and leptin receptor. Mice with transcriptional blockade of Lepr (LeprloxTB/loxTB) were obese and hyperglycemic and reconstitution of Lepr in adipose tissues of LeprloxTB/loxTB mice resulted in males reaching a higher maximal body weight. Although mice with adipose tissue Lepr reconstitution had lower blood glucose levels at several ages, their plasma insulin concentrations during an oral glucose test were elevated. Thus, attenuation or restoration of Lepr in adipocytes alters the plasma insulin profile following glucose ingestion, modifies the glucose-lowering effect of prolonged leptin therapy in insulin-deficient diabetes, and may modulate weight gain.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Experimental , Gene Knockdown Techniques , Receptors, Leptin , Adipose Tissue/pathology , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Mice , Mice, Transgenic , Receptors, Leptin/genetics , Receptors, Leptin/metabolism
10.
Cell Rep ; 22(1): 163-174, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29298418

ABSTRACT

Depolarization of neuroendocrine cells results in calcium influx, which induces vesicle exocytosis and alters gene expression. These processes, along with the restoration of resting membrane potential, are energy intensive. We hypothesized that cellular mechanisms exist to maximize energy production during excitation. Here, we demonstrate that NPAS4, an immediate early basic helix-loop-helix (bHLH)-PAS transcription factor, acts to maximize energy production by suppressing hypoxia-inducible factor 1α (HIF1α). As such, knockout of Npas4 from insulin-producing ß cells results in reduced OXPHOS, loss of insulin secretion, ß cell dedifferentiation, and type 2 diabetes. NPAS4 plays a similar role in the nutrient-sensing cells of the hypothalamus. Its knockout here results in increased food intake, reduced locomotor activity, and elevated peripheral glucose production. In conclusion, NPAS4 is critical for the coordination of metabolism during the stimulation of electrically excitable cells; its loss leads to the defects in cellular metabolism that underlie the cellular dysfunction that occurs in metabolic disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypothalamus/metabolism , Neuroendocrine Cells/metabolism , Oxidative Phosphorylation , Oxygen/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Hypothalamus/cytology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Transgenic , Neuroendocrine Cells/cytology
11.
Mol Metab ; 6(9): 1052-1065, 2017 09.
Article in English | MEDLINE | ID: mdl-28951828

ABSTRACT

BACKGROUND: The hormone leptin is an important regulator of metabolic homeostasis, able to inhibit food intake and increase energy expenditure. Leptin can also independently lower blood glucose levels, particularly in hyperglycemic models of leptin or insulin deficiency. Despite significant efforts and relevance to diabetes, the mechanisms by which leptin acts to regulate blood glucose levels are not fully understood. SCOPE OF REVIEW: Here we assess literature relevant to the glucose lowering effects of leptin. Leptin receptors are widely expressed in multiple cell types, and we describe both peripheral and central effects of leptin that may be involved in lowering blood glucose. In addition, we summarize the potential clinical application of leptin in regulating glucose homeostasis. MAJOR CONCLUSIONS: Leptin exerts a plethora of metabolic effects on various tissues including suppressing production of glucagon and corticosterone, increasing glucose uptake, and inhibiting hepatic glucose output. A more in-depth understanding of the mechanisms of the glucose-lowering actions of leptin may reveal new strategies to treat metabolic disorders.


Subject(s)
Glucose/metabolism , Leptin/metabolism , Leptin/physiology , Animals , Blood Glucose/drug effects , Corticosterone/metabolism , Diabetes Mellitus/drug therapy , Eating/drug effects , Energy Metabolism , Glucagon/metabolism , Homeostasis/drug effects , Humans , Insulin/metabolism , Insulin Secretion/drug effects , Leptin/genetics , Liver/drug effects , Receptors, Leptin/metabolism
12.
Mol Metab ; 5(8): 716-724, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27656409

ABSTRACT

OBJECTIVE: Leptin reverses hyperglycemia in rodent models of type 1 diabetes (T1D). Direct application of leptin to the brain can lower blood glucose in diabetic rodents, and can activate autonomic efferents and non-shivering thermogenesis in brown adipose tissue (BAT). We investigated whether leptin reverses hyperglycemia through a mechanism that requires autonomic innervation, or uncoupling protein 1 (UCP1)-mediated thermogenesis. METHODS: To examine the role of parasympathetic and sympathetic efferents in the glucose-lowering action of leptin, mice with a subdiaphragmatic vagotomy or 6-hydroxydopamine induced chemical sympathectomy were injected with streptozotocin (STZ) to induce hyperglycemia, and subsequently leptin treated. To test whether the glucose-lowering action of leptin requires activation of UCP1-mediated thermogenesis in BAT, we administered leptin in STZ-diabetic Ucp1 knockout (Ucp1 (-/-)) mice and wildtype controls. RESULTS: Leptin ameliorated STZ-induced hyperglycemia in both intact and vagotomised mice. Similarly, mice with a partial chemical sympathectomy did not have an attenuated response to leptin-mediated glucose lowering relative to sham controls, and showed intact leptin-induced Ucp1 expression in BAT. Although leptin activated BAT thermogenesis in STZ-diabetic mice, the anti-diabetic effect of leptin was not blunted in Ucp1 (-/-) mice. CONCLUSIONS: These results suggest that leptin lowers blood glucose in insulin-deficient diabetes through a manner that does not require parasympathetic or sympathetic innervation, and thus imply that leptin lowers blood glucose through an alternative CNS-mediated mechanism or redundant target tissues. Furthermore, we conclude that the glucose lowering action of leptin is independent of UCP1-dependent thermogenesis.

13.
Endocrinology ; 157(7): 2671-85, 2016 07.
Article in English | MEDLINE | ID: mdl-27183315

ABSTRACT

Leptin signaling in the central nervous system, and particularly the arcuate hypothalamic nucleus, is important for regulating energy and glucose homeostasis. However, the roles of extra-arcuate leptin responsive neurons are less defined. In the current study, we generated mice with widespread inactivation of the long leptin receptor isoform in the central nervous system via Synapsin promoter-driven Cre (Lepr(flox/flox) Syn-cre mice). Within the hypothalamus, leptin signaling was disrupted in the lateral hypothalamic area (LHA) and ventral premammillary nucleus (PMV) but remained intact in the arcuate hypothalamic nucleus and ventromedial hypothalamic nucleus, dorsomedial hypothalamic nucleus, and nucleus of the tractus solitarius. To investigate the role of LHA/PMV neuronal leptin signaling, we examined glucose and energy homeostasis in Lepr(flox/flox) Syn-cre mice and Lepr(flox/flox) littermates under basal and diet-induced obese conditions and tested the role of LHA/PMV neurons in leptin-mediated glucose lowering in streptozotocin-induced diabetes. Lepr(flox/flox) Syn-cre mice did not have altered body weight or blood glucose levels but were hyperinsulinemic and had enhanced glucagon secretion in response to experimental hypoglycemia. Surprisingly, when placed on a high-fat diet, Lepr(flox/flox) Syn-cre mice were protected from weight gain, glucose intolerance, and diet-induced hyperinsulinemia. Peripheral leptin administration lowered blood glucose in streptozotocin-induced diabetic Lepr(flox/flox) Syn-cre mice as effectively as in Lepr(flox/flox) littermate controls. Collectively these findings suggest that leptin signaling in LHA/PMV neurons is not critical for regulating glucose levels but has an indispensable role in the regulation of insulin and glucagon levels and, may promote the development of diet-induced hyperinsulinemia and weight gain.


Subject(s)
Glucagon/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Leptin/metabolism , Obesity/metabolism , Receptors, Leptin/metabolism , Signal Transduction/physiology , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Diet, High-Fat , Hypothalamic Area, Lateral/metabolism , Insulin Secretion , Mice , Mice, Knockout , Neurons/metabolism , Receptors, Leptin/genetics , Ventromedial Hypothalamic Nucleus/metabolism
14.
J Diabetes Investig ; 7 Suppl 1: 87-93, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27186362

ABSTRACT

The gut epithelium's large surface area, its direct exposure to ingested nutrients, its vast stem cell population and its immunotolerogenic environment make it an excellent candidate for therapeutic cells to treat diabetes. Thus, several attempts have been made to coax immature gut cells to differentiate into insulin-producing cells by altering the expression patterns of specific transcription factors. Furthermore, because of similarities in enteroendocrine and pancreatic endocrine cell differentiation pathways, other approaches have used genetically engineered enteroendocrine cells to produce insulin in addition to their endogenous secreted hormones. Several studies support the utility of both of these approaches for the treatment of diabetes. Converting a patient's own gut cells into meal-regulated insulin factories in a safe and immunotolerogenic environment is an attractive approach to treat and potentially cure diabetes. Here, we review work on these approaches and indicate where we feel further advancements are required.


Subject(s)
Cell Engineering , Diabetes Mellitus/therapy , Enteroendocrine Cells/physiology , Insulin-Secreting Cells/physiology , Insulin/therapeutic use , Stem Cells/physiology , Animals , Cell Differentiation , Enteroendocrine Cells/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans Transplantation , Mice , Rats , Stem Cell Transplantation
15.
Diabetes ; 62(8): 2738-46, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23656888

ABSTRACT

Islet transplantation is an effective method to obtain long-term glycemic control for patients with type 1 diabetes, yet its widespread use is limited by an inadequate supply of donor islets. The hormone leptin has profound glucose-lowering and insulin-sensitizing action in type 1 diabetic rodent models. We hypothesized that leptin administration could reduce the dose of transplanted islets required to achieve metabolic control in a mouse model of type 1 diabetes. We first performed a leptin dose-response study in C57Bl/6 mice with streptozotocin (STZ)-induced diabetes to determine a leptin dose insufficient to reverse hyperglycemia. Subsequently, we compared the ability of suboptimal islet transplants of 50 or 125 syngeneic islets to achieve glycemic control in STZ-induced diabetic C57Bl/6 mice treated with or without this dose of leptin. The dose-response study revealed that leptin reverses STZ-induced diabetes in a dose-dependent manner. Supraphysiological leptin levels were necessary to restore euglycemia but simultaneously increased risk of hypoglycemia, and also lost efficacy after 12 days of administration. In contrast, 1 µg/day leptin only modestly reduced blood glucose but maintained efficacy throughout the study duration. We then administered 1 µg/day leptin to diabetic mice that underwent transplantation of 50 or 125 islets. Although these islet doses were insufficient to ameliorate hyperglycemia alone, coadministration of leptin with islet transplantation robustly improved control of glucose and lipid metabolism, without increasing circulating insulin levels. This study reveals that low-dose leptin administration can reduce the number of transplanted islets required to achieve metabolic control in STZ-induced diabetic mice.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Hyperglycemia/surgery , Islets of Langerhans Transplantation/methods , Islets of Langerhans/drug effects , Leptin/therapeutic use , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Glucose Tolerance Test , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Insulin/blood , Islets of Langerhans/metabolism , Leptin/metabolism , Leptin/pharmacology , Male , Mice , Mice, Inbred C57BL
16.
Diabetes ; 62(2): 490-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23048186

ABSTRACT

The melanocortin-4 receptor (MC4R) is well recognized as an important mediator of body weight homeostasis. Activation of MC4R causes dramatic weight loss in rodent models, and mutations in human are associated with obesity. This makes MC4R a logical target for pharmacological therapy for the treatment of obesity. However, previous studies in rodents and humans have observed a broad array of side effects caused by acute treatment with MC4R agonists, including increased heart rate and blood pressure. We demonstrate that treatment with a highly-selective novel MC4R agonist (BIM-22493 or RM-493) resulted in transient decreases in food intake (35%), with persistent weight loss over 8 weeks of treatment (13.5%) in a diet-induced obese nonhuman primate model. Consistent with weight loss, these animals significantly decreased adiposity and improved glucose tolerance. Importantly, we observed no increases in blood pressure or heart rate with BIM-22493 treatment. In contrast, treatment with LY2112688, an MC4R agonist previously shown to increase blood pressure and heart rate in humans, caused increases in blood pressure and heart rate, while modestly decreasing food intake. These studies demonstrate that distinct melanocortin peptide drugs can have widely different efficacies and side effects.


Subject(s)
Anti-Obesity Agents/therapeutic use , Cardiovascular System/drug effects , Diet, High-Fat/adverse effects , Insulin Resistance , Obesity/drug therapy , Peptides/therapeutic use , Receptor, Melanocortin, Type 4/agonists , Weight Loss/drug effects , alpha-MSH/analogs & derivatives , Adiposity/drug effects , Animals , Blood Pressure/drug effects , Chronic Disease , Eating/drug effects , Glucose Intolerance/drug therapy , Heart Rate/drug effects , Macaca mulatta , Male , Obesity/etiology , alpha-MSH/therapeutic use
17.
Endocrinology ; 151(11): 5226-36, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20881250

ABSTRACT

The low-density lipoprotein receptor (Ldlr) is a key molecule involved with lipid clearance. The Ldlr(-/-) mouse has been used extensively as a model for studying atherosclerosis. This study sought to characterize the energy balance phenotype of Ldlr(-/-) mice with respect to weight gain, body composition, energy expenditure (EE), glucose homeostasis, and leptin sensitivity. Adult Ldlr(-/-) mice and Ldlr(+/+) controls on a C57Bl/6J background were fed either a chow or a high-fat, high-sucrose Western-type diet (WTD) for eight wk. Physiological studies of food intake, EE, activity, insulin sensitivity, and leptin responsiveness were performed. The effect of these diet interventions on circulating leptin and on leptin gene expression was also examined. On the chow diet, Ldlr(-/-) mice had lower EE and higher activity levels relative to controls. On the WTD, Ldlr(-/-) mice gained less weight relative to Ldlr(+/+) mice, specifically gaining less fat mass. Increased thermogenesis in Ldlr(-/-) mice fed the WTD was detected. Additionally, leptin responsiveness was blunted in chow-fed Ldlr(-/-) mice, suggesting a novel role for the Ldlr pathway that extends to leptin's regulation of energy balance. In addition to its known role in lipid transport, these results demonstrate the importance of the Ldlr in energy homeostasis and suggest a direct physiological link between altered lipid transport and energy balance.


Subject(s)
Dietary Fats/metabolism , Disease Susceptibility/metabolism , Obesity/physiopathology , Receptors, LDL/metabolism , Thermogenesis/physiology , Analysis of Variance , Animals , Body Composition/physiology , Calorimetry , Diet , Eating/physiology , Energy Metabolism/physiology , Enzyme-Linked Immunosorbent Assay , Insulin/metabolism , Insulin Resistance/physiology , Leptin/metabolism , Leptin/pharmacology , Mice , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Receptors, LDL/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Endocrinology ; 151(4): 1598-610, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20194730

ABSTRACT

Childhood obesity increases the risk of adult obesity and diabetes, suggesting that early overnutrition permanently programs altered energy and glucose homeostasis. In the present studies, we used a mouse model to investigate whether early overnutrition increases susceptibility to obesity and insulin resistance in response to a high-fat diet (HFD). Litters from Swiss Webster dams were culled to three [chronic postnatal overnutrition (CPO)] or 10 (control) pups and then weaned onto standard chow at postnatal day (P) 23. At 6 wk of age, a subset of mice was placed on HFD, and glucose and insulin tolerance were examined at 16-17 wk of age. Leptin sensitivity was determined by hypothalamic phosphorylated signal transducer and activator of transcription-3 immunoreactivity at P16 and adulthood after ip leptin. CPO mice exhibited accelerated body weight gain and hyperleptinemia during the preweaning period but only a slightly heavier body weight and normal glucose tolerance in adulthood on standard chow diet. Importantly, CPO mice exhibited significant leptin resistance in the arcuate nucleus, demonstrated by reduced activation of phospho-signal transducer and activator of transcription-3, as early as P16 and throughout life, despite normalized leptin levels. In response to HFD, CPO but not control mice displayed insulin resistance in response to an insulin tolerance test. In conclusion, CPO mice exhibited early and persistent leptin resistance in the arcuate nucleus and, in response to HFD, rapid development of obesity and insulin resistance. These studies suggest that early overnutrition can permanently alter energy homeostasis and significantly increase susceptibility to obesity and insulin resistance.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Dietary Fats/metabolism , Leptin/metabolism , Overnutrition/metabolism , STAT3 Transcription Factor/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Arcuate Nucleus of Hypothalamus/drug effects , Area Under Curve , Blood Glucose , Body Weight/genetics , Eating/physiology , Energy Metabolism/physiology , Glucose Tolerance Test , Immunohistochemistry , Insulin/metabolism , Insulin Resistance/genetics , Leptin/pharmacology , Mice , Motor Activity/physiology , Overnutrition/genetics , Phosphorylation/physiology , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics
19.
J Comp Neurol ; 506(2): 194-210, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-18022952

ABSTRACT

Peptide YY (PYY), a member of the NPY superfamily of peptides, is predominantly synthesized by the colon and is thought to act on both the gut and brain to modulate energy homeostasis. Although neurons expressing PYY mRNA have also been reported in the brainstem, little is known about their physiological role and study of their projections has been problematic due to crossreactivity of PYY antibodies with NPY. In the present study we examined the localization of central PYY cell bodies in the mouse, rat, and monkey. In addition, efferent projections and afferent inputs of central PYY neurons were examined in rodents. Central PYY projections were examined by immunohistochemistry in the NPY knockout mouse, or with an NPY-preabsorbed PYY antibody in the rat to avoid any crossreactivity with NPY. In all species investigated PYY-immunoreactive (ir) cell bodies were localized exclusively to the gigantocellular reticular nucleus (Gi) of the rostral medulla. The highest density of PYY fibers was present within the solitary tract nucleus, specifically within the dorsal and lateral aspects. PYY fibers were also concentrated within the dorsal motor nucleus of the vagus and the hypoglossal nucleus. In addition, both orexin and melanin-concentrating hormone fibers made numerous close appositions with PYY cell bodies in the Gi. Collectively, the projection pattern and association with orexigenic neuropeptides suggest that brainstem PYY neurons may play a role in energy homeostasis through a coordinated effect on visceral, motor, and sympathetic output targets.


Subject(s)
Brain Stem/cytology , Neurons/metabolism , Peptide YY/metabolism , Animals , Brain Stem/metabolism , Hypothalamic Hormones/metabolism , In Situ Hybridization/methods , Intracellular Signaling Peptides and Proteins/metabolism , Macaca fascicularis , Male , Melanins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/metabolism , Neurons/cytology , Neuropeptide Y/metabolism , Neuropeptides/metabolism , Orexins , Peptide YY/genetics , Pituitary Hormones/metabolism , Rats , Rats, Sprague-Dawley
20.
Alcohol Clin Exp Res ; 31(9): 1598-610, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17760789

ABSTRACT

BACKGROUND: Rats prenatally exposed to ethanol (E) exhibit hypothalamic-pituitary-adrenal (HPA) hyperresponsiveness and changes in central HPA regulation following exposure to stressors. Whether ethanol-induced alterations in basal HPA regulation play a role in mediating HPA hyperresponsiveness remains unclear. We utilized adrenalectomy (ADX), with or without corticosterone (CORT) replacement, to investigate basal HPA function and the role of CORT in mediating ethanol-induced alterations. METHODS: Adult males and females from prenatal E, pair-fed (PF), and ad lib-fed control (C) groups were terminated at the circadian peak, 7 days following sham surgery or ADX, with or without CORT replacement. Plasma levels of CORT and adrenocorticotropin (ACTH), and mRNA levels of corticotropin-releasing hormone (CRH) and arginine vasopressin (AVP) in the paraventricular nucleus, CRH Type 1 receptor (CRH-R1) and pro-opiomelanocortin (POMC) in the anterior pituitary, and mineralocorticoid (MR) and glucocorticoid (GR) receptors in the dorsal hippocampus were determined. RESULTS: Adrenalectomy resulted in significantly greater plasma ACTH elevations in E and PF males, and parallel CRH mRNA elevations in both E and PF males and females compared with their C counterparts. In contrast, pituitary CRH-R1 mRNA levels were lower in E compared with C males, with no differences in POMC. In addition, in response to ADX, E females showed a greater MR mRNA response, and E males showed a greater GR mRNA response compared with their C counterparts, and CORT replacement was ineffective in normalizing ADX-induced alterations in ACTH levels in E and PF females, hippocampal MR mRNA levels in E males, and AVP mRNA levels in PF males and females. CONCLUSIONS: Together, these data indicate that the prenatal ethanol exposure induces HPA dysregulation under basal conditions at multiple levels of the axis, resulting in alterations in both HPA drive and feedback regulation and/or in the balance between drive and feedback. While some effects may be nutritionally mediated, it appears that the mechanisms underlying basal HPA dysregulation may differ between E and PF animals rather than occurring along a continuum of effects on the same pathway. Altered basal HPA tone may play a role in mediating the HPA hyperresponsiveness to stressors observed in E offspring.


Subject(s)
Central Nervous System Depressants/pharmacology , Corticosterone/physiology , Ethanol/pharmacology , Hypothalamus/physiology , Limbic System/physiology , Pituitary-Adrenal System/physiology , Prenatal Exposure Delayed Effects/physiopathology , Adrenalectomy , Adrenocorticotropic Hormone/blood , Animals , Central Nervous System Depressants/adverse effects , Corticosterone/blood , Corticotropin-Releasing Hormone/metabolism , Ethanol/adverse effects , Female , Hypothalamus/drug effects , Limbic System/drug effects , Male , Mineralocorticoids/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary Gland/metabolism , Pituitary-Adrenal System/drug effects , Pregnancy , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Glucocorticoid/metabolism , Vasopressins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...