Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2783: 93-107, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38478227

RESUMO

Murine models of obesity or reduced adiposity are a valuable resource for understanding the role of adipocyte dysfunction in metabolic disorders. Adipose tissue stromal vascular cells or primary adipocytes derived from murine adipose tissue and grown in culture are essential tools for studying the mechanisms underlying adipocyte development and function. Herein, we describe methods for the isolation, expansion, and long-term storage of murine adipose-derived stromal/stem cells, along with protocols for inducing adipogenesis to white or beige adipocytes in this cell population and osteogenic differentiation. Isolation of the adipose stromal vascular fraction cells for flow cytometric analysis is also described.


Assuntos
Adipogenia , Adiposidade , Camundongos , Humanos , Animais , Citometria de Fluxo/métodos , Osteogênese , Adipócitos , Tecido Adiposo , Diferenciação Celular , Obesidade/metabolismo , Células-Tronco
2.
Metabolism ; 97: 40-49, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31129047

RESUMO

BACKGROUND: Metabolic flexibility can be assessed by changes in respiratory exchange ratio (RER) following feeding. Though metabolic flexibility (difference in RER between fasted and fed state) is often impaired in individuals with obesity or type 2 diabetes, the cellular processes contributing to this impairment are unclear. MATERIALS AND METHODS: From several clinical studies we identified the 16 most and 14 least metabolically flexible male and female subjects out of >100 participants based on differences between 24-hour and sleep RER measured in a whole-room indirect calorimeter. Global skeletal muscle gene expression profiles revealed that, in metabolically flexible subjects, transcripts regulated by the RNA binding protein, HuR, are enriched. We generated and characterized mice with a skeletal muscle-specific knockout of the HuR encoding gene, Elavl1 (HuRm-/-). RESULTS: Male, but not female, HuRm-/- mice exhibit metabolic inflexibility, with mild obesity, impaired glucose tolerance, impaired fat oxidation and decreased in vitro palmitate oxidation compared to HuRfl/fl littermates. Expression levels of genes involved in mitochondrial fatty acid oxidation and oxidative phosphorylation are decreased in both mouse and human muscle when HuR is inhibited. CONCLUSIONS: HuR inhibition results in impaired metabolic flexibility and decreased lipid oxidation, suggesting a role for HuR as an important regulator of skeletal muscle metabolism.


Assuntos
Proteína Semelhante a ELAV 1/metabolismo , Músculo Esquelético/metabolismo , Proteínas de Ligação a RNA/metabolismo , Roedores/metabolismo , Adulto , Animais , Diabetes Mellitus Tipo 2/metabolismo , Jejum/metabolismo , Ácidos Graxos/metabolismo , Feminino , Intolerância à Glucose/metabolismo , Humanos , Metabolismo dos Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Obesidade/metabolismo , Oxirredução , Fosforilação Oxidativa , Troca Gasosa Pulmonar/fisiologia
3.
Process Biochem ; 59: 312-320, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28966553

RESUMO

Understanding the process of adipogenesis is critical if suitable therapeutics for obesity and related metabolic diseases are to be found. The current study presents proof of feasibility of creating a 3-D spheroid model using human adipose-derived stem cells (hASCs) and their subsequent adipogenic differentiation. hASC spheroids were formed atop an elastin-like polypeptide-polyethyleneimine (ELP-PEI) surface and differentiated using an adipogenic cocktail. Spheroids were matured in the presence of dietary fatty acids (linoleic or oleic acid) and evaluated based on functional markers including intracellular protein, CD36 expression, triglyceride accumulation, and PPAR-γ gene expression. Spheroid size was found to increase as the hASCs matured in the adipocyte maintenance medium, though the fatty acid treatment generally resulted in smaller spheroids compared to control. A stable protein content over the 10-day maturation period indicated contact-inhibited proliferation as well as minimal loss of spheroids during culture. Spheroids treated with fatty acids showed greater amounts of intracellular triglyceride content and greater expression of the key adipogenic gene, PPAR-γ. We also demonstrated that 3-D spheroids outperformed 2-D monolayer cultures in adipogenesis. We then compared the adipogenesis of hASC spheroids to that in 3T3-L1 spheroids and found that the triglyceride accumulation was less profound in hASC spheroids than that in 3T3-L1 adipocytes, correlated with smaller average spheroids, suggesting a relatively slower differentiation process. Taken together, we have shown the feasibility of adipogenic differentiation of patient-derived hASC spheroids, which with further development, may help elucidate key features in the adipogenesis process.

4.
Curr Pharm Des ; 23(25): 3645-3657, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28677505

RESUMO

BACKGROUND: The gp130 cytokine, oncostatin M (OSM), serves several physiological and pathological functions. At the molecular level, OSM can directly or indirectly participate in tumorigenesis and insulin resistance development. Although OSM was initially found to be anti-proliferative in tumors, numerous tumorigenic roles for OSM have been reported in a variety of cancers. In metabolic diseases, OSM signaling may be required for homeostasis in both the liver and the adipose tissue, since abrogation of OSM signaling causes obesity, hepatic steatosis, and insulin resistance. This review aims to: 1) examine the current literature regarding the role of OSM in the development of cancers and insulin resistance; and 2) propose a possible link between cancerassociated OSM and the development of the insulin resistance observed with cancer cachexia. CONCLUSION: In light of the potential links between cancer-associated OSM and cachexia-related insulin resistance, additional research is needed, especially given the possible link between these disease states. When considering OSM as a pharmaceutical target, its tumorigenic effects and role in tissue homeostasis must be carefully considered.


Assuntos
Antineoplásicos/metabolismo , Resistência à Insulina/fisiologia , Neoplasias/metabolismo , Oncostatina M/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Receptor gp130 de Citocina/antagonistas & inibidores , Receptor gp130 de Citocina/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Oncostatina M/farmacologia , Oncostatina M/uso terapêutico
5.
Biochim Biophys Acta ; 1849(6): 637-52, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25882704

RESUMO

Enhanced leukocytic infiltration into pancreatic islets contributes to inflammation-based diminutions in functional ß-cell mass. Insulitis (aka islet inflammation), which can be present in both T1DM and T2DM, is one factor influencing pancreatic ß-cell death and dysfunction. IL-1ß, an inflammatory mediator in both T1DM and T2DM, acutely (within 1h) induced expression of the CCL20 gene in rat and human islets and clonal ß-cell lines. Transcriptional induction of CCL20 required the p65 subunit of NF-κB to replace the p50 subunit at two functional κB sites within the CCL20 proximal gene promoter. The NF-κB p50 subunit prevents CCL20 gene expression during unstimulated conditions and overexpression of p50 reduces CCL20, but enhances cyclooxygenase-2 (COX-2), transcript accumulation after exposure to IL-1ß. We also identified differential recruitment of specific co-activator molecules to the CCL20 gene promoter, when compared with the CCL2 and COX2 genes, revealing distinct transcriptional requirements for individual NF-κB responsive genes. Moreover, IL-1ß, TNF-α and IFN-γ individually increased the expression of CCR6, the receptor for CCL20, on the surface of human neutrophils. We further found that the chemokine CCL20 is elevated in serum from both genetically obese db/db mice and in C57BL6/J mice fed a high-fat diet. Taken together, these results are consistent with a possible activation of the CCL20-CCR6 axis in diseases with inflammatory components. Thus, interfering with this signaling pathway, either at the level of NF-κB-mediated chemokine production, or downstream receptor activation, could be a potential therapeutic target to offset inflammation-associated tissue dysfunction in obesity and diabetes.


Assuntos
Quimiocina CCL20/genética , Diabetes Mellitus/genética , Inflamação/genética , Obesidade/genética , Fator de Transcrição RelA/genética , Animais , Quimiocina CCL20/biossíntese , Quimiocina CCL20/metabolismo , Diabetes Mellitus/patologia , Humanos , Imunidade Inata/genética , Inflamação/patologia , Resistência à Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos , Camundongos Obesos , NF-kappa B/genética , Obesidade/metabolismo , Obesidade/fisiopatologia , Ratos , Receptores CCR6/genética , Transdução de Sinais/genética , Fator de Transcrição RelA/biossíntese , Fator de Transcrição RelA/metabolismo
6.
Am J Physiol Renal Physiol ; 306(8): F896-906, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24500687

RESUMO

Albuminuria is associated with metabolic syndrome and diabetes. It correlates with the progression of chronic kidney disease, particularly with tubular atrophy. The fatty acid load on albumin significantly increases in obesity, presenting a proinflammatory environment to the proximal tubules. However, little is known about changes in the redox milieu during fatty acid overload and how redox-sensitive mechanisms mediate cell death. Here, we show that albumin with fatty acid impurities or conjugated with palmitate but not albumin itself compromised mitochondrial and cell viability, membrane potential and respiration. Fatty acid overload led to a redox imbalance which deactivated the antioxidant protein peroxiredoxin 2 and caused a peroxide-mediated apoptosis through the redox-sensitive pJNK/caspase-3 pathway. Transfection of tubular cells with peroxiredoxin 2 was protective and mitigated apoptosis. Mitochondrial fatty acid entry and ceramide synthesis modulators suggested that mitochondrial ß oxidation but not ceramide synthesis may modulate lipotoxic effects on tubular cell survival. These results suggest that albumin overloaded with fatty acids but not albumin itself changes the redox environment in the tubules, inducing a peroxide-mediated redox-sensitive apoptosis. Thus, mitigating circulating fatty acid levels may be an important factor in both preserving redox balance and preventing tubular cell damage in proteinuric diseases.


Assuntos
Albuminas/metabolismo , Apoptose/efeitos dos fármacos , Ácidos Graxos/farmacologia , Albuminas/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/patologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Oxirredução , Palmitatos/metabolismo , Peroxirredoxinas/metabolismo , Ratos , Soroalbumina Bovina/farmacologia
7.
J Biol Chem ; 289(9): 5960-9, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24391115

RESUMO

Lipocalin-2 (LCN2) is secreted from adipocytes, and its expression is up-regulated in obese and diabetic mice and humans. LCN2 expression and secretion have been shown to be induced by two proinflammatory cytokines, IFNγ and TNFα, in cultured murine and human adipocytes. In these studies, we demonstrated that IFNγ and TNFα induced LCN2 expression and secretion in vivo. Although we observed a strong induction of LCN2 expression and secretion from white adipose tissue (WAT) depots, the induction of LCN2 varied among different insulin-sensitive tissues. Knockdown experiments also demonstrated that STAT1 is required for IFNγ-induced lipocalin-2 expression in murine adipocytes. Similarly, knockdown of p65 in adipocytes demonstrated the necessity of the NF-κB signaling pathway for TNFα-mediated effects on LCN2. Activation of ERKs by IFNγ and TNFα also affected STAT1 and NF-κB signaling through modulation of serine phosphorylation. ERK activation-induced serine phosphorylation of both STAT1 and p65 mediated the additive effects of IFNγ and TNFα on LCN2 expression. Our results suggest that these same mechanisms occur in humans as we observed STAT1 and NF-κB binding to the human LCN2 promoter in chromatin immunoprecipitation assays performed in human fat cells. These studies substantially increase our knowledge regarding the requirements and mechanisms used by proinflammatory cytokines to induce LCN2 expression.


Assuntos
Proteínas de Fase Aguda/biossíntese , Adipócitos/metabolismo , Tecido Adiposo Branco/metabolismo , Regulação da Expressão Gênica/fisiologia , Lipocalinas/biossíntese , Proteínas Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Elementos de Resposta/fisiologia , Células 3T3-L1 , Proteínas de Fase Aguda/genética , Adipócitos/citologia , Tecido Adiposo Branco/citologia , Animais , Técnicas de Silenciamento de Genes , Humanos , Interferon gama/genética , Interferon gama/metabolismo , Lipocalina-2 , Lipocalinas/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Proteínas Oncogênicas/genética , Proteínas Proto-Oncogênicas/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
8.
PLoS One ; 7(10): e46568, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056347

RESUMO

Recent evidence suggests that tumor necrosis factor alpha (TNF) and angiotensin II (ANGII) induce oxidative stress contribute to cardiovascular disease progression. Here, we examined whether an interaction between TNF and ANGII contributes to altered cardiac mitochondrial biogenesis and ATP production to cause cardiac damage in rats. Rats received intraperitoneal injections of TNF (30 µg/kg), TNF + losartan (LOS, 1 mg/kg), or vehicle for 5 days. Left ventricular (LV) function was measured using echocardiography. Rats were sacrificed and LV tissues removed for gene expression, electron paramagnetic resonance and mitochondrial assays. TNF administration significantly increased expression of the NADPH oxidase subunit, gp91phox, and the angiotensin type 1 receptor (AT-1R) and decreased eNOS in the LV of rats. Rats that received TNF only had increased production rates of superoxide, peroxynitrite and total reactive oxygen species (ROS) in the cytosol and increased production rates of superoxide and hydrogen peroxide in mitochondria. Decreased activities of mitochondrial complexes I, II, and III and mitochondrial genes were observed in rats given TNF. In addition, TNF administration also resulted in a decrease in fractional shortening and an increase in Tei index, suggesting diastolic dysfunction. TNF administration with concomitant LOS treatment attenuated mitochondrial damage, restored cardiac function, and decreased expression of AT1-R and NADPH oxidase subunits. Mitochondrial biogenesis and function is severely impaired by TNF as evidenced by downregulation of mitochondrial genes and increased free radical production, and may contribute to cardiac damage. These defects are independent of the downregulation of mitochondrial gene expression, suggesting novel mechanisms for mitochondrial dysfunction in rats given TNF.


Assuntos
Angiotensina II/metabolismo , Mitocôndrias Cardíacas/metabolismo , Miocárdio/patologia , Estresse Oxidativo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Sequência de Bases , Primers do DNA , Espectroscopia de Ressonância de Spin Eletrônica , Masculino , Ratos , Ratos Sprague-Dawley
9.
Antioxid Redox Signal ; 16(2): 139-52, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21895524

RESUMO

AIMS: Exercise training (ExT) is a recommended adjunct to many pharmaceutical antihypertensive therapies. The effects of chronic ExT on the development of hypertension-induced renal injury remain unknown. We examined whether ExT would preserve renal hemodynamics and structure in the spontaneously hypertensive rat (SHR), and whether these effects were mediated by improved redox status and decreased inflammation. Normotensive WKY rats and SHR underwent moderate-intensity ExT for 16 weeks. One group of SHR animals was treated with hydralazine to investigate the pressure-dependent/independent effects of ExT. Acute renal clearance experiments were performed prior to sacrifice. Tissue free radical production rates were measured by electron paramagnetic resonance; gene and protein expression were measured by real time RT-PCR and Western blot or immunofluorescence, respectively. Plasma angiotensin II levels and kidney antioxidants were assessed. Training efficacy was assessed by citrate synthase activity assay in hind-limb muscle. RESULTS: ExT delayed hypertension, prevented oxidative stress and inflammation, preserved antioxidant status, prevented an increase in circulating AngII levels, and preserved renal hemodynamics and structure in SHR. In addition, exercise-induced effects, at least, in part, were found to be pressure-independent. INNOVATION: This study is the first to provide mechanistic evidence for the renoprotective benefits of ExT in a model of hypertension. Our results demonstrate that initiation of ExT in susceptible patients can delay the development of hypertension and provide renoprotection at the functional and ultrastructural level. CONCLUSION: Chronic ExT preserves renal hemodynamics and structure in SHR; these effects are partially mediated by improved redox status and decreased inflammation.


Assuntos
Hemodinâmica , Rim/patologia , Condicionamento Físico Animal , Animais , Antioxidantes/metabolismo , Western Blotting , Hipertensão/metabolismo , Hipertensão/patologia , Hipertensão/fisiopatologia , Rim/metabolismo , Rim/fisiopatologia , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Ácido Peroxinitroso/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
PLoS One ; 6(9): e24028, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21949690

RESUMO

OBJECTIVE AND BACKGROUND: To assess renoprotective effects of a blueberry-enriched diet in a rat model of hypertension. Oxidative stress (OS) appears to be involved in the development of hypertension and related renal injury. Pharmacological antioxidants can attenuate hypertension and hypertension-induced renal injury; however, attention has shifted recently to the therapeutic potential of natural products as antioxidants. Blueberries (BB) have among the highest antioxidant capacities of fruits and vegetables. METHODS AND RESULTS: Male spontaneously hypertensive rats received a BB-enriched diet (2% w/w) or an isocaloric control diet for 6 or 12 weeks or 2 days. Compared to controls, rats fed BB-enriched diet for 6 or 12 weeks exhibited lower blood pressure, improved glomerular filtration rate, and decreased renovascular resistance. As measured by electron paramagnetic resonance spectroscopy, significant decreases in total reactive oxygen species (ROS), peroxynitrite, and superoxide production rates were observed in kidney tissues in rats on long-term dietary treatment, consistent with reduced pathology and improved function. Additionally, measures of antioxidant status improved; specifically, renal glutathione and catalase activities increased markedly. Contrasted to these observations indicating reduced OS in the BB group after long-term feeding, similar measurements made in rats fed the same diet for only 2 days yielded evidence of increased OS; specifically, significant increases in total ROS, peroxynitrite, and superoxide production rates in all tissues (kidney, brain, and liver) assayed in BB-fed rats. These results were evidence of "hormesis" during brief exposure, which dissipated with time as indicated by enhanced levels of catalase in heart and liver of BB group. CONCLUSION: Long-term feeding of BB-enriched diet lowered blood pressure, preserved renal hemodynamics, and improved redox status in kidneys of hypertensive rats and concomitantly demonstrated the potential to delay or attenuate development of hypertension-induced renal injury, and these effects appear to be mediated by a short-term hormetic response.


Assuntos
Antioxidantes/farmacologia , Mirtilos Azuis (Planta)/química , Dieta , Hipertensão/prevenção & controle , Nefropatias/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Catalase/metabolismo , Creatinina/urina , Glutationa/metabolismo , Hormese , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Nefropatias/fisiopatologia , Nefropatias/urina , Masculino , Nitratos/análise , Nitratos/urina , Nitritos/análise , Nitritos/urina , Fitoterapia/métodos , Preparações de Plantas/farmacologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
11.
Curr Hypertens Rep ; 12(2): 99-104, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20424938

RESUMO

Metabolic syndrome (MetS) is a constellation of metabolic derangements and underlying factors that significantly increases the risk for developing type 2 diabetes and cardiovascular diseases. MetS is a low-grade inflammatory condition, with systemic inflammation and inflammation of central abdominal fat as contributors. Systemic inflammation in MetS is thought to involve C-reactive protein and some proinflammatory cytokines; the nuclear factor-kappaB pathway also is believed to play a role. Inflammation of central adipose tissue leads to adipokine production, followed by secretion of adipokines into the general circulation to contribute to the overall inflammatory condition. The molecular mechanisms that contribute to this inflammation are still somewhat unclear, but several serine/threonine kinases are known to be involved. Dietary components may also contribute to central adiposity and the inflammation seen in MetS.


Assuntos
Gordura Abdominal , Adiposidade , Inflamação/patologia , Síndrome Metabólica/patologia , Estresse Oxidativo , Adipocinas/biossíntese , Adiponectina , Proteína C-Reativa , Citocinas , Dieta , Humanos , Inflamação/complicações , Resistência à Insulina , Interleucinas , Leptina , Estilo de Vida , NF-kappa B , Estado Nutricional , Fatores de Risco , Serina , Treonina , Fator de Necrose Tumoral alfa
12.
Cardiovasc Res ; 85(3): 473-83, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19729361

RESUMO

AIMS: Inflammatory molecules and their transcription factor, nuclear factor kappa-B (NF-kappaB), are thought to play important roles in diabetes-induced cardiac dysfunction. Here, we investigated the effects of pyrrolidine dithiocarbamate (PDTC), a NF-kappaB inhibitor, in diabetic mice. METHODS AND RESULTS: Obese db/db mice and heterozygous lean mice (n = 8) were allowed free access to drinking water (control) or water containing PDTC (100 mg/kg) for 20 weeks. Left ventricular (LV) function was measured using echocardiography at baseline and at study end. Mice were sacrificed and LV removed for gene expression, biochemical, immunofluorescence, and mitochondrial assays. LV and mitochondrial reactive oxygen species (ROS), superoxide and peroxynitrite were measured using electron spin resonance spectroscopy. Enhanced NF-kappaB activity in db/db mice was associated with increased oxidative stress as demonstrated by increased ROS, superoxide, and peroxynitrite production, and increased NF-kappaB, gp91phox, and Nox1 expression; PDTC ameliorated these effects. Mitochondrial free radical production and structural damage were higher in the db/db group than in the control, db/db PDTC, and PDTC-treated heterozygous animal groups. CONCLUSION: This study demonstrates that NF-kappaB blockade with PDTC mitigates oxidative stress and improves mitochondrial structural integrity directly, through down-regulation of increased oxygen-free radicals, thereby increasing ATP synthesis and thus restoring cardiac function in type II diabetes.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Coração/fisiopatologia , Mitocôndrias/fisiologia , NF-kappa B/fisiologia , Estresse Oxidativo , Animais , Peso Corporal , Ecocardiografia , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Glutationa/análise , Interleucina-6/sangue , Masculino , Camundongos , NF-kappa B/análise , NF-kappa B/antagonistas & inibidores , Tamanho do Órgão , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Fator de Necrose Tumoral alfa/sangue
13.
Obesity (Silver Spring) ; 17(11): 1994-2002, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19424163

RESUMO

Diabetic nephropathy is the leading cause of renal failure in the United States. The obese Zucker rat (OZR; fa/fa) is a commonly used model of type 2 diabetes and metabolic syndrome (MetS), and of the nephropathy and renal oxidative stress commonly seen in these disorders. Heterozygous lean Zucker rats (LZRs; fa/+) are susceptible to high-fat diet (HFD)-induced obesity and MetS. The present study was designed to investigate whether 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (TEMPOL), a membrane-permeable radical scavenger, could alleviate the renal effects of MetS in OZR and LZR fed a HFD, which resembles the typical "Western" diet. OZR and LZR were fed a HFD (OZR-HFD and LZR-HFD) or regular diet (OZR-RD and LZR-RD) and allowed free access to drinking water or water containing 1 mmol/l TEMPOL for 10 weeks. When compared to OZR-RD animals, OZR-HFD animals exhibited significantly higher levels of total renal cortical reactive oxygen species (ROS) production, plasma lipids, insulin, C-reactive protein, blood urea nitrogen (BUN), creatinine (Cr), and urinary albumin excretion (P < 0.05); these changes were accompanied by a significant decrease in plasma high-density lipoprotein levels (P < 0.05). The mRNA expression levels of desmin, tumor necrosis factor-alpha (TNF-alpha), nuclear factor kappaB (NFkappaB), and NAD(P)H oxidase-1 (NOX-1) were significantly higher in the renal cortical tissues of OZR-HFD animals; NFkappaB p65 DNA binding activity as determined by electrophoretic mobility shift assay was also significantly higher in these animals. The same trends were noted in LZR-HFD animals. Our data demonstrate that TEMPOL may prove beneficial in treating the early stages of the nephropathy often associated with MetS.


Assuntos
Antioxidantes/uso terapêutico , Óxidos N-Cíclicos/uso terapêutico , Nefropatias/tratamento farmacológico , Síndrome Metabólica/complicações , Espécies Reativas de Oxigênio/análise , Animais , Antioxidantes/farmacologia , Pressão Sanguínea , Óxidos N-Cíclicos/farmacologia , Desmina/genética , Desmina/metabolismo , Gorduras na Dieta/administração & dosagem , Espectroscopia de Ressonância de Spin Eletrônica , Homozigoto , Córtex Renal/química , Córtex Renal/patologia , Nefropatias/complicações , Glomérulos Renais/ultraestrutura , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Estresse Oxidativo , Ratos , Ratos Zucker , Marcadores de Spin , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
14.
Free Radic Biol Med ; 46(4): 462-70, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19041937

RESUMO

Mitochondrial damage is implicated in the progression of cardiac disease. Considerable evidence suggests that proinflammatory cytokines induce oxidative stress and contribute to cardiac dysfunction. This study was conducted to determine whether a TNF-induced increase in superoxide (O(2)(*)(-)) contributes to mitochondrial damage in the left ventricle (LV) by impairing respiratory complex I activity. We employed an electron paramagnetic resonance (EPR) method to measure O(2)(*)(-) and oxygen consumption in mitochondrial respiratory complexes, using an oxygen label. Adult male Sprague-Dawley rats were divided into four groups: control, TNF treatment (ip), TNF+ apocynin (APO; 200 micromol/kg bw, orally), and TNF+ Tempol (Temp; 300 micromol/kg bw, orally). TNF was injected daily for 5 days. Rats were sacrificed, LV tissue was collected, and mitochondria were isolated for EPR studies. Total LV ROS production was significantly higher in TNF animals than in controls; APO or Temp treatment ameliorated TNF-induced LV ROS production. Total mitochondrial ROS production was significantly higher in the TNF and TNF+ APO groups than in the control and TNF+ Temp groups. These findings suggest that TNF alters the cellular redox state, reduces the expression of four complex I subunits by increasing mitochondrial O(2)(*)(-) production and depleting ATP synthesis, and decreases oxygen consumption, thereby resulting in mitochondrial damage and leading to LV dysfunction.


Assuntos
Citotoxicidade Imunológica/fisiologia , Complexo I de Transporte de Elétrons/metabolismo , Mitocôndrias Cardíacas/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Disfunção Ventricular Esquerda/enzimologia , Acetofenonas/administração & dosagem , Animais , Respiração Celular/efeitos dos fármacos , Respiração Celular/fisiologia , Óxidos N-Cíclicos/administração & dosagem , Citotoxicidade Imunológica/efeitos dos fármacos , Ecocardiografia , Espectroscopia de Ressonância de Spin Eletrônica , Complexo I de Transporte de Elétrons/genética , Sequestradores de Radicais Livres/administração & dosagem , Masculino , Miocárdio/enzimologia , Miocárdio/imunologia , Miocárdio/ultraestrutura , NADPH Oxidases/antagonistas & inibidores , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Consumo de Oxigênio/efeitos dos fármacos , Consumo de Oxigênio/fisiologia , Ratos , Ratos Sprague-Dawley , Marcadores de Spin , Superóxidos/metabolismo , Fator de Necrose Tumoral alfa/administração & dosagem , Disfunção Ventricular Esquerda/imunologia , Disfunção Ventricular Esquerda/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA