Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Mol Psychiatry ; 26(7): 2805-2819, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33067580

RESUMO

It is essential to elucidate brain-adipocyte interactions in order to tackle obesity and its comorbidities, as the precise control of brain-adipose tissue cross-talk is crucial for energy and glucose homeostasis. Recent studies show that in the peripheral adipose tissue, adenosine induces adipogenesis through peripheral adenosine A1 receptor (pADORA1) signaling; however, it remains unclear whether systemic and adipose tissue metabolism would also be under the control of central (c) ADORA1 signaling. Here, we use tissue-specific pharmacology and metabolic tools to clarify the roles of cADORA1 signaling in energy and adipocyte physiology. We found that cADORA1 signaling reduces body weight while also inducing adipose tissue lipolysis. cADORA1 signaling also increases adipose tissue sympathetic norepinephrine content. In contrast, pADORA1 signaling facilitates a high-fat diet-induced obesity (DIO). We propose here a novel mechanism in which cADORA1 and pADORA1 signaling hinder and aggravate DIO, respectively.


Assuntos
Tecido Adiposo , Metabolismo dos Lipídeos , Adipócitos , Tecido Adiposo/metabolismo , Peso Corporal , Encéfalo , Dieta Hiperlipídica , Metabolismo Energético , Humanos
2.
Aging Cell ; 19(11): e13257, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33146912

RESUMO

Diabetes and metabolic syndrome are associated with the typical American high glycemia diet and result in accumulation of high levels of advanced glycation end products (AGEs), particularly upon aging. AGEs form when sugars or their metabolites react with proteins. Associated with a myriad of age-related diseases, AGEs accumulate in many tissues and are cytotoxic. To date, efforts to limit glycation pharmacologically have failed in human trials. Thus, it is crucial to identify systems that remove AGEs, but such research is scanty. Here, we determined if and how AGEs might be cleared by autophagy. Our in vivo mouse and C. elegans models, in which we altered proteolysis or glycative burden, as well as experiments in five types of cells, revealed more than six criteria indicating that p62-dependent autophagy is a conserved pathway that plays a critical role in the removal of AGEs. Activation of autophagic removal of AGEs requires p62, and blocking this pathway results in accumulation of AGEs and compromised viability. Deficiency of p62 accelerates accumulation of AGEs in soluble and insoluble fractions. p62 itself is subject to glycative inactivation and accumulates as high mass species. Accumulation of p62 in retinal pigment epithelium is reversed by switching to a lower glycemia diet. Since diminution of glycative damage is associated with reduced risk for age-related diseases, including age-related macular degeneration, cardiovascular disease, diabetes, Alzheimer's, and Parkinson's, discovery of methods to limit AGEs or enhance p62-dependent autophagy offers novel potential therapeutic targets to treat AGEs-related pathologies.


Assuntos
Produtos Finais de Glicação Avançada/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Autofagia/fisiologia , Linhagem Celular , Sobrevivência Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Rim/citologia , Rim/metabolismo , Cristalino/citologia , Cristalino/metabolismo , Lisossomos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos
3.
Transl Psychiatry ; 9(1): 111, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30850579

RESUMO

Recent studies indicate that activation of hypothalamic Agouti-related protein (Agrp) neurons can increase forage-related/repetitive behavior and decrease anxiety levels. However, the impact of physiological hunger states and food deprivation on anxiety-related behaviors have not been clarified. In the present study, we evaluated changes in anxiety levels induced by physiological hunger states and food deprivation, and identified the neuron population involved. Ad libitum fed and fasted mice were tested in the open field and elevated plus-maze behavioral tests. The DREADD approach was applied to selectively inhibit and stimulate neurons expressing Agrp in hypothalamic arcuate nucleus in Agrp-Cre transgenic mice. We found that anxiety levels were significantly reduced in the late light period when mice have increased need for food and increased Agrp neurons firing, in contrast to the levels in the early light period. Consistently, we also found that anxiety was potently reduced in 24-h fasted mice, relative to 12-h fasted mice or fed ad libitum. Mechanistically, we found that chemogenetic activation of Agrp neurons reduced anxiety in fed mice, and inactivation of Agrp neurons reduced fasting-induced anxiolytic effects. Our results suggest that anxiety levels may vary physiologically with the increasing need for food, and are influenced by acute fasting in a time-dependent manner. Agrp neurons contribute to fasting-induced anxiolytic effects, supporting the notion that Agrp neuron may serve as an entry point for the treatment of energy states-related anxiety disorders.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Ansiedade , Ritmo Circadiano , Jejum/fisiologia , Vias Neurais/fisiologia , Neurônios/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/fisiologia , Ingestão de Alimentos/fisiologia , Feminino , Privação de Alimentos , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasticidade Neuronal
4.
Sci Rep ; 7(1): 15823, 2017 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-29158477

RESUMO

Sirolimus (rapamycin) is an immunosuppressive drug used in transplantation. One of its major side effects is the increased risk of diabetes mellitus; however, the exact mechanisms underlying such association have not been elucidated. Here we show that sirolimus impairs glucose-stimulated insulin secretion both in human and murine pancreatic islets and in clonal ß cells in a dose- and time-dependent manner. Importantly, we demonstrate that sirolimus markedly depletes calcium (Ca2+) content in the endoplasmic reticulum and significantly decreases glucose-stimulated mitochondrial Ca2+ uptake. Crucially, the reduced mitochondrial Ca2+ uptake is mirrored by a significant impairment in mitochondrial respiration. Taken together, our findings indicate that sirolimus causes depletion of intracellular Ca2+ stores and alters mitochondrial fitness, eventually leading to decreased insulin release. Our results provide a novel molecular mechanism underlying the increased incidence of diabetes mellitus in patients treated with this drug.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Glucose/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Sirolimo/farmacologia , Animais , Cálcio/metabolismo , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Humanos , Insulina/genética , Insulina/metabolismo , Secreção de Insulina/genética , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Sirolimo/efeitos adversos
5.
Proc Natl Acad Sci U S A ; 114(22): E4472-E4481, 2017 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-28507131

RESUMO

Age-related macular degeneration (AMD) is the major cause of blindness in developed nations. AMD is characterized by retinal pigmented epithelial (RPE) cell dysfunction and loss of photoreceptor cells. Epidemiologic studies indicate important contributions of dietary patterns to the risk for AMD, but the mechanisms relating diet to disease remain unclear. Here we investigate the effect on AMD of isocaloric diets that differ only in the type of dietary carbohydrate in a wild-type aged-mouse model. The consumption of a high-glycemia (HG) diet resulted in many AMD features (AMDf), including RPE hypopigmentation and atrophy, lipofuscin accumulation, and photoreceptor degeneration, whereas consumption of the lower-glycemia (LG) diet did not. Critically, switching from the HG to the LG diet late in life arrested or reversed AMDf. LG diets limited the accumulation of advanced glycation end products, long-chain polyunsaturated lipids, and their peroxidation end-products and increased C3-carnitine in retina, plasma, or urine. Untargeted metabolomics revealed microbial cometabolites, particularly serotonin, as protective against AMDf. Gut microbiota were responsive to diet, and we identified microbiota in the Clostridiales order as being associated with AMDf and the HG diet, whereas protection from AMDf was associated with the Bacteroidales order and the LG diet. Network analysis revealed a nexus of metabolites and microbiota that appear to act within a gut-retina axis to protect against diet- and age-induced AMDf. The findings indicate a functional interaction between dietary carbohydrates, the metabolome, including microbial cometabolites, and AMDf. Our studies suggest a simple dietary intervention that may be useful in patients to arrest AMD.


Assuntos
Glicemia/metabolismo , Microbioma Gastrointestinal/fisiologia , Índice Glicêmico/fisiologia , Degeneração Macular/metabolismo , Retina/metabolismo , Animais , Produtos Finais de Glicação Avançada/metabolismo , Metaboloma/fisiologia , Metabolômica , Camundongos
6.
Exp Ther Med ; 10(4): 1364-1374, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26622492

RESUMO

Marjolin's ulcers, which are epidermoid carcinomas arising on non-healing scar tissue, may be of various pathological types, including squamous cell carcinoma. The pathogenesis of squamous cell carcinoma arising in an ulcer differs from that of the primary cutaneous squamous cell carcinoma. This squamous cell carcinoma is aggressive in nature, and has a high rate of metastasis. Between January 2001 and September 2013, 51 patients with Marjolin's ulcers were admitted to the Departments of Plastic Surgery of the Affiliated Foshan Hospital and the Second Affiliated Hospital of Sun Yat-sen University. The ulcers included 43 cases of squamous cell carcinoma, six of melanoma, one of basal cell carcinoma and one of epithelioid sarcoma. The clinical data of these patients were retrospectively analyzed. Patients were followed until mortality. Among the patients with squamous cell carcinoma, 30.23% exhibited sentinel lymph node metastasis and 11.63% had distant metastasis. Among the patients with melanoma, 66.67% had sentinel lymph node metastasis and 33.33% had distant metastasis. Sentinel lymph node metastasis was successfully detected in 11 patients with Marjolin's ulcer using 18F-fluorodeoxyglucose positron emission tomography-computed tomography and B-mode ultrasound guided biopsy. Squamous cell carcinoma was often treated by extended resection and skin grafting or skin flap repair. Patients with deep, aggressive squamous cell carcinoma of an extremity and sentinel lymph node metastasis underwent amputation and lymph node dissection. This treatment was also used for melanoma type Marjolin's ulcers.

7.
Diabetes ; 64(9): 3273-84, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26294429

RESUMO

The assumption underlying current diabetes treatment is that lowering the level of time-averaged glucose concentrations, measured as HbA1c, prevents microvascular complications. However, 89% of variation in risk of retinopathy, microalbuminuria, or albuminuria is due to elements of glycemia not captured by mean HbA1c values. We show that transient exposure to high glucose activates a multicomponent feedback loop that causes a stable left shift of the glucose concentration-reactive oxygen species (ROS) dose-response curve. Feedback loop disruption by the GLP-1 cleavage product GLP-1(9-36)(amide) reverses the persistent left shift, thereby normalizing persistent overproduction of ROS and its pathophysiologic consequences. These data suggest that hyperglycemic spikes high enough to activate persistent ROS production during subsequent periods of normal glycemia but too brief to affect the HbA1c value are a major determinant of the 89% of diabetes complications risk not captured by HbA1c. The phenomenon and mechanism described in this study provide a basis for the development of both new biomarkers to complement HbA1c and novel therapeutic agents, including GLP-1(9-36)(amide), for the prevention and treatment of diabetes complications.


Assuntos
Complicações do Diabetes/metabolismo , Retroalimentação Fisiológica , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Glucose/metabolismo , Hiperglicemia/metabolismo , Mitocôndrias/metabolismo , Peptídeos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular , Células Endoteliais/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Hemoglobinas Glicadas/metabolismo , Humanos , Ferro/metabolismo , Potencial da Membrana Mitocondrial
8.
Atherosclerosis ; 239(2): 393-400, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25682038

RESUMO

OBJECTIVE: The pathogenic events responsible for accelerated atherosclerosis in patients with chronic renal failure (CRF) are poorly understood. Here we investigate the hypothesis that concentrations of urea associated with CRF and increased ROS production in adipocytes might also increase ROS production directly in arterial endothelial cells, causing the same pathophysiologic changes seen with hyperglycemia. METHODS: Primary cultures of human aortic endothelial cells (HAEC) were exposed to 20mM urea for 48 h. C57BL/6J wild-type mice underwent 5/6 nephrectomy or a sham operation. Randomized groups of 5/6 nephrectomized mice and their controls were also injected i.p. with a SOD/catalase mimetic (MnTBAP) for 15 days starting immediately after the final surgical procedure. RESULTS: Urea at concentrations seen in CRF induced mitochondrial ROS production in cultured HAEC. Urea-induced ROS caused the activation of endothelial pro-inflammatory pathways through the inhibition of GAPDH, including increased protein kinase C isoforms activity, increased hexosamine pathway activity, and accumulation of intracellular AGEs (advanced glycation end products). Urea-induced ROS directly inactivated the anti-atherosclerosis enzyme PGI2 synthase and also caused ER stress. Normalization of mitochondrial ROS production prevented each of these effects of urea. In uremic mice, treatment with MnTBAP prevented aortic oxidative stress, PGI2 synthase activity reduction and increased expression of the pro-inflammatory proteins TNFα, IL-6, VCAM1, Endoglin, and MCP-1. CONCLUSIONS: Taken together, these data show that urea itself, at levels common in patients with CRF, causes endothelial dysfunction and activation of proatherogenic pathways.


Assuntos
Endotélio Vascular/patologia , Falência Renal Crônica/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ureia/química , Animais , Antígenos CD/metabolismo , Aorta/metabolismo , Aterosclerose/enzimologia , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Catalase/metabolismo , Quimiocina CCL2/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Endoglina , Células Endoteliais/metabolismo , Endotélio/enzimologia , Endotélio Vascular/efeitos dos fármacos , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Humanos , Interleucina-6/metabolismo , Oxirredutases Intramoleculares/metabolismo , Falência Renal Crônica/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Distribuição Aleatória , Receptores de Superfície Celular/metabolismo , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
9.
Physiol Rep ; 2(6)2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24920125

RESUMO

The reactive dicarbonyls, glyoxal and methylglyoxal (MG), increase in diabetes and may participate in the development of diabetic complications. Glyoxal and MG are detoxified by the sequential activities of glyoxalase 1 (GLO1) and glyoxalase 2. To determine the contribution of these dicarbonyls to the etiology of complications, we have genetically manipulated GLO1 levels in apolipoprotein E-null (Apoe(-/-)) mice. Male Apoe(-/-) mice, hemizygous for a human GLO1 transgene (GLO1TGApoe(-/-) mice) or male nontransgenic Apoe(-/-) litter mates were injected with streptozotocin or vehicle and 6 or 20 weeks later, aortic atherosclerosis was quantified. The GLO1 transgene lessened streptozotocin (STZ)-induced increases in immunoreactive hydroimidazolone (MG-H1). Compared to nondiabetic mice, STZ-treated GLO1TGApoe(-/-) and Apoe(-/-) mice had increased serum cholesterol and triglycerides and increased atherosclerosis at both times after diabetes induction. While the increased GLO1 activity in the GLO1TGApoe(-/-) mice failed to protect against diabetic atherosclerosis, it lessened glomerular mesangial expansion, prevented albuminuria and lowered renal levels of dicarbonyls and protein glycation adducts. Aortic atherosclerosis was also quantified in 22-week-old, male normoglycemic Glo1 knockdown mice on an Apoe(-/-) background (Glo1KDApoe(-/-) mice), an age at which Glo1KD mice exhibit albuminuria and renal pathology similar to that of diabetic mice. In spite of ~75% decrease in GLO1 activity and increased aortic MG-H1, the Glo1KDApoe(-/-) mice did not show increased atherosclerosis compared to age-matched Apoe(-/-) mice. Thus, manipulation of GLO1 activity does not affect the development of early aortic atherosclerosis in Apoe(-/-) mice but can dictate the onset of kidney disease independently of blood glucose levels.

10.
Diabetes ; 63(1): 291-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24062246

RESUMO

Differences in susceptibility to diabetic nephropathy (DN) between mouse strains with identical levels of hyperglycemia correlate with renal levels of oxidative stress, shown previously to play a central role in the pathogenesis of DN. Susceptibility to DN appears to be genetically determined, but the critical genes have not yet been identified. Overexpression of the enzyme glyoxalase 1 (Glo1), which prevents posttranslational modification of proteins by the glycolysis-derived α-oxoaldehyde, methylglyoxal (MG), prevents hyperglycemia-induced oxidative stress in cultured cells and model organisms. In this study, we show that in nondiabetic mice, knockdown of Glo1 increases to diabetic levels both MG modification of glomerular proteins and oxidative stress, causing alterations in kidney morphology indistinguishable from those caused by diabetes. We also show that in diabetic mice, Glo1 overexpression completely prevents diabetes-induced increases in MG modification of glomerular proteins, increased oxidative stress, and the development of diabetic kidney pathology, despite unchanged levels of diabetic hyperglycemia. Together, these data indicate that Glo1 activity regulates the sensitivity of the kidney to hyperglycemic-induced renal pathology and that alterations in the rate of MG detoxification are sufficient to determine the glycemic set point at which DN occurs.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Hiperglicemia/metabolismo , Rim/metabolismo , Lactoilglutationa Liase/metabolismo , Albuminúria/genética , Albuminúria/metabolismo , Albuminúria/fisiopatologia , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/fisiopatologia , Hiperglicemia/genética , Hiperglicemia/fisiopatologia , Rim/fisiopatologia , Lactoilglutationa Liase/genética , Camundongos , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo
11.
J Neurosci ; 32(40): 13701-8, 2012 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23035082

RESUMO

Glucagon-like peptide-1 (GLP-1) is an endogenous intestinal peptide that enhances glucose-stimulated insulin secretion. Its natural cleavage product GLP-1(9-36)(amide) possesses distinct properties and does not affect insulin secretion. Here we report that pretreatment of hippocampal slices with GLP-1(9-36)(amide) prevented impaired long-term potentiation (LTP) and enhanced long-term depression induced by exogenous amyloid ß peptide Aß((1-42)). Similarly, hippocampal LTP impairments in amyloid precursor protein/presenilin 1 (APP/PS1) mutant mice that model Alzheimer's disease (AD) were prevented by GLP-1(9-36)(amide). In addition, treatment of APP/PS1 mice with GLP-1(9-36)(amide) at an age at which they display impaired spatial and contextual fear memory resulted in a reversal of their memory defects. At the molecular level, GLP-1(9-36)(amide) reduced elevated levels of mitochondrial-derived reactive oxygen species and restored dysregulated Akt-glycogen synthase kinase-3ß signaling in the hippocampus of APP/PS1 mice. Our findings suggest that GLP-1(9-36)(amide) treatment may have therapeutic potential for AD and other diseases associated with cognitive dysfunction.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Antioxidantes/uso terapêutico , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Transtornos da Memória/tratamento farmacológico , Plasticidade Neuronal/efeitos dos fármacos , Nootrópicos/uso terapêutico , Peptídeos/uso terapêutico , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/farmacologia , Animais , Antioxidantes/farmacologia , Aprendizagem por Associação/efeitos dos fármacos , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/metabolismo , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Medo , Feminino , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Quinase 3 da Glicogênio Sintase/fisiologia , Glicogênio Sintase Quinase 3 beta , Masculino , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Nootrópicos/farmacologia , Compostos Organofosforados/farmacologia , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Presenilina-1/genética , Proteínas Proto-Oncogênicas c-akt/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ubiquinona/análogos & derivados , Ubiquinona/farmacologia
12.
Braz. j. med. biol. res ; 45(7): 656-664, July 2012. ilus, tab
Artigo em Inglês | LILACS | ID: lil-639460

RESUMO

The objective was to elucidate the relationships between serum concentrations of the gut hormone peptide YY (PYY) and ghrelin and growth development in infants for potential application to the clinical observation index. Serum concentrations of PYY and ghrelin were measured using radioimmunoassay from samples collected at the clinic. For each patient, gestational age, birth weight, time required to return to birth weight, rate of weight gain, time required to achieve recommended daily intake (RDI) standards, time required for full-gastric feeding, duration of hospitalization, and time of administration of total parenteral nutrition were recorded. Serum PYY and ghrelin concentrations were significantly higher in the preterm group (N = 20) than in the full-term group (N = 20; P < 0.01). Within the preterm infant group, the serum concentrations of PYY and ghrelin on postnatal day (PND) 7 (ghrelin = 1485.38 ± 409.24; PYY = 812.37 ± 153.77 ng/L) were significantly higher than on PND 1 (ghrelin = 956.85 ± 223.09; PYY = 545.27 ± 204.51 ng/L) or PND 3 (ghrelin = 1108.44 ± 351.36; PYY = 628.96 ± 235.63 ng/L; P < 0.01). Both serum PYY and ghrelin concentrations were negatively correlated with body weight, and the degree of correlation varied with age. Serum ghrelin concentration correlated negatively with birth weight and positively with the time required to achieve RDI (P < 0.05). In conclusion, serum PYY and ghrelin concentrations reflect a negative energy balance, predict postnatal growth, and enable compensation. Further studies are required to elucidate the precise concentration and roles of PYY and ghrelin in newborns and to determine the usefulness of measuring these hormones in clinical practice.


Assuntos
Feminino , Humanos , Recém-Nascido , Masculino , Peso Corporal/fisiologia , Ingestão de Energia/fisiologia , Grelina/sangue , Recém-Nascido Prematuro/fisiologia , Necessidades Nutricionais/fisiologia , Peptídeo YY/sangue , Aumento de Peso/fisiologia , Estudos de Casos e Controles , Radioimunoensaio
13.
Braz J Med Biol Res ; 45(7): 656-64, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22527125

RESUMO

The objective was to elucidate the relationships between serum concentrations of the gut hormone peptide YY (PYY) and ghrelin and growth development in infants for potential application to the clinical observation index. Serum concentrations of PYY and ghrelin were measured using radioimmunoassay from samples collected at the clinic. For each patient, gestational age, birth weight, time required to return to birth weight, rate of weight gain, time required to achieve recommended daily intake (RDI) standards, time required for full-gastric feeding, duration of hospitalization, and time of administration of total parenteral nutrition were recorded. Serum PYY and ghrelin concentrations were significantly higher in the preterm group (N = 20) than in the full-term group (N = 20; P < 0.01). Within the preterm infant group, the serum concentrations of PYY and ghrelin on postnatal day (PND) 7 (ghrelin = 1485.38 ± 409.24; PYY = 812.37 ± 153.77 ng/L) were significantly higher than on PND 1 (ghrelin = 956.85 ± 223.09; PYY = 545.27 ± 204.51 ng/L) or PND 3 (ghrelin = 1108.44 ± 351.36; PYY = 628.96 ± 235.63 ng/L; P < 0.01). Both serum PYY and ghrelin concentrations were negatively correlated with body weight, and the degree of correlation varied with age. Serum ghrelin concentration correlated negatively with birth weight and positively with the time required to achieve RDI (P < 0.05). In conclusion, serum PYY and ghrelin concentrations reflect a negative energy balance, predict postnatal growth, and enable compensation. Further studies are required to elucidate the precise concentration and roles of PYY and ghrelin in newborns and to determine the usefulness of measuring these hormones in clinical practice.


Assuntos
Peso Corporal/fisiologia , Ingestão de Energia/fisiologia , Grelina/sangue , Recém-Nascido Prematuro/fisiologia , Necessidades Nutricionais/fisiologia , Peptídeo YY/sangue , Aumento de Peso/fisiologia , Estudos de Casos e Controles , Feminino , Humanos , Recém-Nascido , Masculino , Radioimunoensaio
14.
Aging Cell ; 9(3): 420-32, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20346071

RESUMO

Deletions in mitochondrial DNA (mtDNA) accumulate during aging. Expression of the Caenorhabditis elegans apurinic/apyrimidinic endonuclease 1 (APE1) ortholog exo-3, involved in DNA repair, is reduced by 45% (P < 0.05) during aging of C. elegans. Suppression of exo-3 by treatment with RNAi resulted in a threefold increase in mtDNA deletions (P < 0.05), twofold enhanced generation of reactive oxygen species (ROS) (P < 0.01), distortion of the structural integrity of the nervous system, reduction of head motility by 43% (P < 0.01) and whole animal motility by 38% (P < 0.05). Suppression of exo-3 significantly reduced life span: mean life span decreased from 18.5 +/- 0.4 to 15.4 +/- 0.1 days (P < 0.001) and maximum life span from 25.9 +/- 0.4 to 23.2 +/- 0.1 days (P = 0.001). Additional treatment of exo-3-suppressed animals with a mitochondrial uncoupler decreased ROS levels, reduced neuronal damage, and increased motility and life span. Additional suppression of the C. elegans p53 ortholog cep-1 in exo-3 RNAi-treated animals similarly decreased ROS levels, preserved neuronal integrity, and increased motility and life span. In wild-type animals, suppression of cep-1, involved in downregulation of exo-3, increased expression of exo-3 without a significant effect on ROS levels, preserved neuronal integrity, and increased motility and life span. Suppression of the C. elegans thioredoxin orthologs trx-1 and trx-2, involved in the redox chaperone activity of exo-3, overrides the protective effect of cep-1 RNAi treatment on neuronal integrity, neuronal function, mean and maximum life span. These results show that APE1/EXO-3, p53/CEP-1, and thioredoxin affect each other and that these interactions determine aging as well as neuronal structure and function.


Assuntos
Envelhecimento , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Tiorredoxinas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , DNA Mitocondrial/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Deleção de Genes , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Tiorredoxinas/genética , Proteína Supressora de Tumor p53/genética
15.
J Clin Invest ; 120(1): 203-13, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19955654

RESUMO

Although supraphysiological concentrations of urea are known to increase oxidative stress in cultured cells, it is generally thought that the elevated levels of urea in chronic renal failure patients have negligible toxicity. We previously demonstrated that ROS increase intracellular protein modification by O-linked beta-N-acetylglucosamine (O-GlcNAc), and others showed that increased modification of insulin signaling molecules by O-GlcNAc reduces insulin signal transduction. Because both oxidative stress and insulin resistance have been observed in patients with end-stage renal disease, we sought to determine the role of urea in these phenotypes. Treatment of 3T3-L1 adipocytes with urea at disease-relevant concentrations induced ROS production, caused insulin resistance, increased expression of adipokines retinol binding protein 4 (RBP4) and resistin, and increased O-GlcNAc-modified insulin signaling molecules. Investigation of a mouse model of surgically induced renal failure (uremic mice) revealed increased ROS production, modification of insulin signaling molecules by O-GlcNAc, and increased expression of RBP4 and resistin in visceral adipose tissue. Uremic mice also displayed insulin resistance and glucose intolerance, and treatment with an antioxidant SOD/catalase mimetic normalized these defects. The SOD/catalase mimetic treatment also prevented the development of insulin resistance in normal mice after urea infusion. These data suggest that therapeutic targeting of urea-induced ROS may help reduce the high morbidity and mortality caused by end-stage renal disease.


Assuntos
Resistência à Insulina , Falência Renal Crônica/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ureia/sangue , Células 3T3-L1 , Animais , Glicemia/análise , Glucose/metabolismo , Intolerância à Glucose/etiologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Fosforilação , Resistina/sangue , Proteínas Plasmáticas de Ligação ao Retinol/análise
16.
Diabetes ; 58(11): 2450-6, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19675139

RESUMO

OBJECTIVE: Establishing Caenorhabditis elegans as a model for glucose toxicity-mediated life span reduction. RESEARCH DESIGN AND METHODS: C. elegans were maintained to achieve glucose concentrations resembling the hyperglycemic conditions in diabetic patients. The effects of high glucose on life span, glyoxalase-1 activity, advanced glycation end products (AGEs), and reactive oxygen species (ROS) formation and on mitochondrial function were studied. RESULTS: High glucose conditions reduced mean life span from 18.5 + or - 0.4 to 16.5 + or - 0.6 days and maximum life span from 25.9 + or - 0.4 to 23.2 + or - 0.4 days, independent of glucose effects on cuticle or bacterial metabolization of glucose. The formation of methylglyoxal-modified mitochondrial proteins and ROS was significantly increased by high glucose conditions and reduced by mitochondrial uncoupling and complex IIIQo inhibition. Overexpression of the methylglyoxal-detoxifying enzyme glyoxalase-1 attenuated the life-shortening effect of glucose by reducing AGE accumulation (by 65%) and ROS formation (by 50%) and restored mean (16.5 + or - 0.6 to 20.6 + or - 0.4 days) and maximum life span (23.2 + or - 0.4 to 27.7 + or - 2.3 days). In contrast, inhibition of glyoxalase-1 by RNAi further reduced mean (16.5 + or - 0.6 to 13.9 + or - 0.7 days) and maximum life span (23.2 + or - 0.4 to 20.3 + or - 1.1 days). The life span reduction by glyoxalase-1 inhibition was independent from the insulin signaling pathway because high glucose conditions also affected daf-2 knockdown animals in a similar manner. CONCLUSIONS: C. elegans is a suitable model organism to study glucose toxicity, in which high glucose conditions limit the life span by increasing ROS formation and AGE modification of mitochondrial proteins in a daf-2 independent manner. Most importantly, glucose toxicity can be prevented by improving glyoxalase-1-dependent methylglyoxal detoxification or preventing mitochondrial dysfunction.


Assuntos
Caenorhabditis elegans/metabolismo , Glucose/toxicidade , Hiperglicemia/metabolismo , Longevidade/fisiologia , Animais , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/ultraestrutura , Modelos Animais de Doenças , Humanos , Expectativa de Vida/tendências , Microscopia Eletrônica de Varredura , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo
17.
Proc Natl Acad Sci U S A ; 106(32): 13505-10, 2009 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-19666581

RESUMO

Diabetes is associated with poor outcomes following acute vascular occlusive events. This results in part from a failure to form adequate compensatory microvasculature in response to ischemia. Since vascular endothelial growth factor (VEGF) is an essential mediator of neovascularization, we examined whether hypoxic up-regulation of VEGF was impaired in diabetes. Both fibroblasts isolated from type 2 diabetic patients, and normal fibroblasts exposed chronically to high glucose, were defective in their capacity to up-regulate VEGF in response to hypoxia. In vivo, diabetic animals demonstrated an impaired ability to increase VEGF production in response to soft tissue ischemia. This resulted from a high glucose-induced decrease in transactivation by the transcription factor hypoxia-inducible factor-1alpha (HIF-1alpha), which mediates hypoxia-stimulated VEGF expression. Decreased HIF-1alpha functional activity was specifically caused by impaired HIF-1alpha binding to the coactivator p300. We identify covalent modification of p300 by the dicarbonyl metabolite methylglyoxal as being responsible for this decreased association. Administration of deferoxamine abrogated methylglyoxal conjugation, normalizing both HIF-1alpha/p300 interaction and transactivation by HIF-1alpha. In diabetic mice, deferoxamine promoted neovascularization and enhanced wound healing. These findings define molecular defects that underlie impaired VEGF production in diabetic tissues and offer a promising direction for therapeutic intervention.


Assuntos
Complicações do Diabetes/metabolismo , Complicações do Diabetes/patologia , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Hipóxia/complicações , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Desferroxamina/farmacologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Modelos Animais de Doenças , Glucose/farmacologia , Humanos , Hiperglicemia/complicações , Hiperglicemia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Neovascularização Patológica/complicações , Neovascularização Patológica/patologia , Ligação Proteica/efeitos dos fármacos , Aldeído Pirúvico/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Ativação Transcricional/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Fatores de Transcrição de p300-CBP/metabolismo
18.
Aging Cell ; 7(2): 260-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18221415

RESUMO

Studies of mutations affecting lifespan in Caenorhabditis elegans show that mitochondrial generation of reactive oxygen species (ROS) plays a major causative role in organismal aging. Here, we describe a novel mechanism for regulating mitochondrial ROS production and lifespan in C. elegans: progressive mitochondrial protein modification by the glycolysis-derived dicarbonyl metabolite methylglyoxal (MG). We demonstrate that the activity of glyoxalase-1, an enzyme detoxifying MG, is markedly reduced with age despite unchanged levels of glyoxalase-1 mRNA. The decrease in enzymatic activity promotes accumulation of MG-derived adducts and oxidative stress markers, which cause further inhibition of glyoxalase-1 expression. Over-expression of the C. elegans glyoxalase-1 orthologue CeGly decreases MG modifications of mitochondrial proteins and mitochondrial ROS production, and prolongs C. elegans lifespan. In contrast, knock-down of CeGly increases MG modifications of mitochondrial proteins and mitochondrial ROS production, and decreases C. elegans lifespan.


Assuntos
Caenorhabditis elegans/enzimologia , Lactoilglutationa Liase/biossíntese , Lactoilglutationa Liase/deficiência , Longevidade , Mitocôndrias/enzimologia , Modificação Traducional de Proteínas , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Inibidores Enzimáticos , Retroalimentação Fisiológica , Expressão Gênica , Lactoilglutationa Liase/antagonistas & inibidores , Proteínas Mitocondriais/metabolismo , Mutação , Fosforilação Oxidativa , Estresse Oxidativo , Aldeído Pirúvico/metabolismo , RNA de Helmintos , RNA Mensageiro/análise , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo
19.
J Clin Invest ; 116(4): 1071-80, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16528409

RESUMO

Insulin resistance markedly increases cardiovascular disease risk in people with normal glucose tolerance, even after adjustment for known risk factors such as LDL, triglycerides, HDL, and systolic blood pressure. In this report, we show that increased oxidation of FFAs in aortic endothelial cells without added insulin causes increased production of superoxide by the mitochondrial electron transport chain. FFA-induced overproduction of superoxide activated a variety of proinflammatory signals previously implicated in hyperglycemia-induced vascular damage and inactivated 2 important antiatherogenic enzymes, prostacyclin synthase and eNOS. In 2 nondiabetic rodent models--insulin-resistant, obese Zucker (fa/fa) rats and high-fat diet-induced insulin-resistant mice--inactivation of prostacyclin synthase and eNOS was prevented by inhibition of FFA release from adipose tissue; by inhibition of the rate-limiting enzyme for fatty acid oxidation in mitochondria, carnitine palmitoyltransferase I; and by reduction of superoxide levels. These studies identify what we believe to be a novel mechanism contributing to the accelerated atherogenesis and increased cardiovascular disease risk occurring in people with insulin resistance.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Resistência à Insulina/fisiologia , Oxirredutases Intramoleculares/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Animais , Aorta , Carnitina O-Palmitoiltransferase/metabolismo , Proteínas de Transporte/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Canais Iônicos , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais , Modelos Biológicos , Oxirredução , Ratos , Ratos Zucker , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo , Proteína Desacopladora 1
20.
J Biol Chem ; 280(6): 4617-26, 2005 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-15536073

RESUMO

Hyperglycemia is a major independent risk factor for diabetic macrovascular disease. The consequences of exposure of endothelial cells to hyperglycemia are well established. However, little is known about how adipocytes respond to both acute as well as chronic exposure to physiological levels of hyperglycemia. Here, we analyze adipocytes exposed to hyperglycemia both in vitro as well as in vivo. Comparing cells differentiated at 4 mm to cells differentiated at 25 mm glucose (the standard differentiation protocol) reveals severe insulin resistance in cells exposed to 25 mm glucose. A global assessment of transcriptional changes shows an up-regulation of a number of mitochondrial proteins. Exposure to hyperglycemia is associated with a significant induction of reactive oxygen species (ROS), both in vitro as well as in vivo in adipocytes isolated from streptozotocin-treated hyperglycemic mice. Furthermore, hyperglycemia for a few hours in a clamped setting will trigger the induction of a pro-inflammatory response in adipose tissue from rats that can effectively be reduced by co-infusion of N-acetylcysteine (NAC). ROS levels in 3T3-L1 adipocytes can be reduced significantly with pharmacological agents that lower the mitochondrial membrane potential, or by overexpression of uncoupling protein 1 or superoxide dismutase. In parallel with ROS, interleukin-6 secretion from adipocytes is significantly reduced. On the other hand, treatments that lead to a hyperpolarization of the mitochondrial membrane, such as overexpression of the mitochondrial dicarboxylate carrier result in increased ROS formation and decreased insulin sensitivity, even under normoglycemic conditions. Combined, these results highlight the importance ROS production in adipocytes and the associated insulin resistance and inflammatory response.


Assuntos
Adipócitos/patologia , Desoxiguanosina/análogos & derivados , Hiperglicemia/metabolismo , Células 3T3-L1 , 8-Hidroxi-2'-Desoxiguanosina , Acetilcisteína/química , Adenoviridae/genética , Adipócitos/metabolismo , Animais , Northern Blotting , Diferenciação Celular , Desoxiguanosina/química , Diabetes Mellitus Experimental , Transportadores de Ácidos Dicarboxílicos/metabolismo , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica , Glucose/metabolismo , Immunoblotting , Inflamação , Insulina/metabolismo , Interleucina-6/metabolismo , Potenciais da Membrana , Camundongos , Mitocôndrias/metabolismo , Estresse Oxidativo , Oxigênio/metabolismo , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estreptozocina/farmacologia , Fatores de Tempo , Transcrição Gênica , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...