Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
2.
Mol Metab ; 5(10): 807-822, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27688995

RESUMO

OBJECTIVE: Central melanocortin pathways are well-established regulators of energy balance. However, scant data exist about the role of systemic melanocortin peptides. We set out to determine if peripheral α-melanocyte stimulating hormone (α-MSH) plays a role in glucose homeostasis and tested the hypothesis that the pituitary is able to sense a physiological increase in circulating glucose and responds by secreting α-MSH. METHODS: We established glucose-stimulated α-MSH secretion using humans, non-human primates, and mouse models. Continuous α-MSH infusions were performed during glucose tolerance tests and hyperinsulinemic-euglycemic clamps to evaluate the systemic effect of α-MSH in glucose regulation. Complementary ex vivo and in vitro techniques were employed to delineate the direct action of α-MSH via the melanocortin 5 receptor (MC5R)-PKA axis in skeletal muscles. Combined treatment of non-selective/selective phosphodiesterase inhibitor and α-MSH was adopted to restore glucose tolerance in obese mice. RESULTS: Here we demonstrate that pituitary secretion of α-MSH is increased by glucose. Peripheral α-MSH increases temperature in skeletal muscles, acts directly on soleus and gastrocnemius muscles to significantly increase glucose uptake, and enhances whole-body glucose clearance via the activation of muscle MC5R and protein kinase A. These actions are absent in obese mice, accompanied by a blunting of α-MSH-induced cAMP levels in skeletal muscles of obese mice. Both selective and non-selective phosphodiesterase inhibition restores α-MSH induced skeletal muscle glucose uptake and improves glucose disposal in obese mice. CONCLUSION: These data describe a novel endocrine circuit that modulates glucose homeostasis by pituitary α-MSH, which increases muscle glucose uptake and thermogenesis through the activation of a MC5R-PKA-pathway, which is disrupted in obesity.

3.
PLoS One ; 11(7): e0157027, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27467141

RESUMO

The melanocortin system includes five G-protein coupled receptors (family A) defined as MC1R-MC5R, which are stimulated by endogenous agonists derived from proopiomelanocortin (POMC). The melanocortin system has been intensely studied for its central actions in body weight and energy expenditure regulation, which are mainly mediated by MC4R. The pituitary gland is the source of various POMC-derived hormones released to the circulation, which raises the possibility that there may be actions of the melanocortins on peripheral energy homeostasis. In this study, we examined the molecular signaling pathway involved in α-MSH-stimulated glucose uptake in differentiated L6 myotubes and mouse muscle explants. In order to examine the involvement of AMPK, we investigate -MSH stimulation in both wild type and AMPK deficient mice. We found that -MSH significantly induces phosphorylation of TBC1 domain (TBC1D) family member 1 (S237 and T596), which is independent of upstream PKA and AMPK. We find no evidence to support that -MSH-stimulated glucose uptake involves TBC1D4 phosphorylation (T642 and S704) or GLUT4 translocation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Glucose/metabolismo , Músculo Esquelético/efeitos dos fármacos , alfa-MSH/farmacologia , Proteínas Quinases Ativadas por AMP/genética , Animais , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Fosforilação , Transdução de Sinais
5.
Mol Cell Endocrinol ; 418 Pt 2: 108-19, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26316427

RESUMO

The incidence of obesity and its related disorders are increasing at a rate of pandemic proportions. Understanding the mechanisms behind the maintenance of energy balance is fundamental in developing treatments for clinical syndromes including obesity and diabetes. A neural network located in the nucleus of the solitary tract-area postrema complex in the hindbrain and the hypothalamus in the forebrain has long been implicated in the control of energy balance. In the hypothalamus this central neuronal network consists of small populations of nuclei with distinct functions such as the arcuate nucleus (ARH), the paraventricular nuclei of the hypothalamus (PVH), the dorsomedial (DMH), the ventromedial (VMH) and the lateral hypothalamus (LH). These hypothalamic areas form interconnected neuronal circuits that respond to fluctuations in energy status by altering the expression of neuropeptides, leading to changes in energy intake and expenditure. Regulation of these hypothalamic nuclei involves the actions of orexigenic peptides (ie ghrelin), which act to stimulate energy intake and decrease energy expenditure, and anorexigenic peptides (ie. leptin and insulin), which act to reduce energy intake and stimulate energy expenditure. Here we review the role of the ARH, DMH and PVH in the control of energy homeostasis and how recent advances in research technologies (Cre-loxP technology, optogenetics and pharmacogenetics) have shed light on the role of these hypothalamic nuclei in the control of energy balance. Such novel findings include the implication of ARH POMC and AgRP neurons in the browning of white adipose tissue to regulate energy expenditure as well as the likely existence of divergent hypothalamic pathways in the DMH and PVH in the control of food intake and energy expenditure.


Assuntos
Tecido Adiposo/metabolismo , Peso Corporal/fisiologia , Encéfalo/metabolismo , Hipotálamo/metabolismo , Pâncreas/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Hipotalâmico Dorsomedial/metabolismo , Ingestão de Energia , Metabolismo Energético , Humanos , Leptina/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo
6.
Cell ; 159(6): 1404-16, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25480301

RESUMO

Obesity is associated with increased blood pressure (BP), which in turn increases the risk of cardiovascular diseases. We found that the increase in leptin levels seen in diet-induced obesity (DIO) drives an increase in BP in rodents, an effect that was not seen in animals deficient in leptin or leptin receptors (LepR). Furthermore, humans with loss-of-function mutations in leptin and the LepR have low BP despite severe obesity. Leptin's effects on BP are mediated by neuronal circuits in the dorsomedial hypothalamus (DMH), as blocking leptin with a specific antibody, antagonist, or inhibition of the activity of LepR-expressing neurons in the DMH caused a rapid reduction of BP in DIO mice, independent of changes in weight. Re-expression of LepRs in the DMH of DIO LepR-deficient mice caused an increase in BP. These studies demonstrate that leptin couples changes in weight to changes in BP in mammalian species.


Assuntos
Hipertensão/metabolismo , Leptina/metabolismo , Obesidade/metabolismo , Animais , Leptina/genética , Camundongos Endogâmicos C57BL , Mutação , Neurônios/metabolismo , Obesidade/patologia , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Transdução de Sinais
7.
Endocrinology ; 155(11): 4494-506, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25211588

RESUMO

Polycystic ovarian syndrome (PCOS), the most common female endocrine disorder of unknown etiology, is characterized by reproductive abnormalities and associated metabolic conditions comprising insulin resistance, type 2 diabetes mellitus, and dyslipidemia. We previously reported that transgenic overexpression of nerve growth factor (NGF), a marker of sympathetic hyperactivity, directed to the ovary by the mouse 17α-hydroxylase/C17-20 lyase promoter (17NF mice), results in ovarian abnormalities similar to those seen in PCOS women. To investigate whether ovarian overproduction of NGF also induces common metabolic alterations of PCOS, we assessed glucose homeostasis by glucose tolerance test, plasma insulin levels, and body composition by dual-energy x-ray absorptiometry scan in young female 17NF mice and wild-type mice. 17NF mice exhibited increased body weight and alterations in body fat distribution with a greater accumulation of visceral fat compared with sc fat (P < .01). 17NF mice also displayed glucose intolerance (P < .01), decreased insulin-mediated glucose disposal (P < .01), and hyperinsulinemia (P < .05), which, similar to PCOS patients, occurred independently of body weight. Additionally, 17NF mice exhibited increased sympathetic outflow observed as increased interscapular brown adipose tissue temperature. This change was evident during the dark period (7 pm to 7 am) and occurred concomitant with increased interscapular brown adipose tissue uncoupling protein 1 expression. These findings suggest that overexpression of NGF in the ovary may suffice to cause both reproductive and metabolic alterations characteristic of PCOS and support the hypothesis that sympathetic hyperactivity may contribute to the development and/or progression of PCOS.


Assuntos
Infertilidade Feminina/genética , Fator de Crescimento Neural/genética , Ovário/metabolismo , Síndrome do Ovário Policístico/genética , Animais , Modelos Animais de Doenças , Feminino , Humanos , Infertilidade Feminina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Crescimento Neural/metabolismo , Ovário/patologia , Fenótipo , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/patologia , Reprodução/genética , Regulação para Cima/genética
8.
Endocrinology ; 155(11): 4447-60, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25147981

RESUMO

In premenopausal and menopausal women in particular, suboptimal estrogens have been linked to the development of the metabolic syndrome as major contributors to fat accumulation. At the same time, estrogens have been described to have a role in regulating body metabolic status. We evaluated how endogenous or administered estrogens impact on the changes associated with high-fat diet (HFD) consumption in 2 different paradigms; ovarian-intact and in ovariectomized mice. When estradiol (E2) was cyclically administered to ovarian-intact HFD-fed mice for 12 weeks, animals gained significantly less weight than ovarian-intact vehicle controls (P < .01). This difference was mainly due to a reduced caloric intake but not to an increase in energy expenditure or locomotor activity. This E2 treatment regime to mice exposed to HFD was overall able to avoid the increase of visceral fat content to levels of those found in mice fed a regular chow diet. In the ovariectomized model, the main body weight and fat content reducing action of E2 was not only through decreasing food intake but also by increasing the whole-body energy expenditure, locomotor activity, and by inducing fat oxidation. Importantly, these animals became responsive to the anorexigenic effects of leptin in contrast to the vehicle-treated and the pair-fed control groups (P < .01). Further, in vitro hypothalamic secretion experiments revealed that treatment of obese mice with E2 is able to modulate the secretion of appetite-regulating neuropeptides; namely, E2 increased the secretion of the anorectic neuropeptide α-melanocyte-stimulating hormone and decreased the secretion of the orexigenic neuropetides neuropeptide Y and Agouti-related peptide. In conclusion, differences in response to E2 treatment of HFD-fed animals depend on their endogenous estrogenic status. Overall, E2 administration overcomes arcuate leptin resistance and partially prevents fat accumulation on these mice.


Assuntos
Resistência a Medicamentos/efeitos dos fármacos , Estradiol/farmacologia , Leptina/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Obesidade/metabolismo , Obesidade/prevenção & controle , Animais , Dieta Hiperlipídica , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/etiologia , Ovariectomia , Fatores Sexuais
9.
Am J Physiol Endocrinol Metab ; 303(5): E635-43, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22761161

RESUMO

Clinical studies have demonstrated a strong relationship between visceral fat content and metabolic diseases, such as type 2 diabetes and liver steatosis. Obese mouse models are an excellent tool to study metabolic diseases; however, there are limited methods for the noninvasive measurement of fat distribution in mice. Although micromagnetic resonance imaging and microcomputed tomography are the "gold standards" in the measurement of fat distribution, more economical and accessible methods are required. Dual energy X-ray absorptiometry (DEXA) is an effective method in characterizing fat content; however, it cannot discriminate between visceral and subcutaneous fat depots. We demonstrate that an evaluation of abdominal fat content measured by DEXA through the selection of one localized abdominal area strongly correlates with visceral fat content in C57BL/6J mice. We found that DEXA is able to measure fat pad volume ex vivo with high accuracy; however, the measurement of visceral fat in vivo shows an overestimation caused by subcutaneous tissue interference. The overestimation is almost constant for a wide range of values, and thus it is possible to correct the data for a more accurate estimation of visceral fat content. We demonstrate the utility of this technique in characterizing phenotypes of several obese mouse models (ob/ob, db/db, MC4R-KO, and DIO) and evaluating the effect of treatments on visceral fat content in longitudinal studies. Additionally, we also establish abdominal obesity as a potential biomarker for metabolic abnormalities (liver fat accumulation, insulin resistance/diabetes) in mice, similar to that described in humans.


Assuntos
Absorciometria de Fóton/métodos , Adiposidade , Modelos Animais de Doenças , Gordura Intra-Abdominal/diagnóstico por imagem , Doenças Metabólicas/diagnóstico por imagem , Animais , Feminino , Resistência à Insulina , Metabolismo dos Lipídeos , Fígado/metabolismo , Masculino , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Obesos , Obesidade Abdominal/diagnóstico por imagem , Obesidade Abdominal/metabolismo , Reprodutibilidade dos Testes , Caracteres Sexuais , Gordura Subcutânea Abdominal/diagnóstico por imagem , Imagem Corporal Total
10.
Adipocyte ; 1(3): 177-181, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23700530

RESUMO

Obesity is a global problem and effective drug therapy treatment is still unavailable. Obesity develops due to an imbalance between energy intake and energy expenditure (EE). Understanding what happens to EE in obesity may be the key to developing new treatments for obesity. If EE in obesity can be elevated, it could be a potential therapeutic target. We recently discovered that in baseline conditions obese mice have increased EE, in terms of thermogenesis. However, this increase in EE is not great enough to offset the elevated calorie intake that leads to the development of obesity. In obesity, the adipose derived hormone leptin is significantly elevated. This elevated leptin concentration appears to cause an increase in thermogenesis through increased sympathetic nerve activity (SNA) to brown adipose tissue deposits. The brain region of the dorsomedial hypothalamus (DMH) appears to be a key region that leptin activates in obesity to cause this increased thermogenesis. One unsettling finding is that the sympathetic nervous system (SNS) in obesity is elevated via leptin and it seems unlikely that SNA would be selectivity increased to only brown adipose tissue. Previously, it has been observed that leptin can increase SNA to numerous organs including the kidney. Furthermore, in obesity, SNA is increased in numerous organs. This leads to the critical question: is the leptin-mediated elevation of SNA and thermogenesis also chronically activating the kidney and contributing to the development of hypertension in obesity?

11.
Cell Metab ; 14(5): 684-99, 2011 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-22000926

RESUMO

In obesity, anorectic responses to leptin are diminished, giving rise to the concept of "leptin resistance." Increased expression of protein tyrosine phosphatase 1B (PTP1B) has been associated with the attenuation of leptin signaling and development of cellular leptin resistance. Here we report that hypothalamic levels of the tyrosine phosphatase TCPTP are also elevated in obesity to attenuate the leptin response. We show that mice that lack TCPTP in neuronal cells have enhanced leptin sensitivity and are resistant to high-fat-diet-induced weight gain and the development of leptin resistance. Also, intracerebroventricular administration of a TCPTP inhibitor enhances leptin signaling and responses in mice. Moreover, the combined deletion of TCPTP and PTP1B in neuronal cells has additive effects in the prevention of diet-induced obesity. Our results identify TCPTP as a critical negative regulator of hypothalamic leptin signaling and causally link elevated TCPTP to the development of cellular leptin resistance in obesity.


Assuntos
Hipotálamo/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 2/deficiência , Transdução de Sinais , Animais , Glicemia/análise , Composição Corporal/efeitos dos fármacos , Dieta Hiperlipídica , Inibidores Enzimáticos/farmacologia , Feminino , Expressão Gênica , Hipotálamo/citologia , Infusões Intraventriculares , Insulina/sangue , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Obesidade/genética , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , Proteína Tirosina Fosfatase não Receptora Tipo 2/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 2/genética , Receptores para Leptina/metabolismo , Técnicas de Cultura de Tecidos
12.
J Neurosci ; 31(34): 12189-97, 2011 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-21865462

RESUMO

Leptin regulates body weight in mice by decreasing appetite and increasing sympathetic nerve activity (SNA), which increases energy expenditure in interscapular brown adipose tissue (iBAT). Diet-induced obese mice (DIO) are resistant to the anorectic actions of leptin. We evaluated whether leptin still stimulated sympathetic outflow in DIO mice. We measured iBAT temperature as a marker of SNA. We found that obese hyperleptinemic mice have higher iBAT temperature than mice on regular diet. Conversely, obese leptin-deficient ob/ob mice have lower iBAT temperature. Additionally, leptin increased SNA in obese (DIO and ob/ob) and control mice, despite DIO mice being resistant to anorectic action of leptin. We demonstrated that neurons in the dorsomedial hypothalamus (DMH) of DIO mice mediate the thermogenic responses to hyperleptinemia in obese mammals because blockade of leptin receptors in the DMH prevented the thermogenic effects of leptin. Peripheral Melotan II (MTII) injection increased iBAT temperature, but it was blunted by blockade of DMH melanocortin receptors (MC4Rs) by injecting agouti-related peptide (AgRP) directly into the DMH, suggesting a physiological role of the DMH on temperature regulation in animals with normal body weight. Nevertheless, obese mice without a functional melanocortin system (MC4R KO mice) have an increased sympathetic outflow to iBAT compared with their littermates, suggesting that higher leptin levels drive sympathoexcitation to iBAT by a melanocortin-independent pathway. Because the sympathetic nervous system contributes in regulating blood pressure, heart rate, and hepatic glucose production, selective leptin resistance may be a crucial mechanism linking adiposity and metabolic syndrome.


Assuntos
Tecido Adiposo Marrom/fisiologia , Núcleo Hipotalâmico Dorsomedial/fisiologia , Leptina/fisiologia , Sistema Nervoso Simpático/fisiologia , Termogênese/fisiologia , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/inervação , Animais , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Modelos Animais de Doenças , Núcleo Hipotalâmico Dorsomedial/efeitos dos fármacos , Resistência a Medicamentos/efeitos dos fármacos , Resistência a Medicamentos/fisiologia , Leptina/deficiência , Leptina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/genética , Obesidade/metabolismo , Receptores para Leptina/antagonistas & inibidores , Receptores para Leptina/fisiologia , Sistema Nervoso Simpático/efeitos dos fármacos , Termogênese/efeitos dos fármacos
13.
Endocrinology ; 151(10): 4745-55, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20826561

RESUMO

Circulating ghrelin is decreased in obesity, and peripheral ghrelin does not induce food intake in obese mice. We investigated whether ghrelin resistance was a centrally mediated phenomenon involving dysregulated neuropeptide Y (NPY) and agouti-related peptide (AgRP) circuits. We show that diet-induced obesity (DIO) (12 wk) suppresses the neuroendocrine ghrelin system by decreasing acylated and total plasma ghrelin, decreasing ghrelin and Goat mRNA in the stomach, and decreasing expression of hypothalamic GHSR. Peripheral (ip) or central (intracerebroventricular) ghrelin injection was able to induce food intake and arcuate nucleus Fos immunoreactivity in chow-fed but not high-fat diet-fed mice. DIO decreased expression of Npy and Agrp mRNA, and central ghrelin was unable to promote expression of these genes. Ghrelin did not induce AgRP or NPY secretion in hypothalamic explants from DIO mice. Injection of NPY intracerebroventricularly increased food intake in both chow-fed and high-fat diet-fed mice, indicating that downstream NPY/AgRP neural targets are intact and that defective NPY/AgRP function is a primary cause of ghrelin resistance. Ghrelin resistance in DIO is not confined to the NPY/AgRP neurons, because ghrelin did not stimulate growth hormone secretion in DIO mice. Collectively, our data suggests that DIO causes ghrelin resistance by reducing NPY/AgRP responsiveness to plasma ghrelin and suppressing the neuroendocrine ghrelin axis to limit further food intake. Ghrelin has a number of functions in the brain aside from appetite control, including cognitive function, mood regulation, and protecting against neurodegenerative diseases. Thus, central ghrelin resistance may potentiate obesity-related cognitive decline, and restoring ghrelin sensitivity may provide therapeutic outcomes for maintaining healthy aging.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Resistência a Medicamentos , Grelina/metabolismo , Neuropeptídeo Y/metabolismo , Obesidade/metabolismo , Animais , Cognição/efeitos dos fármacos , Cognição/fisiologia , Dieta Aterogênica , Resistência a Medicamentos/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Grelina/sangue , Grelina/genética , Grelina/farmacologia , Cabras , Hormônio do Crescimento/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Neurônios/metabolismo , Obesidade/sangue , Obesidade/complicações , Obesidade/etiologia
14.
Proc Natl Acad Sci U S A ; 107(33): 14875-80, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20679202

RESUMO

The neuronal circuits involved in the regulation of feeding behavior and energy expenditure are soft-wired, reflecting the relative activity of the postsynaptic neuronal system, including the anorexigenic proopiomelanocortin (POMC)-expressing cells of the arcuate nucleus. We analyzed the synaptic input organization of the melanocortin system in lean rats that were vulnerable (DIO) or resistant (DR) to diet-induced obesity. We found a distinct difference in the quantitative and qualitative synaptology of POMC cells between DIO and DR animals, with a significantly greater number of inhibitory inputs in the POMC neurons in DIO rats compared with DR rats. When exposed to a high-fat diet (HFD), the POMC cells of DIO animals lost synapses, whereas those of DR rats recruited connections. In both DIO rats and mice, the HFD-triggered loss of synapses on POMC neurons was associated with increased glial ensheathment of the POMC perikarya. The altered synaptic organization of HFD-fed animals promoted increased POMC tone and a decrease in the stimulatory connections onto the neighboring neuropeptide Y (NPY) cells. Exposure to HFD was associated with reactive gliosis, and this affected the structure of the blood-brain barrier such that the POMC and NPY cell bodies and dendrites became less accessible to blood vessels. Taken together, these data suggest that consumption of an HFD has a major impact on the cytoarchitecture of the arcuate nucleus in vulnerable subjects, with changes that might be irreversible due to reactive gliosis.


Assuntos
Dieta , Gliose/metabolismo , Melanocortinas/metabolismo , Obesidade/metabolismo , Sinapses/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Núcleo Arqueado do Hipotálamo/fisiopatologia , Gorduras na Dieta/efeitos adversos , Feminino , Gliose/etiologia , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica , Neurônios/metabolismo , Neurônios/ultraestrutura , Neuropeptídeo Y/metabolismo , Obesidade/etiologia , Pró-Opiomelanocortina/metabolismo , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica/fisiologia
15.
J Pediatr Endocrinol Metab ; 23(1-2): 101-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20432813

RESUMO

AIM: Gastrointestinal (GI) hormones are involved in satiety regulation and in glucose metabolism. Most GI hormones are hydrolyzed and inactivated by the same enzyme, dipeptidyl peptidase IV (DPP-IV). We analyzed changes of DPP-IV after weight loss in obese children and its relationships to the GI hormones pancreatic peptide (PP), peptide YY (PYY), and insulin sensitivity. METHODS: We measured at baseline and one year later anthropometrics, percentage body fat based on skinfold thickness, DPP-IV, PP, PYY, insulin, and glucose concentrations in 18 obese children (mean age 10.9 years, 44% male, mean BMI 28.5 kg/m2) who participated in a one-year lifestyle intervention program based on physical activity, nutrition course, and behavioral therapy. Insulin sensitivity was calculated using QUICKI. RESULTS: Changes of DPP-IV correlated significantly to the changes of percentage body fat (r = 0.47) and BMI SDS (r = 0.60). In partial regression analysis adjusted for change in weight status, changes of DPP-IV correlated significantly to changes of PYY (r = -0.43), PP (r = -0.49), QUICKI (r = -0.53), and insulin (r = 0.57). The 10 children with substantial weight loss significantly reduced their DPP-IV and insulin concentrations, while QUICKI, PYY, and PP levels significantly increased. In children without substantial weight loss no significant changes were observed. CONCLUSIONS: These findings suggest that the increase of fasting PP and PYY in weight loss is influenced at least in part by a decrease of their cleavage enzyme DPP-IV. Further research is necessary to evaluate the mechanisms in weight loss leading to a decrease of DPP-IV activity and consequently to an improvement of insulin sensitivity.


Assuntos
Dipeptidil Peptidase 4/sangue , Resistência à Insulina , Obesidade/sangue , Obesidade/terapia , Polipeptídeo Pancreático/sangue , Peptídeo YY/sangue , Adolescente , Assistência Ambulatorial , Glicemia/metabolismo , Criança , Serviços de Saúde da Criança , Feminino , Humanos , Insulina/sangue , Estilo de Vida , Masculino , Resultado do Tratamento , Redução de Peso
16.
Endocrinology ; 151(4): 1598-610, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20194730

RESUMO

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.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Gorduras na Dieta/metabolismo , Leptina/metabolismo , Hipernutrição/metabolismo , Fator de Transcrição STAT3/metabolismo , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Área Sob a Curva , Glicemia , Peso Corporal/genética , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Teste de Tolerância a Glucose , Imuno-Histoquímica , Insulina/metabolismo , Resistência à Insulina/genética , Leptina/farmacologia , Camundongos , Atividade Motora/fisiologia , Hipernutrição/genética , Fosforilação/fisiologia , Radioimunoensaio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética
17.
Metabolism ; 59(2): 186-94, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19766264

RESUMO

Relationships of blood circulating melanocortins to childhood obesity are not well established. We evaluated serum alpha-melanocyte-stimulating hormone (alpha-MSH) in lean children and different study groups of childhood obesity. We examined serum alpha-MSH in 52 otherwise healthy children with childhood obesity (Ob; mean age, 11 years; 32 girls/20 boys), 27 normal-weight children of same age, 7 additional obese patients with reduced melanocortin-4 receptor function (MC4Rmut), and 22 patients with craniopharyngioma (CP). Fasting serum alpha-MSH and leptin were measured by radioimmunoassay. Serum alpha-MSH was also evaluated 1 hour after 500-kcal liquid meal (CP and Ob) and at the end of 1-year lifestyle intervention in 24 Ob patients. The alpha-MSH levels were similar in obese vs lean children but significantly lower in CP (P < .001) and significantly higher (P < .05) in MC4Rmut patients compared with Ob. One hour after liquid meal, alpha-MSH increased in patients with Ob but not with CP. After 1 year, alpha-MSH levels increased significantly in the successful weight reduction Ob subgroup despite unchanged cortisol levels. The alpha-MSH changes correlated to weight status changes (r = 0.67, P = .0003) but not to changes of cortisol, insulin, or homeostasis model assessment of insulin resistance index. Persistently low alpha-MSH levels in CP patients are suspected to be due to pituitary or hypothalamic damage. High peripheral levels in MC4Rmut carriers indicate up-regulation of alpha-MSH. Changes of weight status are associated with changes of peripheral alpha-MSH.


Assuntos
Obesidade/sangue , alfa-MSH/sangue , Adolescente , Índice de Massa Corporal , Peso Corporal , Criança , Craniofaringioma/sangue , Feminino , Genótipo , Humanos , Hidrocortisona/sangue , Insulina/sangue , Resistência à Insulina , Leptina/sangue , Masculino , Mutação , Obesidade/genética , Obesidade/terapia , Neoplasias Hipofisárias/sangue , Receptor Tipo 4 de Melanocortina/genética
18.
Endocrinology ; 149(10): 4837-45, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18583425

RESUMO

Agouti-related protein (AgRP) is an orexigenic neuropeptide produced by neurons in the hypothalamic arcuate nucleus (ARC) that is a key component of central neural circuits that control food intake and energy expenditure. Disorders in energy homeostasis, characterized by hypophagia and increased metabolic rate, frequently develop in animals with either acute or chronic diseases. Recently, studies have demonstrated that proopiomelanocortin-expressing neurons in the ARC are activated by the proinflammatory cytokine IL-1beta. In the current study, we sought to determine whether inflammatory processes regulate the expression of AgRP mRNA and to characterize the response of AgRP neurons to IL-1beta. Here, we show by real-time RT-PCR and in situ hybridization analysis that AgRP mRNA expression in rodents is increased in models of acute and chronic inflammation. AgRP neurons were found to express the type I IL-1 receptor, and the percentage of expression was significantly increased after peripheral administration of lipopolysaccharide. Furthermore, we demonstrate that IL-1beta inhibits the release of AgRP from hypothalamic explants. Collectively, these data indicate that proinflammatory signals decrease the secretion of AgRP while increasing the transcription of the AgRP gene. These observations suggest that AgRP neurons may participate with ARC proopiomelanocortin neurons in mediating the anorexic and metabolic responses to acute and chronic disease processes.


Assuntos
Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Inflamação/fisiopatologia , Transcrição Gênica/imunologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Núcleo Arqueado do Hipotálamo/citologia , Transplante de Tecido Encefálico , Doença Crônica , Modelos Animais de Doenças , Inflamação/induzido quimicamente , Inflamação/imunologia , Interleucina-1beta/metabolismo , Cetorolaco/farmacologia , Falência Renal Crônica/imunologia , Falência Renal Crônica/fisiopatologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Neoplasias/fisiopatologia , Neurônios/fisiologia , Prostaglandinas/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Receptores de Interleucina-1/genética
19.
PLoS One ; 3(5): e2202, 2008 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-18493584

RESUMO

BACKGROUND: Cannabinoids, the active components of marijuana, stimulate appetite, and cannabinoid receptor-1 (CB1-R) antagonists suppress appetite and promote weight loss. Little is known about how CB1-R antagonists affect the central neurocircuitry, specifically the melanocortin system that regulates energy balance. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show that peripherally administered CB1-R antagonist (AM251) or agonist equally suppressed or stimulated feeding respectively in A(y) , which lack a functional melanocortin system, and wildtype mice, demonstrating that cannabinoid effects on feeding do not require melanocortin circuitry. CB1-R antagonist or agonist administered into the ventral tegmental area (VTA) equally suppressed or stimulated feeding respectively, in both genotypes. In addition, peripheral and central cannabinoid administration similarly induced c-Fos activation in brain sites suggesting mediation via motivational dopaminergic circuitry. Amperometry-detected increases in evoked dopamine (DA) release by the CB1-R antagonist in nucleus accumbens slices indicates that AM251 modulates DA release from VTA terminals. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that the effects of cannabinoids on energy balance are independent of hypothalamic melanocortin circuitry and is primarily driven by the reward system.


Assuntos
Canabinoides/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Melanocortinas/fisiologia , Animais , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley
20.
Nature ; 449(7159): 228-32, 2007 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-17728716

RESUMO

A subset of neurons in the brain, known as 'glucose-excited' neurons, depolarize and increase their firing rate in response to increases in extracellular glucose. Similar to insulin secretion by pancreatic beta-cells, glucose excitation of neurons is driven by ATP-mediated closure of ATP-sensitive potassium (K(ATP)) channels. Although beta-cell-like glucose sensing in neurons is well established, its physiological relevance and contribution to disease states such as type 2 diabetes remain unknown. To address these issues, we disrupted glucose sensing in glucose-excited pro-opiomelanocortin (POMC) neurons via transgenic expression of a mutant Kir6.2 subunit (encoded by the Kcnj11 gene) that prevents ATP-mediated closure of K(ATP) channels. Here we show that this genetic manipulation impaired the whole-body response to a systemic glucose load, demonstrating a role for glucose sensing by POMC neurons in the overall physiological control of blood glucose. We also found that glucose sensing by POMC neurons became defective in obese mice on a high-fat diet, suggesting that loss of glucose sensing by neurons has a role in the development of type 2 diabetes. The mechanism for obesity-induced loss of glucose sensing in POMC neurons involves uncoupling protein 2 (UCP2), a mitochondrial protein that impairs glucose-stimulated ATP production. UCP2 negatively regulates glucose sensing in POMC neurons. We found that genetic deletion of Ucp2 prevents obesity-induced loss of glucose sensing, and that acute pharmacological inhibition of UCP2 reverses loss of glucose sensing. We conclude that obesity-induced, UCP2-mediated loss of glucose sensing in glucose-excited neurons might have a pathogenic role in the development of type 2 diabetes.


Assuntos
Glucose/metabolismo , Homeostase , Neurônios/metabolismo , Obesidade/fisiopatologia , Pró-Opiomelanocortina/metabolismo , Trifosfato de Adenosina/biossíntese , Trifosfato de Adenosina/metabolismo , Animais , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/farmacologia , Humanos , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/genética , Canais Iônicos/metabolismo , Glicosídeos Iridoides , Iridoides/farmacologia , Camundongos , Camundongos Obesos , Camundongos Transgênicos , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Obesidade/induzido quimicamente , Obesidade/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Proteína Desacopladora 2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...