Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell Metab ; 5(3): 181-94, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17339026

RESUMO

Despite high leptin levels, most obese humans and rodents lack responsiveness to its appetite-suppressing effects. We demonstrate that leptin modulates NPY/AgRP and alpha-MSH secretion from the ARH of lean mice. High-fat diet-induced obese (DIO) mice have normal ObRb levels and increased SOCS-3 levels, but leptin fails to modulate peptide secretion and any element of the leptin signaling cascade. Despite this leptin resistance, the melanocortin system downstream of the ARH in DIO mice is over-responsive to melanocortin agonists, probably due to upregulation of MC4R. Lastly, we show that by decreasing the fat content of the mouse's diet, leptin responsiveness of NPY/AgRP and POMC neurons recovered simultaneously, with mice regaining normal leptin sensitivity and glycemic control. These results highlight the physiological importance of leptin sensing in the melanocortin circuits and show that their loss of leptin sensing likely contributes to the pathology of leptin resistance.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Leptina/farmacologia , Neurônios/metabolismo , Obesidade/metabolismo , Proteína Relacionada com Agouti , Animais , Núcleo Arqueado do Hipotálamo/citologia , Composição Corporal , Dieta , Gorduras na Dieta/administração & dosagem , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo/metabolismo , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Leptina/administração & dosagem , Masculino , Melanocortinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/metabolismo , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro , Transdução de Sinais , Redução de Peso , alfa-MSH/metabolismo
2.
Endocr Rev ; 42(1): 1-28, 2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33150398

RESUMO

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.


Assuntos
Glucose/metabolismo , Leptina/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Humanos , Insulina/metabolismo , Especificidade de Órgãos/efeitos dos fármacos
3.
Sci Rep ; 11(1): 14032, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34234216

RESUMO

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.


Assuntos
Dieta Hiperlipídica , Gorduras Insaturadas na Dieta/metabolismo , Metabolismo Energético , Ácidos Graxos Ômega-3/metabolismo , Hipernutrição/metabolismo , Fatores Etários , Animais , Biomarcadores , Diabetes Mellitus Experimental , Glucose/metabolismo , Hormônios/sangue , Hormônios/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Fosfatidilcolinas/sangue
4.
Diabetes ; 70(12): 2771-2784, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34544729

RESUMO

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.


Assuntos
Dieta Hiperlipídica , Glucose/metabolismo , Fenômenos Fisiológicos da Nutrição Pré-Natal , Animais , Animais Recém-Nascidos , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Suscetibilidade a Doenças , Fígado Gorduroso/etiologia , Fígado Gorduroso/metabolismo , Feminino , Idade Gestacional , Homeostase/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos , Hipernutrição/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Caracteres Sexuais , Fatores de Tempo
5.
Cell Rep Med ; 2(11): 100434, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34841287

RESUMO

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.


Assuntos
Progressão da Doença , Deleção de Genes , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , MicroRNAs/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Animais , Apoptose , Sequência de Bases , Linhagem Celular Tumoral , Movimento Celular , Dieta Hiperlipídica , Humanos , Secreção de Insulina , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/metabolismo , Especificidade de Órgãos , Ratos
6.
Sci Rep ; 11(1): 18394, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34526546

RESUMO

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.


Assuntos
Glucose/metabolismo , Leptina/metabolismo , Células Mieloides/metabolismo , Transdução de Sinais , Animais , Biomarcadores , Glicemia/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Metabolismo Energético , Técnicas de Silenciamento de Genes , Homeostase , Leptina/genética , Masculino , Doenças Metabólicas/etiologia , Doenças Metabólicas/metabolismo , Camundongos
7.
Sci Rep ; 10(1): 10518, 2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32601405

RESUMO

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.


Assuntos
Dependovirus , Células Secretoras de Insulina/metabolismo , Insulina/genética , Regiões Promotoras Genéticas , Recombinação Genética , Animais , Insulina/metabolismo , Integrases , Camundongos , Camundongos Transgênicos
8.
Sci Rep ; 9(1): 3619, 2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30842440

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental/patologia , Dieta Hiperlipídica/efeitos adversos , Proteínas de Homeodomínio/metabolismo , Resistência à Insulina , Células Secretoras de Insulina/patologia , Hipernutrição/complicações , Transativadores/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , Diabetes Mellitus Experimental/etiologia , Diabetes Mellitus Experimental/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Transativadores/genética , Aumento de Peso
9.
Sci Rep ; 9(1): 10829, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31346189

RESUMO

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.


Assuntos
Dependovirus , Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Proteínas de Fluorescência Verde , Animais , Glicemia , Peso Corporal/fisiologia , Insulina/sangue , Ilhotas Pancreáticas/metabolismo , Camundongos , Regiões Promotoras Genéticas , Ratos
10.
Sci Rep ; 9(1): 3307, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30824713

RESUMO

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.


Assuntos
Tecido Adiposo/metabolismo , Diabetes Mellitus Experimental , Técnicas de Silenciamento de Genes , Receptores para Leptina , Tecido Adiposo/patologia , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Camundongos , Camundongos Transgênicos , Receptores para Leptina/genética , Receptores para Leptina/metabolismo
11.
J Comp Neurol ; 506(2): 194-210, 2008 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-18022952

RESUMO

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.


Assuntos
Tronco Encefálico/citologia , Neurônios/metabolismo , Peptídeo YY/metabolismo , Animais , Tronco Encefálico/metabolismo , Hormônios Hipotalâmicos/metabolismo , Hibridização In Situ/métodos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macaca fascicularis , Masculino , Melaninas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vias Neurais/metabolismo , Neurônios/citologia , Neuropeptídeo Y/metabolismo , Neuropeptídeos/metabolismo , Orexinas , Peptídeo YY/genética , Hormônios Hipofisários/metabolismo , Ratos , Ratos Sprague-Dawley
12.
Cell Rep ; 22(1): 163-174, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29298418

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Hipotálamo/metabolismo , Células Neuroendócrinas/metabolismo , Fosforilação Oxidativa , Oxigênio/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipotálamo/citologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Transgênicos , Células Neuroendócrinas/citologia
13.
Endocrinology ; 148(7): 3279-87, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17412803

RESUMO

The hypothalamic neurocircuitry that regulates energy homeostasis in adult rats is not fully developed until the third postnatal week. In particular, fibers from the hypothalamic arcuate nucleus, including both neuropeptide Y (NPY) and alpha-MSH fibers, do not begin to innervate downstream hypothalamic targets until the second postnatal week. However, alpha-MSH fibers from the brainstem and melanocortin receptors are present in the hypothalamus at birth. The present study investigated the melanocortin system in the early postnatal period by examining effects of the melanocortin receptor agonist melanotan II (MTII) on body weight, energy expenditure, and hypothalamic NPY expression. Rat pups were injected ip with MTII (3 mg/kg body weight) or saline on postnatal day (P) 5 to P6, P10-P11, or P15-P16 at 1700 and 0900 h and then killed at 1300 h. Stomach weight and brown adipose tissue uncoupling protein 1 mRNA were determined. In addition, we assessed central c-Fos activation 90 min after MTII administration and hypothalamic NPY mRNA after twice daily MTII administration from P5-P10 or P10-P15. MTII induced hypothalamic c-Fos activation as well as attenuating body weight gain in rat pups. Stomach weight was significantly decreased and uncoupling protein 1 mRNA was increased at all ages, indicating decreased food intake and increased energy expenditure, respectively. However, MTII had no effect on NPY mRNA levels in any hypothalamic region. These findings demonstrate that MTII can inhibit food intake and stimulate energy expenditure before the full development of hypothalamic feeding neurocircuitry. These effects do not appear to be mediated by changes in NPY expression.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Canais Iônicos/genética , Proteínas Mitocondriais/genética , Peptídeos Cíclicos/farmacologia , Receptores da Corticotropina/metabolismo , alfa-MSH/análogos & derivados , Animais , Animais Lactentes , Relação Dose-Resposta a Droga , Feminino , Mucosa Gástrica/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Masculino , Neuropeptídeo Y/genética , Tamanho do Órgão/efeitos dos fármacos , Peptídeos Cíclicos/administração & dosagem , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores da Corticotropina/agonistas , Estômago/efeitos dos fármacos , Estômago/crescimento & desenvolvimento , Proteína Desacopladora 1 , alfa-MSH/administração & dosagem , alfa-MSH/farmacologia
14.
Endocrinology ; 148(9): 4150-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17525123

RESUMO

Excess weight gain during the early postnatal period increases the risk of persistent obesity into adulthood and impacts on the subsequent risk for metabolic and cardiovascular diseases. The current study investigated the long-term effect of early excess weight gain, through reduced nursing litter size, on body weight regulation and its relation to brown adipose tissue (BAT) thermogenesis. Animals raised in a small litter (SL, three pups per litter) were compared with those raised in a normal litter size (NL, eight pups per litter). BAT from young adult NL and SL rats, maintained under either ambient or cold conditions, were used for gene expression, morphological, and functional analysis. Compared with NL, SL rats showed excess weight gain, and adult SL animals had a reduced thermogenic capacity as displayed by lower levels of uncoupling protein 1 (UCP1). When exposed to cold, BAT from SL rats was less active and demonstrated reduced responsiveness to cold. Furthermore, reduction in transcript abundance of several lipid lipases and transcriptional regulators was observed in SL rats either at ambient temperature or under cold conditions. Finally, the expression of sympathetic beta 3-adrenergic receptor and the response to the sympathetic receptor agonist isoproterenol were decreased in SL rats. Overall, these observations provide the first evidence that postnatal excess weight gain results in abnormalities in BAT thermogenesis and sympathetic outflow, which likely increases susceptibility to obesity in adulthood.


Assuntos
Tecido Adiposo/fisiologia , Termogênese/fisiologia , Aumento de Peso/fisiologia , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/fisiologia , Animais , Humanos , Lactente , Insulina/sangue , Leptina/sangue , Lipólise , Tamanho da Ninhada de Vivíparos , Modelos Animais , Obesidade/etiologia , Ratos , Ratos Sprague-Dawley
15.
Mol Metab ; 6(9): 1052-1065, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28951828

RESUMO

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.


Assuntos
Glucose/metabolismo , Leptina/metabolismo , Leptina/fisiologia , Animais , Glicemia/efeitos dos fármacos , Corticosterona/metabolismo , Diabetes Mellitus/tratamento farmacológico , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético , Glucagon/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Insulina/metabolismo , Secreção de Insulina/efeitos dos fármacos , Leptina/genética , Fígado/efeitos dos fármacos , Receptores para Leptina/metabolismo
16.
J Diabetes Investig ; 7 Suppl 1: 87-93, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27186362

RESUMO

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.


Assuntos
Engenharia Celular , Diabetes Mellitus/terapia , Células Enteroendócrinas/fisiologia , Células Secretoras de Insulina/fisiologia , Insulina/uso terapêutico , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Células Enteroendócrinas/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Transplante das Ilhotas Pancreáticas , Camundongos , Ratos , Transplante de Células-Tronco
17.
Endocrinology ; 157(7): 2671-85, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27183315

RESUMO

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.


Assuntos
Glucagon/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Obesidade/metabolismo , Receptores para Leptina/metabolismo , Transdução de Sinais/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Dieta Hiperlipídica , Região Hipotalâmica Lateral/metabolismo , Secreção de Insulina , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Receptores para Leptina/genética , Núcleo Hipotalâmico Ventromedial/metabolismo
18.
Mol Metab ; 5(8): 716-724, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27656409

RESUMO

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.

19.
Brain Res Dev Brain Res ; 157(1): 74-82, 2005 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-15939087

RESUMO

Postnatal handling has been shown to attenuate some of the deficits in developmental outcome observed following prenatal ethanol exposure (E) although it appears to be ineffective at ameliorating the hypothalamic-pituitary-adrenal (HPA) hyperresponsiveness to stressors that has been observed in adult E animals. However, the effects of postnatal handling on central regulation of HPA activity in E animals, particularly with regard to alterations in steady-state hypothalamic corticotropin-releasing factor (CRF) activity, have not been examined. In the present study, offspring from E, pair-fed (PF), and ad-libitum-fed control (C) groups were exposed to daily handling during the first 2 weeks of life (H) or were left entirely undisturbed until weaning (NH). Basal CRF and arginine vasopressin (AVP) mRNA in the parvocellular portion of the paraventricular nucleus (pPVN) of the hypothalamus were assessed at 90-110 days of age. Prenatal ethanol exposure resulted in elevated basal pPVN CRF mRNA levels compared to those in ad-libitum-fed controls. Handling altered CRF mRNA levels in a sex-specific and prenatal treatment-specific manner. Females showed no significant effects of handling. In contrast, handling decreased CRF mRNA levels in PF and C but not E males compared to their NH counterparts. There were no effects of prenatal ethanol or postnatal handling on AVP mRNA levels. These findings indicate that prenatal ethanol exposure results in elevated basal CRF mRNA levels in adulthood and that handling appears to be ineffective in normalizing those elevations, supporting the suggestion that altered basal HPA regulation in E animals may, at least in part, underlie their HPA hyperresponsiveness to stressors.


Assuntos
Depressores do Sistema Nervoso Central/toxicidade , Hormônio Liberador da Corticotropina/metabolismo , Etanol/toxicidade , Manobra Psicológica , Núcleo Hipotalâmico Paraventricular/metabolismo , Efeitos Tardios da Exposição Pré-Natal , RNA Mensageiro/metabolismo , Análise de Variância , Animais , Contagem de Células , Hormônio Liberador da Corticotropina/genética , Feminino , Hibridização In Situ/métodos , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley
20.
Physiol Behav ; 86(5): 646-60, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16289141

RESUMO

In the normal adult rodent and primate, arcuate nucleus (ARH) neurons function as conduits for transmitting metabolic hormonal signals into the hypothalamic circuitry that modulates feeding and energy expenditure. We and others have shown that ARH projections do not fully develop until the 3rd postnatal week in the rodent. This is in stark contrast to the nonhuman primate (NHP) in which ARH projections develop during the 3rd trimester of pregnancy. This species difference suggests that maternal diet and health are likely key factors for the development of ARH projections in the primate, whereas the postnatal environment (i.e., diet) would be more important in the rodent. Furthermore, pertubations in these circuits during critical periods of development may have long-term consequences on feeding behavior and body weight management. Our group has used a rat model of overfeeding and underfeeding specifically during the postnatal period to begin to investigate the metabolic adaptions that may cause developmental abnormalities in the hypothalamic circuitry. While the overfed animals become obese as adults and the underfed maintain a lean phenotype, both models display low basal insulin and IGFII levels as adults. Furthermore, both models have abnormal expression of several key genes in peripheral metabolic tissue that are suggestive of changes in sympathetic outflow. Human studies show that gestational diabetes can also contribute to the development of obesity and diabetes in children; however, the mechanism is unknown. Since the critical periods for the development of hypothalamic circuits are different between rodents and primates our group has begun studies using NHP model to determine if maternal obesity/diabetes causes abnormalities in the development of metabolic systems, including the brain, in the offspring. To do this we have placed female NHPs on either a control diet or a high fat/calorie diet to induce obesity and diabetes. We have characterized the onset of insulin resistance and hyperleptinemia in these animals over the last 2(1/2) years and have collected offspring. Ongoing studies will investigate the metabolic abnormalities in these offspring.


Assuntos
Metabolismo/fisiologia , Rede Nervosa/fisiologia , Obesidade/fisiopatologia , Animais , Criança , Diabetes Gestacional/fisiopatologia , Feminino , Humanos , Obesidade/etiologia , Gravidez , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA