Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Cell ; 164(3): 353-64, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26824653

RESUMO

More than one-half billion people are obese, and despite progress in genetic research, much of the heritability of obesity remains enigmatic. Here, we identify a Trim28-dependent network capable of triggering obesity in a non-Mendelian, "on/off" manner. Trim28(+/D9) mutant mice exhibit a bi-modal body-weight distribution, with isogenic animals randomly emerging as either normal or obese and few intermediates. We find that the obese-"on" state is characterized by reduced expression of an imprinted gene network including Nnat, Peg3, Cdkn1c, and Plagl1 and that independent targeting of these alleles recapitulates the stochastic bi-stable disease phenotype. Adipose tissue transcriptome analyses in children indicate that humans too cluster into distinct sub-populations, stratifying according to Trim28 expression, transcriptome organization, and obesity-associated imprinted gene dysregulation. These data provide evidence of discrete polyphenism in mouse and man and thus carry important implications for complex trait genetics, evolution, and medicine.


Assuntos
Epigênese Genética , Haploinsuficiência , Proteínas Nucleares/genética , Obesidade/genética , Proteínas Repressoras/genética , Magreza/genética , Adolescente , Animais , Índice de Massa Corporal , Criança , Pré-Escolar , Humanos , Camundongos , Inquéritos Nutricionais , Polimorfismo Genético , Proteína 28 com Motivo Tripartido
3.
Nature ; 578(7795): 444-448, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31875646

RESUMO

Metformin, the world's most prescribed anti-diabetic drug, is also effective in preventing type 2 diabetes in people at high risk1,2. More than 60% of this effect is attributable to the ability of metformin to lower body weight in a sustained manner3. The molecular mechanisms by which metformin lowers body weight are unknown. Here we show-in two independent randomized controlled clinical trials-that metformin increases circulating levels of the peptide hormone growth/differentiation factor 15 (GDF15), which has been shown to reduce food intake and lower body weight through a brain-stem-restricted receptor. In wild-type mice, oral metformin increased circulating GDF15, with GDF15 expression increasing predominantly in the distal intestine and the kidney. Metformin prevented weight gain in response to a high-fat diet in wild-type mice but not in mice lacking GDF15 or its receptor GDNF family receptor α-like (GFRAL). In obese mice on a high-fat diet, the effects of metformin to reduce body weight were reversed by a GFRAL-antagonist antibody. Metformin had effects on both energy intake and energy expenditure that were dependent on GDF15, but retained its ability to lower circulating glucose levels in the absence of GDF15 activity. In summary, metformin elevates circulating levels of GDF15, which is necessary to obtain its beneficial effects on energy balance and body weight, major contributors to its action as a chemopreventive agent.


Assuntos
Peso Corporal/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Fator 15 de Diferenciação de Crescimento/metabolismo , Metformina/farmacologia , Administração Oral , Adulto , Idoso , Animais , Glicemia/análise , Glicemia/metabolismo , Dieta Hiperlipídica , Método Duplo-Cego , Ingestão de Energia/efeitos dos fármacos , Enterócitos/citologia , Enterócitos/efeitos dos fármacos , Feminino , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/antagonistas & inibidores , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator 15 de Diferenciação de Crescimento/sangue , Fator 15 de Diferenciação de Crescimento/deficiência , Fator 15 de Diferenciação de Crescimento/genética , Homeostase/efeitos dos fármacos , Humanos , Intestinos/citologia , Intestinos/efeitos dos fármacos , Masculino , Metformina/administração & dosagem , Camundongos , Camundongos Obesos , Pessoa de Meia-Idade , Redução de Peso/efeitos dos fármacos
4.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34187898

RESUMO

An acute increase in the circulating concentration of glucocorticoid hormones is essential for the survival of severe somatic stresses. Circulating concentrations of GDF15, a hormone that acts in the brain to reduce food intake, are frequently elevated in stressful states. We now report that GDF15 potently activates the hypothalamic-pituitary-adrenal (HPA) axis in mice and rats. A blocking antibody to the GDNF-family receptor α-like receptor completely prevented the corticosterone response to GDF15 administration. In wild-type mice exposed to a range of stressful stimuli, circulating levels of both corticosterone and GDF15 rose acutely. In the case of Escherichia coli or lipopolysaccharide injections, the vigorous proinflammatory cytokine response elicited was sufficient to produce a near-maximal HPA response, regardless of the presence or absence of GDF15. In contrast, the activation of the HPA axis seen in wild-type mice in response to the administration of genotoxic or endoplasmic reticulum toxins, which do not provoke a marked rise in cytokines, was absent in Gdf15-/- mice. In conclusion, consistent with its proposed role as a sentinel hormone, endogenous GDF15 is required for the activation of the protective HPA response to toxins that do not induce a substantial cytokine response. In the context of efforts to develop GDF15 as an antiobesity therapeutic, these findings identify a biomarker of target engagement and a previously unrecognized pharmacodynamic effect, which will require monitoring in human studies.


Assuntos
Fator 15 de Diferenciação de Crescimento/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Animais , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Glucocorticoides/metabolismo , Fator 15 de Diferenciação de Crescimento/administração & dosagem , Humanos , Lipopolissacarídeos , Camundongos , Ratos , Tunicamicina/farmacologia
5.
Phys Rev Lett ; 130(11): 111801, 2023 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-37001070

RESUMO

We present the first search for the pair production of dark particles X via K_{L}^{0}→XX with X decaying into two photons using the data collected by the KOTO experiment. No signal was observed in the mass range of 40-110 MeV/c^{2} and 210-240 MeV/c^{2}. This sets upper limits on the branching fractions as B(K_{L}^{0}→XX)<(1-4)×10^{-7} and B(K_{L}^{0}→XX)<(1-2)×10^{-6} at the 90% confidence level for the two mass regions, respectively.

6.
Eur Cell Mater ; 40: 133-145, 2020 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-32951194

RESUMO

Vascularisation efficiency plays an essential role in the success of bulk transplantation, while biocompatibility and safety are major concerns in clinical applications. Fibrin-based hydrogels have been exploited as scaffolds for their advantages in biocompatibility, degradability and mass transportation in various forms. However, the mechanical strength and degree of vascularisation remain unsatisfactory for clinical usage. An interpenetrating hydrogel was developed by adding hyaluronic acid (HA) to a fibrin-based natural hydrogel. The vasculogenesis of endothelial cells (human umbilical vein endothelial cells, HUVECs) was characterised within the gel using both in vitro and in vivo animal studies. The in vitro vascular morphology analysis showed 17.9 % longer mean tube length and 14.3 % higher average thickness in 7 d cultivation within the HA-supplemented hydrogel. The in vivo results showed 51.6 % larger total tube area, 1.8 × longer average tube length and 81.6 % higher cell number in the HA-supplemented hydrogel compared to the hydrogel without HA. The experimental results demonstrated better vascularisation and cell recruitment in the HA- supplemented hydrogel. The material properties of the hydrogels were also analysed using atomic force microscopy (AFM). The results revealed 3.7 × higher elasticity of the HA-supplemented hydrogel, which provided better mechanical strength and support for easy handling during procedures. With the demonstrated advantages, the developed hydrogels showed promise for exploitation in various practical clinical applications.


Assuntos
Fibrina/farmacologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Ácido Hialurônico/farmacologia , Hidrogéis/farmacologia , Modelos Biológicos , Neovascularização Fisiológica , Animais , Elasticidade , Fluorescência , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Morfogênese/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos
7.
Proc Natl Acad Sci U S A ; 114(35): 9421-9426, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28811369

RESUMO

An intergenic region of human chromosome 2 (2p25.3) harbors genetic variants which are among those most strongly and reproducibly associated with obesity. The gene closest to these variants is TMEM18, although the molecular mechanisms mediating these effects remain entirely unknown. Tmem18 expression in the murine hypothalamic paraventricular nucleus (PVN) was altered by changes in nutritional state. Germline loss of Tmem18 in mice resulted in increased body weight, which was exacerbated by high fat diet and driven by increased food intake. Selective overexpression of Tmem18 in the PVN of wild-type mice reduced food intake and also increased energy expenditure. We provide evidence that TMEM18 has four, not three, transmembrane domains and that it physically interacts with key components of the nuclear pore complex. Our data support the hypothesis that TMEM18 itself, acting within the central nervous system, is a plausible mediator of the impact of adjacent genetic variation on human adiposity.


Assuntos
Apetite/genética , Peso Corporal/genética , Proteínas de Membrana/metabolismo , Obesidade/genética , Animais , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteínas de Transporte Vesicular
8.
PLoS Genet ; 9(1): e1003166, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23300482

RESUMO

The strongest BMI-associated GWAS locus in humans is the FTO gene. Rodent studies demonstrate a role for FTO in energy homeostasis and body composition. The phenotypes observed in loss of expression studies are complex with perinatal lethality, stunted growth from weaning, and significant alterations in body composition. Thus understanding how and where Fto regulates food intake, energy expenditure, and body composition is a challenge. To address this we generated a series of mice with distinct temporal and spatial loss of Fto expression. Global germline loss of Fto resulted in high perinatal lethality and a reduction in body length, fat mass, and lean mass. When ratio corrected for lean mass, mice had a significant increase in energy expenditure, but more appropriate multiple linear regression normalisation showed no difference in energy expenditure. Global deletion of Fto after the in utero and perinatal period, at 6 weeks of age, removed the high lethality of germline loss. However, there was a reduction in weight by 9 weeks, primarily as loss of lean mass. Over the subsequent 10 weeks, weight converged, driven by an increase in fat mass. There was a switch to a lower RER with no overall change in food intake or energy expenditure. To test if the phenotype can be explained by loss of Fto in the mediobasal hypothalamus, we sterotactically injected adeno-associated viral vectors encoding Cre recombinase to cause regional deletion. We observed a small reduction in food intake and weight gain with no effect on energy expenditure or body composition. Thus, although hypothalamic Fto can impact feeding, the effect of loss of Fto on body composition is brought about by its actions at sites elsewhere. Our data suggest that Fto may have a critical role in the control of lean mass, independent of its effect on food intake.


Assuntos
Composição Corporal/genética , Ingestão de Alimentos/genética , Metabolismo Energético/genética , Oxigenases de Função Mista/genética , Obesidade , Oxo-Ácido-Liases/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Peso Corporal/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Homeostase , Humanos , Masculino , Camundongos , Obesidade/genética , Obesidade/metabolismo
9.
Eur J Neurol ; 21(5): 797-801, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24629033

RESUMO

BACKGROUND AND PURPOSE: Anosognosia and neglect may coexist in stroke patients. Neglect patients often report poor quality of life (QOL), whereas patients suffering from other cognition disorders with poor insight report better QOL. This study investigates the relationship between anosognosia, neglect and QOL amongst stroke survivors. METHODS: Stroke survivors who met the criteria were used as a sampling pool. Sixty stroke patients were observed in this study, amongst whom 20 patients with anosognosia and neglect (A+N+), 20 patients with neglect but not anosognosia (A-N+) and 20 patients with neither anosognosia nor neglect (A-N-) were selected from the sampling pool based on demographic characteristics matched with the A+N+ group. A questionnaire (SS-QOL) was used to collect the QOL perceived by the stroke survivors. RESULTS: The perceived QOL of the A+N+ group was significantly better than those of the other groups, including the subscales of self-care, mobility, work/productivity, upper extremity, mood, family role and social role. However, the A+N+ group had poor balance level and more fall incidents were reported. CONCLUSION: The A+N+ group perceived better QOL but had more falls and poorer balance than the other groups. Health providers should work with caregivers aggressively in preventing accidents.


Assuntos
Agnosia/etiologia , Lateralidade Funcional/fisiologia , Transtornos da Percepção/etiologia , Qualidade de Vida , Acidente Vascular Cerebral , Adulto , Idoso , Análise de Variância , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/etiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/mortalidade , Acidente Vascular Cerebral/psicologia
10.
Proc Natl Acad Sci U S A ; 106(23): 9350-5, 2009 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-19470471

RESUMO

Tre-2, BUB2, CDC16, 1 domain family member 4 (TBC1D4) (AS160) is a Rab-GTPase activating protein implicated in insulin-stimulated glucose transporter 4 (GLUT4) translocation in adipocytes and myotubes. To determine whether loss-of-function mutations in TBC1D4 might impair GLUT4 translocation and cause insulin resistance in humans, we screened the coding regions of this gene in 156 severely insulin-resistant patients. A female presenting at age 11 years with acanthosis nigricans and extreme postprandial hyperinsulinemia was heterozygous for a premature stop mutation (R363X) in TBC1D4. After demonstrating reduced expression of wild-type TBC1D4 protein and expression of the truncated protein in lymphocytes from the proband, we further characterized the biological effects of the truncated protein in 3T3L1 adipocytes. Prematurely truncated TBC1D4 protein tended to increase basal cell membrane GLUT4 levels (P = 0.053) and significantly reduced insulin-stimulated GLUT4 cell membrane translocation (P < 0.05). When coexpressed with wild-type TBC1D4, the truncated protein dimerized with full-length TBC1D4, suggesting that the heterozygous truncated variant might interfere with its wild-type counterpart in a dominant negative fashion. Two overweight family members with the mutation also manifested normal fasting glucose and insulin levels but disproportionately elevated insulin levels following an oral glucose challenge. This family provides unique genetic evidence of TBC1D4 involvement in human insulin action.


Assuntos
Acantose Nigricans/genética , Proteínas Ativadoras de GTPase/genética , Hiperinsulinismo/genética , Códon sem Sentido , Feminino , Transportador de Glucose Tipo 4/genética , Humanos , Masculino , Linhagem , Mutação Puntual
11.
Mol Metab ; 43: 101127, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33242659

RESUMO

OBJECTIVE: More than 300 genetic variants have been robustly associated with measures of human adiposity. Highly penetrant mutations causing human obesity do so largely by disrupting satiety pathways in the brain and increasing food intake. Most of the common obesity-predisposing variants are in, or near, genes expressed highly in the brain, but little is known of their function. Exploring the biology of these genes at scale in mammalian systems is challenging. We sought to establish and validate the use of a multicomponent screen for feeding behaviour phenotypes, taking advantage of the tractable model organism Drosophila melanogaster. METHODS: We validated a screen for feeding behaviour in Drosophila by comparing results after disrupting the expression of centrally expressed genes that influence energy balance in flies to those of 10 control genes. We then used this screen to explore the effects of disrupted expression of genes either a) implicated in energy homeostasis through human genome-wide association studies (GWAS) or b) expressed and nutritionally responsive in specific populations of hypothalamic neurons with a known role in feeding/fasting. RESULTS: Using data from the validation study to classify responses, we studied 53 Drosophila orthologues of genes implicated by human GWAS in body mass index and found that 15 significantly influenced feeding behaviour or energy homeostasis in the Drosophila screen. We then studied 50 Drosophila homologues of 47 murine genes reciprocally nutritionally regulated in POMC and agouti-related peptide neurons. Seven of these 50 genes were found by our screen to influence feeding behaviour in flies. CONCLUSION: We demonstrated the utility of Drosophila as a tractable model organism in a high-throughput genetic screen for food intake phenotypes. This simple, cost-efficient strategy is ideal for high-throughput interrogation of genes implicated in feeding behaviour and obesity in mammals and will facilitate the process of reaching a functional understanding of obesity pathogenesis.


Assuntos
Apetite/genética , Apetite/fisiologia , Comportamento Alimentar/fisiologia , Animais , Índice de Massa Corporal , Encéfalo , Drosophila melanogaster/genética , Metabolismo Energético , Estudo de Associação Genômica Ampla , Genótipo , Homeostase , Hipotálamo/metabolismo , Neurônios/metabolismo , Estado Nutricional , Obesidade/metabolismo , Fenótipo
12.
Sci Rep ; 11(1): 17571, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34475432

RESUMO

Neuronatin (Nnat) has previously been reported to be part of a network of imprinted genes downstream of the chromatin regulator Trim28. Disruption of Trim28 or of members of this network, including neuronatin, results in an unusual phenotype of a bimodal body weight. To better characterise this variability, we examined the key contributors to energy balance in Nnat+/-p mice that carry a paternal null allele and do not express Nnat. Consistent with our previous studies, Nnat deficient mice on chow diet displayed a bimodal body weight phenotype with more than 30% of Nnat+/-p mice developing obesity. In response to both a 45% high fat diet and exposure to thermoneutrality (30 °C) Nnat deficient mice maintained the hypervariable body weight phenotype. Within a calorimetry system, food intake in Nnat+/-p mice was hypervariable, with some mice consuming more than twice the intake seen in wild type littermates. A hyperphagic response was also seen in Nnat+/-p mice in a second, non-home cage environment. An expected correlation between body weight and energy expenditure was seen, but corrections for the effects of positive energy balance and body weight greatly diminished the effect of neuronatin deficiency on energy expenditure. Male and female Nnat+/-p mice displayed subtle distinctions in the degree of variance body weight phenotype and food intake and further sexual dimorphism was reflected in different patterns of hypothalamic gene expression in Nnat+/-p mice. Loss of the imprinted gene Nnat is associated with a highly variable food intake, with the impact of this phenotype varying between genetically identical individuals.


Assuntos
Ingestão de Alimentos/fisiologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Obesidade/metabolismo , Animais , Biomarcadores/metabolismo , Peso Corporal , Dieta Hiperlipídica , Metabolismo Energético , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/etiologia , Obesidade/patologia
13.
Diabetologia ; 53(2): 309-20, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19902174

RESUMO

AIMS/HYPOTHESIS: Numerous new genes have recently been identified in genome-wide association studies for type 2 diabetes. Most are highly expressed in beta cells and presumably play important roles in their function. However, these genes account for only a small proportion of total risk and there are likely to be additional candidate genes not detected by current methodology. We therefore investigated islets from the polygenic New Zealand mouse (NZL) model of diet-induced beta cell dysfunction to identify novel genes and pathways that may play a role in the pathogenesis of diabetes. METHODS: NZL mice were fed a diabetogenic high-fat diet (HF) or a diabetes-protective carbohydrate-free HF diet (CHF). Pancreatic islets were isolated by laser capture microdissection (LCM) and subjected to genome-wide transcriptome analyses. RESULTS: In the prediabetic state, 2,109 islet transcripts were differentially regulated (>1.5-fold) between HF and CHF diets. Of the genes identified, 39 (e.g. Cacna1d, Chd2, Clip2, Igf2bp2, Dach1, Tspan8) correlated with data from the Diabetes Genetics Initiative and Wellcome Trust Case Control Consortium genome-wide scans for type 2 diabetes, thus validating our approach. HF diet induced early changes in gene expression associated with increased cell-cycle progression, proliferation and differentiation of islet cells, and oxidative stress (e.g. Cdkn1b, Tmem27, Pax6, Cat, Prdx4 and Txnip). In addition, pathway analysis identified oxidative phosphorylation as the predominant gene-set that was significantly upregulated in response to the diabetogenic HF diet. CONCLUSIONS/INTERPRETATION: We demonstrated that LCM of pancreatic islet cells in combination with transcriptional profiling can be successfully used to identify novel candidate genes for diabetes. Our data strongly implicate glucose-induced oxidative stress in disease progression.


Assuntos
Dieta para Diabéticos , Dieta , Regulação da Expressão Gênica , Ilhotas Pancreáticas/fisiologia , Síndrome Metabólica/genética , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Amplificação de Genes , Perfilação da Expressão Gênica , Hiperglicemia/genética , Hiperglicemia/prevenção & controle , Ilhotas Pancreáticas/citologia , Cinética , Masculino , Síndrome Metabólica/veterinária , Camundongos , Herança Multifatorial , Reação em Cadeia da Polimerase , RNA/genética , RNA/isolamento & purificação , Transcrição Gênica
14.
J Nanosci Nanotechnol ; 10(7): 4562-7, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21128457

RESUMO

This work seeks to characterize the residual stresses of titanium thin films as they are affected by various substrate temperatures during the sputtering process. The titanium thin films are deposited on silicon wafers by a RF magnetron sputter while different substrate temperatures are considered. The residual stresses are measured by both X-ray diffraction and a substrate curvature method, and consistent results are obtained by both methods. The results show that the residual stress decreases as the substrate temperature increases, in which the stress changes from tensile to compressive when the substrate temperature increases from 25 to 50 degrees C. Furthermore, the elastic modulus and hardness of the titanium thin films are tested with a nanoindenter using a standard Berkovich probe. Correlations between the residual stresses and mechanical properties measured by nanoindentation are also discussed.

15.
Cell Metab ; 32(1): 8-10, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32640247

RESUMO

In a recent issue of Cell, Orthofer et al. (2020) present a body of data from mice, flies, and humans that suggest (1) a physiological role for Alk in the normal control of energy balance across several species and (2) a possible contribution of genetic variants in or near ALK to the variability of adiposity in humans, more specifically at its lower end.


Assuntos
Obesidade , Magreza , Animais , Metabolismo Energético , Camundongos , Receptores Proteína Tirosina Quinases
16.
Mol Cell Endocrinol ; 300(1-2): 147-51, 2009 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-18840502

RESUMO

Human genetic data indicate impaired synthesis or processing of POMC results in obesity. We have used a mouse model of POMC deficiency (Pomc null) to explore the role of POMC-derived peptides in energy homeostasis. The phenotype of Pomc null mice recapitulates the clinical syndrome seen in humans congenitally lacking POMC. Loss of only one copy of the Pomc gene is sufficient to render mice susceptible to the effects of high fat feeding, emphasizing an important gene-environment interaction predisposing to obesity. Our studies indicate that POMC-derived peptides have influences on the response to a high fat diet, including a major influence on the dietary preference for fat. Pomc null mice are unusual in that obesity and hyperphagia develop in the absence of circulating glucocorticoid (GC). To investigate the interaction between GCs and the melanocortin system, we administered corticosterone to Pomc null mice. They appear hypersensitive to the adverse metabolic effects of GCs, developing hypertension, an exacerbation of both hyperphagia and obesity and a profound insulin resistance. GC treatment of Pomc null mice significantly increases the expression of the melanocortin antagonist agouti-related protein (AgRP). On-going studies in mice lacking both AgRP and Pomc will determine whether the metabolic phenotype seen with this GC therapy is due to a lack of melanocortin peptide, the unopposed action of AgRP or a combination of both.


Assuntos
Metabolismo Energético , Homeostase , Peptídeos/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Dieta , Alimentos , Preferências Alimentares , Glucocorticoides/metabolismo , Humanos , Melanocortinas/metabolismo , Hormônios Estimuladores de Melanócitos/genética , Hormônios Estimuladores de Melanócitos/metabolismo , Peptídeos/genética , Pró-Opiomelanocortina/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
17.
Endocrinology ; 149(11): 5432-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18635658

RESUMO

The metabolic effects of leptin may involve both centrally and peripherally mediated actions with a component of the central actions potentially independent of alterations in food intake. Ob/ob mice have significant abnormalities in lipid metabolism, correctable by leptin administration. We used ob/ob mice to study the relative importance of the subtypes of actions of leptin (central vs. peripheral; food intake dependent vs. independent) on lipid metabolism. Mice were treated for 3 d with leptin, either centrally [intracerebroventricular (icv)] or peripherally (ip), and compared with mice pair-fed to the leptin-treated mice (PF) and with ad libitum-fed controls (C). All treatment groups (icv, ip, PF) showed indistinguishable changes in liver weight; hepatic steatosis; hepatic lipidemic profile; and circulating free fatty acids, triglycerides, and cholesterol lipoprotein profile. Changes in the expression of genes involved in lipogenesis and fatty acid oxidation in liver, muscle, and white fat were broadly similar in ip, icv, and PF groups. Leptin (both icv and ip) stimulated expression of both mitochondrial and peroxisomal acyl-coenzyme A oxidase (liver) and peroxisomal proliferator-activated receptor-alpha (skeletal muscle) to an extent not replicated by pair feeding. Leptin had profound effects on peripheral lipid metabolism, but the majority were explained by its effects on food intake. Leptin had additional centrally mediated effects to increase the expression of a limited number of genes concerned with fatty acid oxidation. Whereas we cannot exclude direct peripheral effects of leptin on certain aspects of lipid metabolism, we were unable to detect any such effects on the parameters measured in this study.


Assuntos
Encéfalo/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Leptina/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Ácidos Graxos/sangue , Regulação da Expressão Gênica/efeitos dos fármacos , Injeções Intraperitoneais , Injeções Intraventriculares , Leptina/administração & dosagem , Metabolismo dos Lipídeos/genética , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Obesos , Proteínas Musculares/genética , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Triglicerídeos/sangue , Redução de Peso/efeitos dos fármacos , Redução de Peso/genética
18.
Endocrinology ; 148(11): 5331-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17717049

RESUMO

Complete proopiomelanocortin (POMC) deficiency causes a human syndrome of hypoadrenalism, altered skin and hair pigmentation, and severe hyperphagic obesity. Heterozygote carriers of nonsense mutations are strongly predisposed to obesity. Pomc(+/-) mice have normal body weight on a chow diet but increase food intake and become more obese than wild-type littermates when placed on a high-fat diet. To further explore the mechanisms whereby dietary fat interacts with Pomc genotype to produce obesity, we examined Pomc-null, Pomc(+/-), and wild-type mice for changes in the components of energy balance in response to provision of a high-fat diet and macronutrient preference when presented with a selection of dietary choices. In contrast to wild-type mice, Pomc null mice did not increase their resting energy expenditure or their spontaneous physical activity when given a high-fat diet. Pomc(+/-) mice increased resting energy expenditure similarly to wild types, but their increase in physical activity was significantly less than that seen in wild-type mice. In two independent experimental tests of macronutrient preference, Pomc genotype was a strong predictor of dietary fat preference with Pomc null animals choosing to eat approximately twice as much fat, but similar amounts of carbohydrate and protein, as wild-type animals. Pomc(+/-) mice showed an intermediate response. In summary, POMC-derived peptides have influences on multiple aspects of the organism's response to the presentation of high-fat diet. This includes a major influence, readily discernible even in heterozygote animals, on the dietary preference for fat.


Assuntos
Dieta Aterogênica , Gorduras na Dieta/farmacologia , Preferências Alimentares/efeitos dos fármacos , Atividade Motora/genética , Pró-Opiomelanocortina/fisiologia , Termogênese/genética , Animais , Gorduras na Dieta/administração & dosagem , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Pró-Opiomelanocortina/genética , Termogênese/efeitos dos fármacos
19.
Diabetes ; 55(12): 3366-71, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17130481

RESUMO

The neurotrophin brain-derived neurotrophic factor (BDNF) inhibits food intake, and rodent models of BDNF disruption all exhibit increased food intake and obesity, as well as hyperactivity. We report an 8-year-old girl with hyperphagia and severe obesity, impaired cognitive function, and hyperactivity who harbored a de novo chromosomal inversion, 46,XX,inv(11)(p13p15.3), a region encompassing the BDNF gene. We have identified the proximal inversion breakpoint that lies 850 kb telomeric of the 5' end of the BDNF gene. The patient's genomic DNA was heterozygous for a common coding polymorphism in BDNF, but monoallelic expression was seen in peripheral lymphocytes. Serum concentration of BDNF protein was reduced compared with age- and BMI-matched subjects. Haploinsufficiency for BDNF was associated with increased ad libitum food intake, severe early-onset obesity, hyperactivity, and cognitive impairment. These findings provide direct evidence for the role of the neurotrophin BDNF in human energy homeostasis, as well as in cognitive function, memory, and behavior.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Aberrações Cromossômicas , Inversão Cromossômica/genética , Transtornos Cognitivos/genética , Hipercinese/genética , Hiperfagia/genética , Mutação , Obesidade/genética , Adulto , Fator Neurotrófico Derivado do Encéfalo/sangue , Cromossomos Artificiais Bacterianos , DNA/genética , DNA/isolamento & purificação , Feminino , Humanos
20.
J Neuroendocrinol ; 19(9): 703-7, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17680885

RESUMO

The hypothalamic melanocortin system plays a fundamental role in the regulation of energy homeostasis. Orexins (hypocretins) are also involved in a diverse range of physiological processes, including food intake. Previous evidence has suggested that hypothalamic orexin expression may be influenced by the central melanocortin system. Here, we studied orexin mRNA levels in pro-opiomelanocortin-deficient (Pomc(-/-)) mice, a mouse model lacking all endogenously produced melanocortin peptides. Orexin expression in the lateral hypothalamus was significantly increased in corticosterone deficient Pomc(-/-) mice. Furthermore, when circulating glucocorticoids were restored to levels within the physiological range, orexin expression remained elevated. However, i.c.v. administration of the melanocortin alpha-melanocyte-stimulating hormone (MSH) to Pomc(-/-) mice reduced orexin expression back down to wild-type levels. This was independent of the effects of alpha-MSH on food intake because elevated orexin expression persisted in Pomc(-/-) mice pairfed to alpha-MSH-treated animals. These data indicate that alpha-MSH may play a role in the regulation of orexin expression in Pomc(-/-), with an elevation in orexin levels contributing to the hyperphagia seen in these animals.


Assuntos
Hormônios/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neuropeptídeos/metabolismo , alfa-MSH/metabolismo , Animais , Peso Corporal , Corticosterona/administração & dosagem , Ingestão de Alimentos , Hipotálamo/anatomia & histologia , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Knockout , Neuropeptídeos/genética , Orexinas , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro/metabolismo , alfa-MSH/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA