Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
ASAIO J ; 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38483814

RESUMO

Extracorporeal membrane oxygenation (ECMO) is often associated with disturbances in acid/base status that can be triggered by the underlying pathology or the ECMO circuit itself. Extracorporeal membrane oxygenation is known to cause hypocapnia, but the impact of reduced partial pressure of carbon dioxide (pCO2) on biomarkers of tissue perfusion during veno-arterial (VA)-ECMO has not been evaluated. To study the impact of low pCO2 on perfusion indices in VA-ECMO, we placed Sprague-Dawley rats on an established VA-ECMO circuit using either an oxygen/carbon dioxide mixture (O2 95%, CO2 5%) or 100% O2 delivered through the oxygenator (n = 5 per cohort). Animals receiving 100% O2 developed a significant VA CO2 difference (pCO2 gap) and rising blood lactate levels that were inversely proportional to the decrease in pCO2 values. In contrast, pCO2 gap and lactate levels remained similar to pre-ECMO baseline levels in animals receiving the O2/CO2 mixture. More importantly, there was no significant difference in venous oxygen saturation (SvO2) between the two groups, suggesting that elevated blood lactate levels observed in the rats receiving 100% O2 were a response to oxygenator induced hypocapnia and alkaline pH rather than reduced perfusion or underlying tissue hypoxia. These findings have implications in clinical and experimental extracorporeal support contexts.

2.
Artigo em Inglês | MEDLINE | ID: mdl-37683721

RESUMO

BACKGROUND: Factor XII (FXII) is a multifunctional protease capable of activating thrombotic and inflammatory pathways. FXII has been linked to thrombosis in extracorporeal membrane oxygenation (ECMO), but the role of FXII in ECMO-induced inflammatory complications has not been studied. We used novel gene-targeted FXII- deficient rats to evaluate the role of FXII in ECMO-induced thromboinflammation. METHODS: FXII-deficient (FXII-/-) Sprague-Dawley rats were generated using CRISPR/Cas9. A minimally invasive venoarterial (VA) ECMO model was used to compare wild-type (WT) and FXII-/- rats in 2 separate experimental cohorts: rats placed on ECMO without pharmacologic anticoagulation and rats anticoagulated with argatroban. Rats were maintained on ECMO for 1 hour or until circuit failure occurred. Comparisons were made with unchallenged rats and rats that underwent a sham surgical procedure without ECMO. RESULTS: FXII-/- rats were maintained on ECMO without pharmacologic anticoagulation with low resistance throughout the 1-hour experiment. In contrast, WT rats placed on ECMO without anticoagulation developed thrombotic circuit failure within 10 minutes. Argatroban provided a means to maintain WT and FXII-/- rats on ECMO for the 1-hour time frame without thrombotic complications. Analyses of these rats demonstrated that ECMO resulted in increased neutrophil migration into the liver that was significantly blunted by FXII deficiency. ECMO also resulted in increases in high molecular weight kininogen cleavage and complement activation that were abrogated by genetic deletion of FXII. CONCLUSIONS: FXII initiates hemostatic system activation and key inflammatory sequelae in ECMO, suggesting that therapies targeting FXII could limit both thromboembolism and inopportune inflammatory complications in this setting.

3.
J Thorac Cardiovasc Surg ; 166(5): e164-e173, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37164051

RESUMO

OBJECTIVE: Several human studies have associated nitric oxide administration via the cardiopulmonary bypass circuit with decreased incidence of cardiopulmonary bypass-associated acute kidney injury, but histopathologic and serologic evidence of nitric oxide efficacy for acute kidney injury attenuation are lacking. METHODS: By using a survival ovine model (72 hours), acute kidney injury was induced by implementing low-flow cardiopulmonary bypass for 2 hours, followed by full-flow cardiopulmonary bypass for 2 hours. The nitric oxide cohort (n = 6) received exogenous nitric oxide through the cardiopulmonary bypass circuit via the oxygenator, and the control group (n = 5) received no nitric oxide. Serial serologic biomarkers and renal histopathology were obtained. RESULTS: Baseline characteristics (age, weight) and intraoperative parameters (cardiopulmonary bypass time, urine output, heart rate, arterial pH, and lactate) were equivalent (P > .10) between groups. Postoperatively, urine output, heart rate, respiratory rate, and peripheral arterial saturation were equivalent (P > .10) between groups. Post-cardiopulmonary bypass creatinine elevations from baseline were significantly greater in the control group versus the nitric oxide group at 16, 24, and 48 hours (all P < .05). Histopathologic evidence of moderate/severe acute kidney injury (epithelial necrosis, tubular slough, cast formation, glomerular edema) occurred in 60% (3/5) of the control group versus 0% (0/6) of the nitric oxide group. Cortical tubular epithelial cilia lengthening (a sensitive sign of cellular injury) was significantly greater in the control group than in the nitric oxide group (P = .012). CONCLUSIONS: In a survival ovine cardiopulmonary bypass model, nitric oxide administered with cardiopulmonary bypass demonstrated serologic and histologic evidence of renal protection from acute kidney injury. These results provide insight into 1 potential mechanism for cardiopulmonary bypass-associated acute kidney injury and supports continued study of nitric oxide via cardiopulmonary bypass circuit for prevention of acute kidney injury.

4.
Thromb Res ; 185: 160-166, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31821908

RESUMO

INTRODUCTION: During storage, packed red blood cells undergo a series of physical, metabolic, and chemical changes collectively known as the red blood cell storage lesion. One key component of the red blood cell storage lesion is the accumulation of microparticles, which are submicron vesicles shed from erythrocytes as part of the aging process. Previous studies from our laboratory indicate that transfusion of these microparticles leads to lung injury, but the mechanism underlying this process is unknown. In the present study, we hypothesized that microparticles from aged packed red blood cell units induce pulmonary thrombosis. MATERIALS AND METHODS: Leukoreduced, platelet-depleted, murine packed red blood cells (pRBCS) were prepared then stored for up to 14 days. Microparticles were isolated from stored units via high-speed centrifugation. Mice were transfused with microparticles. The presence of pulmonary microthrombi was determined with light microscopy, Martius Scarlet Blue, and thrombocyte stains. In additional studies microparticles were labelled with CFSE prior to injection. Murine lung endothelial cells were cultured and P-selectin concentrations determined by ELISA. In subsequent studies, P-selectin was inhibited by PSI-697 injection prior to transfusion. RESULTS: We observed an increase in microthrombi formation in lung vasculature in mice receiving microparticles from stored packed red blood cell units as compared with controls. These microthrombi contained platelets, fibrin, and microparticles. Treatment of cultured lung endothelial cells with microparticles led to increased P-selectin in the media. Treatment of mice with a P-selectin inhibitor prior to microparticle infusion decreased microthrombi formation. CONCLUSIONS: These data suggest that microparticles isolated from aged packed red blood cell units promote the development of pulmonary microthrombi in a murine model of transfusion. This pro-thrombotic event appears to be mediated by P-selectin.


Assuntos
Micropartículas Derivadas de Células , Trombose , Animais , Preservação de Sangue , Células Endoteliais , Eritrócitos , Pulmão , Camundongos , Camundongos Endogâmicos C57BL , Selectina-P
5.
Shock ; 49(3): 288-294, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29438268

RESUMO

Microparticles are submicron vesicles shed from aging erythrocytes as a characteristic feature of the red blood cell (RBC) storage lesion. Exposure of pulmonary endothelial cells to RBC-derived microparticles promotes an inflammatory response, but the mechanisms underlying microparticle-induced endothelial cell activation are poorly understood. In the present study, cultured murine lung endothelial cells (MLECs) were treated with microparticles isolated from aged murine packed RBCs or vehicle. Microparticle-treated cells demonstrated increased expression of the adhesion molecules ICAM and E-selectin, as well as the cytokine, IL-6. To identify mechanisms that mediate these effects of microparticles on MLECs, cells were treated with microparticles covalently bound to carboxyfluorescein succinimidyl ester (CFSE) and cellular uptake of microparticles was quantified via flow cytometry. Compared with controls, there was a greater proportion of CFSE-positive MLECs from 15 min up to 24 h, suggesting endocytosis of the microparticles by endothelial cells. Colocalization of microparticles with lysosomes was observed via immunofluorescence, indicating endocytosis and endolysosomal trafficking. This process was inhibited by endocytosis inhibitors. SiRNA knockdown of Rab5 signaling protein in endothelial cells resulted in impaired microparticle uptake as compared with nonsense siRNA-treated cells, as well as an attenuation of the inflammatory response to microparticle treatment. Taken together, these data suggest that endocytosis of RBC-derived microparticles by lung endothelial cells results in endothelial cell activation. This response seems to be mediated, in part, by the Rab5 signaling protein.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Endocitose , Células Epiteliais/metabolismo , Eritrócitos/metabolismo , Pulmão/metabolismo , Mucosa Respiratória/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Células Epiteliais/citologia , Eritrócitos/citologia , Pulmão/citologia , Masculino , Camundongos , Mucosa Respiratória/citologia
6.
Surgery ; 163(2): 423-429, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29198748

RESUMO

BACKGROUND: Red blood cell-derived microparticles are biologically active, submicron vesicles shed by erythrocytes during storage. Recent clinical studies have linked the duration of red blood cell storage with thromboembolic events in critically ill transfusion recipients. In the present study, we hypothesized that microparticles from aged packed red blood cell units promote a hypercoagulable state in a murine model of transfusion. METHODS: Microparticles were isolated from aged, murine packed red blood cell units via serial centrifugation. Healthy male C57BL/6 mice were transfused with microparticles or an equivalent volume of vehicle, and whole blood was harvested for analysis via rotational thromboelastometry. Serum was harvested from a separate set of mice after microparticles or saline injection, and analyzed for fibrinogen levels. Red blood cell-derived microparticles were analyzed for their ability to convert prothrombin to thrombin. Finally, mice were transfused with either red blood cell microparticles or saline vehicle, and a tail bleeding time assay was performed after an equilibration period of 2, 6, 12, or 24 hours. RESULTS: Mice injected with red blood cell-derived microparticles demonstrated an accelerated clot formation time (109.3 ± 26.9 vs 141.6 ± 28.2 sec) and increased α angle (68.8 ± 5.0 degrees vs 62.8 ± 4.7 degrees) compared with control (each P < .05). Clotting time and maximum clot firmness were not significantly different between the 2 groups. Red blood cell-derived microparticles exhibited a hundredfold greater conversion of prothrombin substrate to its active thrombin form (66.60 ± 0.03 vs 0.70 ± 0.01 peak OD; P<.0001). Additionally, serum fibrinogen levels were lower in microparticles-injected mice compared with saline vehicle, suggesting thrombin-mediated conversion to insoluble fibrin (14.0 vs 16.5 µg/mL, P<.05). In the tail bleeding time model, there was a more rapid cessation of bleeding at 2 hours posttransfusion (90.6 vs 123.7 sec) and 6 hours posttransfusion (87.1 vs 141.4 sec) in microparticles-injected mice as compared with saline vehicle (each P<.05). There was no difference in tail bleeding time at 12 or 24 hours. CONCLUSION: Red blood cell-derived microparticles induce a transient hypercoagulable state through accelerated activation of clotting factors.


Assuntos
Micropartículas Derivadas de Células , Trombofilia , Reação Transfusional , Animais , Transfusão de Sangue , Masculino , Camundongos Endogâmicos C57BL , Modelos Animais
7.
Cancer Cell ; 26(5): 738-53, 2014 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-25517751

RESUMO

Autophagy promotes tumor growth by generating nutrients from the degradation of intracellular structures. Here we establish, using shRNAs, a dominant-negative mutant, and a pharmacologic inhibitor, mefenamic acid (MFA), that the Transient Receptor Potential Melastatin 3 (TRPM3) channel promotes the growth of clear cell renal cell carcinoma (ccRCC) and stimulates MAP1LC3A (LC3A) and MAP1LC3B (LC3B) autophagy. Increased expression of TRPM3 in RCC leads to Ca(2+) influx, activation of CAMKK2, AMPK, and ULK1, and phagophore formation. In addition, TRPM3 Ca(2+) and Zn(2+) fluxes inhibit miR-214, which directly targets LC3A and LC3B. The von Hippel-Lindau tumor suppressor (VHL) represses TRPM3 directly through miR-204 and indirectly through another miR-204 target, Caveolin 1 (CAV1).


Assuntos
Autofagia , Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , MicroRNAs/fisiologia , Canais de Cátion TRPM/genética , Animais , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Neoplasias Renais/genética , Camundongos Nus , Transplante de Neoplasias , Oncogenes , Interferência de RNA , Canais de Cátion TRPM/metabolismo , Carga Tumoral , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
8.
Biochim Biophys Acta ; 1841(6): 888-95, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24315825

RESUMO

Mice deficient in group 1b phospholipase A2 have decreased plasma lysophosphatidylcholine and increased hepatic oxidation that is inhibited by intraperitoneal lysophosphatidylcholine injection. This study sought to identify a mechanism for lysophosphatidylcholine-mediated inhibition of hepatic oxidative function. Results showed that in vitro incubation of isolated mitochondria with 40-200µM lysophosphatidylcholine caused cyclosporine A-resistant swelling in a concentration-dependent manner. However, when mitochondria were challenged with 220µM CaCl2, cyclosporine A protected against permeability transition induced by 40µM, but not 80µM lysophosphatidylcholine. Incubation with 40-120µM lysophosphatidylcholine also increased mitochondrial permeability to 75µM CaCl2 in a concentration-dependent manner. Interestingly, despite incubation with 80µM lysophosphatidylcholine, the mitochondrial membrane potential was steady in the presence of succinate, and oxidation rates and respiratory control indices were similar to controls in the presence of succinate, glutamate/malate, and palmitoyl-carnitine. However, mitochondrial oxidation rates were inhibited by 30-50% at 100µM lysophosphatidylcholine. Finally, while 40µM lysophosphatidylcholine has no effect on fatty acid oxidation and mitochondria remained impermeable in intact hepatocytes, 100µM lysophosphatidylcholine inhibited fatty acid stimulated oxidation and caused intracellular mitochondrial permeability. Taken together, these present data demonstrated that LPC concentration dependently modulates mitochondrial microenvironment, with low micromolar concentrations of lysophosphatidylcholine sufficient to change hepatic oxidation rate whereas higher concentrations are required to disrupt mitochondrial integrity.


Assuntos
Lisofosfatidilcolinas/administração & dosagem , Mitocôndrias Hepáticas/metabolismo , Oxirredução/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos , Animais , Cloreto de Cálcio/administração & dosagem , Ciclosporina/administração & dosagem , Relação Dose-Resposta a Droga , Fosfolipases A2 do Grupo IV/deficiência , Fosfolipases A2 do Grupo IV/genética , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Lisofosfatidilcolinas/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/genética , Camundongos , Mitocôndrias Hepáticas/efeitos dos fármacos
9.
Circulation ; 128(22): 2364-71, 2013 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-24170386

RESUMO

BACKGROUND: Abnormal glucose metabolism is a central feature of disorders with increased rates of cardiovascular disease. Low levels of high-density lipoprotein (HDL) are a key predictor for cardiovascular disease. We used genetic mouse models with increased HDL levels (apolipoprotein A-I transgenic [apoA-I tg]) and reduced HDL levels (apoA-I-deficient [apoA-I ko]) to investigate whether HDL modulates mitochondrial bioenergetics in skeletal muscle. METHODS AND RESULTS: ApoA-I ko mice exhibited fasting hyperglycemia and impaired glucose tolerance test compared with wild-type mice. Mitochondria isolated from gastrocnemius muscle of apoA-I ko mice displayed markedly blunted ATP synthesis. Endurance capacity during exercise exhaustion test was impaired in apoA-I ko mice. HDL directly enhanced glucose oxidation by increasing glycolysis and mitochondrial respiration rate in C2C12 muscle cells. ApoA-I tg mice exhibited lower fasting glucose levels, improved glucose tolerance test, increased lactate levels, reduced fat mass, associated with protection against age-induced decline of endurance capacity compared with wild-type mice. Circulating levels of fibroblast growth factor 21, a novel biomarker for mitochondrial respiratory chain deficiencies and inhibitor of white adipose lipolysis, were significantly reduced in apoA-I tg mice. Consistent with an increase in glucose utilization of skeletal muscle, genetically increased HDL and apoA-I levels in mice prevented high-fat diet-induced impairment of glucose homeostasis. CONCLUSIONS: In view of impaired mitochondrial function and decreased HDL levels in type 2 diabetes mellitus, our findings indicate that HDL-raising therapies may preserve muscle mitochondrial function and address key aspects of type 2 diabetes mellitus beyond cardiovascular disease.


Assuntos
Glicemia/metabolismo , Intolerância à Glucose/metabolismo , Hiperglicemia/metabolismo , Lipoproteínas HDL/metabolismo , Músculo Esquelético/metabolismo , Animais , Apolipoproteína A-I/genética , Respiração Celular/fisiologia , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/sangue , Fatores de Crescimento de Fibroblastos/sangue , Homeostase/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Musculares/metabolismo , Resistência Física/fisiologia
10.
Mol Cell Endocrinol ; 381(1-2): 280-90, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23994018

RESUMO

Signaling through the mammalian target of rapamycin complex 1 (mTORC1) and its effectors the S6-kinases (S6K) in the hypothalamus is thought to be involved in nutrient sensing and control of food intake. Given the anatomical proximity of this pathway to circuits for the hormone ghrelin, we investigated the potential role of the mTORC1/S6K pathway in mediating the metabolic effects of ghrelin. We found that ghrelin promoted phosphorylation of S6K1 in the mouse hypothalamic cell line N-41 and in the rat hypothalamus after intracerebroventricular administration. Rapamycin, an inhibitor of mTORC1, suppressed ghrelin-induced phosphorylation of hypothalamic S6K1 and increased food intake and insulin in rats. Chronic peripheral administration of ghrelin induced a significant increase in body weight, fat mass and food efficiency in wild-type and S6K2-knockout but not in S6K1-knockout mice. We therefore propose that ghrelin-induced hyperphagia, adiposity and insulin secretion are controlled by a central nervous system involving the mTORC1/S6K1 pathway.


Assuntos
Adiposidade/fisiologia , Ingestão de Energia , Grelina/fisiologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Tecido Adiposo Marrom/fisiologia , Animais , Linhagem Celular , Insulina/sangue , Canais Iônicos/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/metabolismo , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/metabolismo , Ratos , Ratos Wistar , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteína Desacopladora 1
11.
Nat Commun ; 4: 1968, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23744028

RESUMO

The G protein-coupled receptor 83 (Gpr83) is widely expressed in brain regions regulating energy metabolism. Here we report that hypothalamic expression of Gpr83 is regulated in response to nutrient availability and is decreased in obese mice compared with lean mice. In the arcuate nucleus, Gpr83 colocalizes with the ghrelin receptor (Ghsr1a) and the agouti-related protein. In vitro analyses show heterodimerization of Gpr83 with Ghsr1a diminishes activation of Ghsr1a by acyl-ghrelin. The orexigenic and adipogenic effect of ghrelin is accordingly potentiated in Gpr83-deficient mice. Interestingly, Gpr83 knock-out mice have normal body weight and glucose tolerance when fed a regular chow diet, but are protected from obesity and glucose intolerance when challenged with a high-fat diet, despite hyperphagia and increased hypothalamic expression of agouti-related protein, Npy, Hcrt and Ghsr1a. Together, our data suggest that Gpr83 modulates ghrelin action but also indicate that Gpr83 regulates systemic metabolism through other ghrelin-independent pathways.


Assuntos
Metabolismo Energético , Grelina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Dieta Hiperlipídica , Metabolismo Energético/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Perfilação da Expressão Gênica , Grelina/administração & dosagem , Grelina/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Fenótipo , Multimerização Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Ratos , Receptor Tipo 3 de Melanocortina/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de Grelina/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
Diabetes ; 61(11): 2734-42, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22787140

RESUMO

Although obesity rates are rapidly rising, caloric restriction remains one of the few safe therapies. Here we tested the hypothesis that obesity-associated disorders are caused by increased adipose tissue as opposed to excess dietary lipids. Fat mass (FM) of lean C57B6 mice fed a high-fat diet (HFD; FMC mice) was "clamped" to match the FM of mice maintained on a low-fat diet (standard diet [SD] mice). FMC mice displayed improved glucose and insulin tolerance as compared with ad libitum HFD mice (P < 0.001) or SD mice (P < 0.05). These improvements were associated with fewer signs of inflammation, consistent with the less-impaired metabolism. In follow-up studies, diet-induced obese mice were food restricted for 5 weeks to achieve FM levels identical with those of age-matched SD mice. Previously, obese mice exhibited improved glucose and insulin tolerance but showed markedly increased fasting-induced hyperphagia (P < 0.001). When mice were given ad libitum access to the HFD, the hyperphagia of these mice led to accelerated body weight gain as compared with otherwise matched controls without a history of obesity. These results suggest that although caloric restriction on a HFD provides metabolic benefits, maintaining those benefits may require lifelong continuation, at least in individuals with a history of obesity.


Assuntos
Tecido Adiposo Branco/metabolismo , Restrição Calórica/efeitos adversos , Metabolismo Energético , Adiposidade , Animais , Dieta Hiperlipídica/efeitos adversos , Dieta Redutora/efeitos adversos , Regulação da Expressão Gênica , Intolerância à Glucose/sangue , Intolerância à Glucose/etiologia , Intolerância à Glucose/imunologia , Intolerância à Glucose/metabolismo , Hiperfagia/etiologia , Hipotálamo/metabolismo , Mediadores da Inflamação/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/dietoterapia , Obesidade/imunologia , Obesidade/metabolismo , Obesidade/prevenção & controle , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Distribuição Aleatória , Prevenção Secundária , Aumento de Peso
13.
PLoS One ; 7(6): e38997, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22720011

RESUMO

UNLABELLED: Low-carbohydrate, high-fat (LC-HF) diets are popular for inducing weight loss in overweighed adults. Adaptive thermogenesis increased by specific effects of macronutrients on energy expenditure has been postulated to induce this weight loss. We studied brown adipose tissue (BAT) morphology and function following exposure to different LC-HF diets. METHODS: Male Wistar rats were fed a standard control diet ad libitum or pair-fed isoenergetic amounts of three experimental diets for 4 weeks. The diets had the following macronutrient composition (% metabolizable energy: carbohydrates, fat, protein): control (64.3/16.7/19), LC-HF-low protein (LC-HF-LP, 1.7/92.8/5.5), LC-HF-normal-protein (LC-HF-NP, 2.2/78.7/19.1), and a high fat diet with carbohydrates ("high fat", 19.4/61.9/18.7). RESULTS: Body weight gain was reduced in all pair-fed experimental groups as compared to rats fed the control diet, with more pronounced effect in rats on LC-HF diets than on the high fat diet with carbohydrates. High fat diets increased expression of PGC1α and ADRB3 in BAT indicating higher SNS outflow. However, UCP1 mRNA expression and expression of UCP1 assessed by immunohistochemistry was not different between diet groups. In accordance, analysis of mitochondrial function in-vitro by extracellular flux analyser (Seahorse Bioscience) and measurement of inducible thermogenesis in vivo (primary endpoint), explored by indirect calorimetry following norepinephrine injection, did not show significant differences between groups. Histology of BAT revealed increased lipid droplet size in rats fed the high-fat diet and both LC-HF diets. CONCLUSION: All experimental diets upregulated expression of genes which are indicative for increased BAT activity. However, the functional measurements in vivo revealed no increase of inducible BAT thermogenesis. This indicates that lower body weight gain with LC-HF diets and a high fat diet in a pair-feeding setting is not caused by increased adaptive thermogenesis in BAT.


Assuntos
Tecido Adiposo Marrom/efeitos dos fármacos , Carboidratos da Dieta/administração & dosagem , Ingestão de Energia , Termogênese , Tecido Adiposo Marrom/metabolismo , Animais , Sequência de Bases , Temperatura Corporal , Peso Corporal , Primers do DNA , Carboidratos da Dieta/farmacologia , Imuno-Histoquímica , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Norepinefrina/farmacologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Termogênese/efeitos dos fármacos
14.
PLoS One ; 7(3): e32631, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22412899

RESUMO

Targeted deletion of S6 kinase (S6K) 1 in mice leads to higher energy expenditure and improved glucose metabolism. However, the molecular mechanisms controlling these effects remain to be fully elucidated. Here, we analyze the potential role of dietary lipids in regulating the mTORC1/S6K system. Analysis of S6K phosphorylation in vivo and in vitro showed that dietary lipids activate S6K, and this effect is not dependent upon amino acids. Comparison of male mice lacking S6K1 and 2 (S6K-dko) with wt controls showed that S6K-dko mice are protected against obesity and glucose intolerance induced by a high-fat diet. S6K-dko mice fed a high-fat diet had increased energy expenditure, improved glucose tolerance, lower fat mass gain, and changes in markers of lipid metabolism. Importantly, however, these metabolic phenotypes were dependent upon dietary lipids, with no such effects observed in S6K-dko mice fed a fat-free diet. These changes appear to be mediated via modulation of cellular metabolism in skeletal muscle, as shown by the expression of genes involved in energy metabolism. Taken together, our results suggest that the metabolic functions of S6K in vivo play a key role as a molecular interface connecting dietary lipids to the endogenous control of energy metabolism.


Assuntos
Gorduras na Dieta/metabolismo , Metabolismo dos Lipídeos , Proteínas Quinases S6 Ribossômicas/metabolismo , Aminoácidos de Cadeia Ramificada/metabolismo , Animais , Linhagem Celular , Colesterol/sangue , Dieta Hiperlipídica/efeitos adversos , Ativação Enzimática , Ácidos Graxos/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Feminino , Deleção de Genes , Intolerância à Glucose/genética , Intolerância à Glucose/prevenção & controle , Leptina/sangue , Metabolismo dos Lipídeos/genética , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Obesidade/genética , Obesidade/metabolismo , Fenótipo , Proteínas Quinases S6 Ribossômicas/deficiência , Proteínas Quinases S6 Ribossômicas/genética , Triglicerídeos/sangue
15.
Diabetes ; 61(2): 339-45, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22210324

RESUMO

Glucocorticoid receptors are highly expressed in the hypothalamic paraventricular nucleus (PVN) and arcuate nucleus (ARC). As glucocorticoids have pronounced effects on neuropeptide Y (NPY) expression and as NPY neurons projecting from the ARC to the PVN are pivotal for balancing feeding behavior and glucose metabolism, we investigated the effect of glucocorticoid signaling in these areas on endogenous glucose production (EGP) and insulin sensitivity by local retrodialysis of the glucocorticoid receptor agonist dexamethasone into the ARC or the PVN, in combination with isotope dilution and hyperinsulinemic-euglycemic clamp techniques. Retrodialysis of dexamethasone for 90 min into the ARC or the PVN did not have significant effects on basal plasma glucose concentration. During the hyperinsulinemic-euglycemic clamp, retrodialysis of dexamethasone into the ARC largely prevented the suppressive effect of hyperinsulinemia on EGP. Antagonizing the NPY1 receptors by intracerebroventricular infusion of its antagonist largely blocked the hepatic insulin resistance induced by dexamethasone in the ARC. The dexamethasone-ARC-induced inhibition of hepatic insulin sensitivity was also prevented by hepatic sympathetic denervation. These data suggest that glucocorticoid signaling specifically in the ARC neurons modulates hepatic insulin responsiveness via NPY and the sympathetic system, which may add to our understanding of the metabolic impact of clinical conditions associated with hypercortisolism.


Assuntos
Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Dexametasona/farmacologia , Resistência à Insulina , Fígado/metabolismo , Transdução de Sinais/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Glucose/metabolismo , Fígado/inervação , Masculino , Neuropeptídeo Y/farmacologia , Neuropeptídeo Y/fisiologia , Ratos , Ratos Wistar , Receptores de Neuropeptídeo Y/fisiologia , Simpatectomia
16.
J Clin Invest ; 119(9): 2577-89, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19726875

RESUMO

Insulin signaling can be modulated by several isoforms of PKC in peripheral tissues. Here, we assessed whether one specific isoform, PKC-theta, was expressed in critical CNS regions that regulate energy balance and whether it mediated the deleterious effects of diets high in fat, specifically palmitic acid, on hypothalamic insulin activity in rats and mice. Using a combination of in situ hybridization and immunohistochemistry, we found that PKC-theta was expressed in discrete neuronal populations of the arcuate nucleus, specifically the neuropeptide Y/agouti-related protein neurons and the dorsal medial nucleus in the hypothalamus. CNS exposure to palmitic acid via direct infusion or by oral gavage increased the localization of PKC-theta to cell membranes in the hypothalamus, which was associated with impaired hypothalamic insulin and leptin signaling. This finding was specific for palmitic acid, as the monounsaturated fatty acid, oleic acid, neither increased membrane localization of PKC-theta nor induced insulin resistance. Finally, arcuate-specific knockdown of PKC-theta attenuated diet-induced obesity and improved insulin signaling. These results suggest that many of the deleterious effects of high-fat diets, specifically those enriched with palmitic acid, are CNS mediated via PKC-theta activation, resulting in reduced insulin activity.


Assuntos
Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Resistência à Insulina/fisiologia , Isoenzimas/metabolismo , Ácido Palmítico/toxicidade , Proteína Quinase C/metabolismo , Animais , Sequência de Bases , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/toxicidade , Gluconeogênese/efeitos dos fármacos , Isoenzimas/deficiência , Isoenzimas/genética , Leptina/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ácido Palmítico/administração & dosagem , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Proteína Quinase C-theta , RNA Interferente Pequeno/genética , Ratos , Ratos Long-Evans , Transdução de Sinais/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/enzimologia
17.
Nat Chem Biol ; 5(10): 749-57, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19597507

RESUMO

We report the efficacy of a new peptide with agonism at the glucagon and GLP-1 receptors that has potent, sustained satiation-inducing and lipolytic effects. Selective chemical modification to glucagon resulted in a loss of specificity, with minimal change to inherent activity. The structural basis for the co-agonism appears to be a combination of local positional interactions and a change in secondary structure. Two co-agonist peptides differing from each other only in their level of glucagon receptor agonism were studied in rodent obesity models. Administration of PEGylated peptides once per week normalized adiposity and glucose tolerance in diet-induced obese mice. Reduction of body weight was achieved by a loss of body fat resulting from decreased food intake and increased energy expenditure. These preclinical studies indicate that when full GLP-1 agonism is augmented with an appropriate degree of glucagon receptor activation, body fat reduction can be substantially enhanced without any overt adverse effects.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/agonistas , Obesidade/tratamento farmacológico , Peptídeos Cíclicos/uso terapêutico , Polietilenoglicóis/química , Receptores de Glucagon/agonistas , Tecido Adiposo/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Peso Corporal/efeitos dos fármacos , AMP Cíclico/biossíntese , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Teste de Tolerância a Glucose , Camundongos , Camundongos Obesos , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Conformação Proteica
18.
J Biol Chem ; 282(9): 6183-91, 2007 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-17200118

RESUMO

Newly synthesized thyroglobulin (Tg), the secretory glycoprotein that serves as precursor in thyroid hormone synthesis, normally forms transient covalent protein complexes with oxidoreductases of the endoplasmic reticulum (ER). The Tg-G2320R mutation is responsible for congenital hypothyroidism in rdw/rdw rats, in which a lack of secondary thyroid enlargement (goiter) implicates death of thyrocytes as part of disease pathogenesis. We found that mutant Tg-G2320R was retained within the ER with no detectable synthesis of thyroxine, had persistent exposure of free cysteine thiols, and was associated with activated ER stress response but incomplete ER-associated degradation (ERAD). Tg-G2320R associated with multiple ER resident proteins, most notably ERp72, including covalent Tg-ERp72 interactions. In PC Cl3 thyrocytes, inducible overexpression of ERp72 increased the ability of cells to maintain Tg cysteines in a reduced state. Noncovalent interactions of several ER chaperones with newly synthesized Tg-G2320R diminished over time in parallel with ERAD of the mutant protein, yet a small ERAD-resistant Tg fraction remained engaged in covalent association with ERp72 even 2 days post-synthesis. Such covalent protein aggregates may set the stage for apoptotic thyrocyte cell death, preventing thyroid goiter formation in rdw/rdw rats.


Assuntos
Nanismo/etiologia , Glicoproteínas de Membrana/metabolismo , Oxirredutases/metabolismo , Tireoglobulina/genética , Animais , Apoptose , Nanismo/metabolismo , Retículo Endoplasmático , Bócio , Glicoproteínas de Membrana/genética , Mutação de Sentido Incorreto , Ligação Proteica , Transporte Proteico , Ratos , Ratos Mutantes , Tireoglobulina/metabolismo , Glândula Tireoide/citologia
19.
Artigo em Inglês | MEDLINE | ID: mdl-16935483

RESUMO

The phosphoinositide (PI)-protein kinase C (PKC) signal transduction pathway is initiated by pre- and postsynaptic Galphaq-coupled receptors, and regulates several clinically relevant neurochemical events, including neurotransmitter release efficacy, monoamine receptor function and trafficking, monoamine transporter function and trafficking, axonal myelination, and gene expression. Mounting evidence for PI-PKC signaling hyperactivity in the peripheral (platelets) and central (premortem and postmortem brain) tissues of patients with schizophrenia, bipolar disorder, and major depressive disorder, coupled with evidence that PI-PKC signal transduction is down-regulated in rat brain following chronic, but not acute, treatment with antipsychotic, mood-stabilizer, and antidepressant medications, suggest that PI-PKC hyperactivity is central to an underlying pathophysiology. Evidence that membrane omega-3 fatty acids act as endogenous antagonists of the PI-PKC signal transduction pathway, coupled with evidence that omega-3 fatty acid deficiency is observed in peripheral and central tissues of patients with schizophrenia, bipolar disorder, and major depressive disorder, support the hypothesis that omega-3 fatty acid deficiency may contribute to elevated PI-PKC activity in these illnesses. The data reviewed in this paper outline a potential molecular mechanism by which omega-3 fatty acids could contribute to the pathophysiology and treatment of recurrent neuropsychiatric illness.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Transtornos Mentais/tratamento farmacológico , Transtornos Mentais/etiologia , Fosfatidilinositóis/metabolismo , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Ácidos Graxos/fisiologia , Ácidos Graxos Ômega-3/uso terapêutico , Humanos , Lipídeos de Membrana/fisiologia , Modelos Biológicos , Recidiva
20.
J Lab Clin Med ; 139(6): 357-63, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12066134

RESUMO

Estradiol-17beta (E2) exhibits potent antioxidant effects that cause continuous suppression of metal-catalyzed oxidation of low-density-lipoprotein in vitro. We sought to learn whether unidentified oxidation products retaining strong antioxidant property may be generated from E2 incubated with lipoproteins and subjected to oxidation by reactive oxygen species generators. E2 oxidation was markedly stimulated in the presence of both LDL and high-density lipoprotein. We have isolated two novel products (less polar than E2), formed when E2 was oxidized with copper sulfate and hydrogen peroxide in the presence of lipoproteins). Both compounds had molecular weights of 306 on gas chromatography/mass spectrometry. They appear to be as strong as E2 in inhibiting LDL oxidation in vitro. Because of their increased hydrophobicity, they have the potential of being associated with LDL and offer promise as agents that can limit LDL oxidation, thereby contributing to cardioprotection.


Assuntos
Antioxidantes/química , Antioxidantes/farmacologia , Estradiol/química , Peroxidação de Lipídeos/efeitos dos fármacos , Lipoproteínas/sangue , Lipoproteínas/farmacologia , Cromatografia Líquida de Alta Pressão , Sulfato de Cobre/química , Estradiol/farmacologia , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Peróxido de Hidrogênio/química , Lipoproteínas HDL/farmacologia , Lipoproteínas LDL/farmacologia , Peso Molecular , Oxirredução , Espécies Reativas de Oxigênio/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA