Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Geroscience ; 2024 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-38761287

RESUMO

Aging is associated with the onset and progression of multiple diseases, which limit health span. Chronic low-grade inflammation in the absence of overt infection is considered the simmering source that triggers age-associated diseases. Failure of many cellular processes during aging is mechanistically linked to inflammation; however, the overall decline in the cellular homeostasis mechanism of autophagy has emerged as one of the top and significant inducers of inflammation during aging, frequently known as inflammaging. Thus, physiological or pharmacological interventions aimed at improving autophagy are considered geroprotective. Rapamycin analogs (rapalogs) are known for their ability to inhibit mTOR and thus regulate autophagy. This study assessed the efficacy of everolimus, a rapalog, in regulating inflammatory cytokine production in T cells from older adults. CD4+ T cells from older adults were treated with a physiological dose of everolimus (0.01 µM), and indices of autophagy and inflammation were assessed to gain a mechanistic understanding of the effect of everolimus on inflammation. Everolimus (Ever) upregulated autophagy and broadly alleviated inflammatory cytokines produced by multiple T cell subsets. Everolimus's ability to alleviate the cytokines produced by Th17 subsets of T cells, such as IL-17A and IL-17F, was dependent on autophagy and antioxidant signaling pathways. Repurposing the antineoplastic drug everolimus for curbing inflammaging is promising, given the drug's ability to restore multiple cellular homeostasis mechanisms.

2.
Aging Cell ; 22(11): e13996, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37837188

RESUMO

Aging promotes numerous intracellular changes in T cells that impact their effector function. Our data show that aging promotes an increase in the localization of STAT3 to the mitochondria (mitoSTAT3), which promotes changes in mitochondrial dynamics and function and T-cell cytokine production. Mechanistically, mitoSTAT3 increased the activity of aging T-cell mitochondria by increasing complex II. Limiting mitoSTAT3 using a mitochondria-targeted STAT3 inhibitor, Mtcur-1 lowered complex II activity, prevented age-induced changes in mitochondrial dynamics and function, and reduced Th17 inflammation. Exogenous expression of a constitutively phosphorylated form of STAT3 in T cells from young adults mimicked changes in mitochondrial dynamics and function in T cells from older adults and partially recapitulated aging-related cytokine profiles. Our data show the mechanistic link among mitoSTAT3, mitochondrial dynamics, function, and T-cell cytokine production.


Assuntos
Mitocôndrias , Dinâmica Mitocondrial , Mitocôndrias/metabolismo , Células Th17/metabolismo , Citocinas/metabolismo , Fator de Transcrição STAT3/metabolismo
3.
Cardiovasc Res ; 119(1): 252-267, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35420120

RESUMO

AIM: The importance of endothelial cell (EC) autophagy to vascular homeostasis in the context of health and disease is evolving. Earlier, we reported that intact EC autophagy is requisite to maintain shear-stress-induced nitric oxide (NO) generation via glycolysis-dependent purinergic signalling to endothelial NO synthase (eNOS). Here, we illustrate the translational and functional significance of these findings. METHODS AND RESULTS: First, we assessed translational relevance using older male humans and mice that exhibit blunted EC autophagy and impaired arterial function vs. adult controls. Active hyperaemia evoked by rhythmic handgrip exercise-elevated radial artery shear-rate similarly from baseline in adult and older subjects for 60 min. Compared with baseline, indexes of autophagy initiation, p-eNOSS1177 activation, and NO generation, occurred in radial artery ECs obtained from adult but not older volunteers. Regarding mice, indexes of autophagy and p-eNOSS1177 activation were robust in ECs from adult but not older animals that completed 60-min treadmill-running. Furthermore, 20 dyne • cm2 laminar shear stress × 45-min increased autophagic flux, glycolysis, ATP production, and p-eNOSS1177 in primary arterial ECs obtained from adult but not older mice. Concerning functional relevance, we next questioned whether the inability to initiate EC autophagy, glycolysis, and p-eNOSS1177in vitro precipitates arterial dysfunction ex vivo. Compromised intraluminal flow-mediated vasodilation displayed by arteries from older vs. adult mice was recapitulated in vessels from adult mice by (i) NO synthase inhibition; (ii) acute autophagy impairment using 3-methyladenine (3-MA); (iii) EC Atg3 depletion (iecAtg3KO mice); (iv) purinergic 2Y1-receptor (P2Y1-R) blockade; and (v) germline depletion of P2Y1-Rs. Importantly, P2Y1-R activation using 2-methylthio-ADP (2-Me-ADP) improved vasodilatory capacity in arteries from (i) adult mice treated with 3-MA; (ii) adult iecAtg3KO mice; and (iii) older animals with repressed EC autophagy. CONCLUSIONS: Arterial dysfunction concurrent with pharmacological, genetic, and age-associated EC autophagy compromise is improved by activating P2Y1-Rs.


Assuntos
Artérias , Força da Mão , Adulto , Humanos , Masculino , Animais , Camundongos , Receptores Purinérgicos P2Y1 , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase , Autofagia , Óxido Nítrico
5.
Front Aging ; 3: 999299, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091470
6.
Front Aging ; 3: 924003, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35928250

RESUMO

Obesity promotes the onset and progression of metabolic and inflammatory diseases such as type 2 diabetes. The chronic low-grade inflammation that occurs during obesity triggers multiple signaling mechanisms that negatively affect organismal health. One such mechanism is the persistent activation and mitochondrial translocation of STAT3, which is implicated in inflammatory pathologies and many types of cancers. STAT3 in the mitochondria (mitoSTAT3) alters electron transport chain activity, thereby influencing nutrient metabolism and immune response. PBMCs and CD4+ T cells from obese but normal glucose-tolerant (NGT) middle-aged subjects had higher phosphorylation of STAT3 on residue serine 727 and more mitochondrial accumulation of STAT3 than cells from lean subjects. To evaluate if circulating lipid overabundance in obesity is responsible for age- and sex-matched mitoSTAT3, cells from lean subjects were challenged with physiologically relevant doses of the saturated and monounsaturated fatty acids, palmitate and oleate, respectively. Fatty acid treatment caused robust accumulation of mitoSTAT3 in all cell types, which was independent of palmitate-induced impairments in autophagy. Co-treatment of cells with fatty acid and trehalose prevented STAT3 phosphorylation and mitochondrial accumulation in an autophagy-independent but cellular peroxide-dependent mechanism. Pharmacological blockade of mitoSTAT3 either by a mitochondria-targeted STAT3 inhibitor or ROS scavenging prevented obesity and fatty acid-induced production of proinflammatory cytokines IL-17A and IL-6, thus establishing a mechanistic link between mitoSTAT3 and inflammatory cytokine production.

7.
Endocrinology ; 163(10)2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35932471

RESUMO

The appreciation of metabolic regulation of T-cell function has exploded over the past decade, as has our understanding of how inflammation fuels comorbidities of obesity, including type 2 diabetes. The likelihood that obesity fundamentally alters T-cell metabolism and thus chronic obesity-associated inflammation is high, but studies testing causal relationships remain underrepresented. We searched PubMed for key words including mitochondria, obesity, T cell, type 2 diabetes, cristae, fission, fusion, redox, and reactive oxygen species to identify foundational and more recent studies that address these topics or cite foundational work. We investigated primary papers cited by reviews found in these searches and highlighted recent work with >100 citations to illustrate the state of the art in understanding mechanisms that control metabolism and thus function of various T-cell subsets in obesity. However, "popularity" of a paper over the first 5 years after publication cannot assess long-term impact; thus, some likely important work with fewer citations is also highlighted. We feature studies of human cells, supplementing with studies from animal models that suggest future directions for human cell research. This approach identified gaps in the literature that will need to be filled before we can estimate efficacy of mitochondria-targeted drugs in clinical trials to alleviate pathogenesis of obesity-associated inflammation.


Assuntos
Diabetes Mellitus Tipo 2 , Animais , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Inflamação/metabolismo , Mitocôndrias/metabolismo , Obesidade/metabolismo , Linfócitos T/metabolismo
8.
Am J Physiol Cell Physiol ; 322(4): C666-C673, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35138175

RESUMO

Redox homeostasis is elemental for the normal physiology of all cell types. Cells use multiple mechanisms to tightly regulate the redox balance. The onset and progression of many metabolic and aging-associated diseases occur due to the dysregulation of redox homeostasis. Thus, it is critical to identify and therapeutically target mechanisms that precipitate abnormalities in redox balance. Reactive oxygen species (ROS) produced within the immune cells regulate homeostasis, hyperimmune and hypoimmune cell responsiveness, apoptosis, immune response to pathogens, and tumor immunity. Immune cells have both cytosolic and organelle-specific redox regulatory systems to maintain appropriate levels of ROS. Nicotinamide nucleotide transhydrogenase (NNT) is an essential mitochondrial redox regulatory protein. Dysregulation of NNT function prevents immune cells from mounting an adequate immune response to pathogens, promotes a chronic inflammatory state associated with aging and metabolic diseases, and initiates conditions related to a dysregulated immune system such as autoimmunity. Although many studies have reported on NNT in different cell types, including cancer cells, relatively few studies have explored NNT in immune cells. This review provides an overview of NNT and focuses on the current knowledge of NNT in the immune cells.


Assuntos
NADP Trans-Hidrogenases , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , NADP Trans-Hidrogenases/genética , NADP Trans-Hidrogenases/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo
9.
Int J Phytoremediation ; 24(1): 12-24, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34000928

RESUMO

Silver nanoparticles (AgNPs) are particularly among the widely used nanomaterials in medicine, industry, and agriculture. The small size and large surface area of AgNPs and other nanomaterials result in their high reactivity in biological systems. To better understand the effects of AgNPs on plants at the molecular level, tomato (Lycopersicon esculentum L.) seedlings were exposed to 30 mg/L silver in the form of nanoparticle (AgNPs), ionic (AgNO3), or bulk (Ag0) in 50% Hoagland media for 7 days. The effects of silver on the expression of plant membrane transporters H+-ATPase, vacuolar type H+-ATPase (V-ATPase), and enzymes isocitrate dehydrogenase (IDH), and catalase in roots was assessed using RT-qPCR and immunofluorescence-confocal microscopy. We observed significantly higher expression of catalase in plants exposed to AgNPs (Fold of expression 1.1) and AgNO3 (Fold of expression 1.2) than the control group. The immunofluorescence imaging of the proteins confirmed the gene expression data; the expression of the enzyme catalase was upregulated 41, 216, and 770% higher than the control group in plants exposed to AgNPs, Ag0, and AgNO3, respectively. Exposure to AgnO3 resulted in the upregulation (fold of expression 1.2) of H+-ATPase and downregulation (fold of expression 0.7) of V-ATPase. A significant reduction in the expression of the redox-sensitive tricarboxylic cycle (TCA) enzyme mitochondrial IDH was observed in plants exposed to AgNPs (38%), AgNO3 (48%), or Ag0 (77%) compared to the control. This study shows that exposure to silver affects the expression of genes and protein involved in membrane transportation and oxidative response. The ionic form of silver had the most significant effect on the expression of genes and proteins compared to other forms of silver. The results from this study improve our understanding about the molecular effects of different forms of silver on important crop species. Novelty statementSilver nanoparticles released into the environment can be oxidized and be transformed into ionic form. Both the particulate and ionic forms of silver can be taken by plants and affect plants physiological and molecular responses. Despite the extensive research in this area, there is a scarce of information about the effects of silver nanoparticles on the expression of membrane transporters especially H+-ATPase involved in regulating cells' electrochemical charge, and the activity of enzymes involved in oxidative stress responses. This is a unique study that evaluates the expression of cellular proton transporters and enzymes of redox balance and energy metabolisms such as membrane transporters, H+-ATPase, and V-ATPases, and enzymes catalase and IDH. The results provide us valuable information about the impact of silver on plants at the molecular level by evaluating the expression of genes and proteins. Key MessageThe exposure of plants to silver as an environmental stressor affects the expression of genes and proteins involved in maintaining cell's electrochemical gradient (H+-ATPase, V-ATPase) and redox potential (IDH, catalase).


Assuntos
Nanopartículas Metálicas , Solanum lycopersicum , Biodegradação Ambiental , Solanum lycopersicum/genética , Prata/toxicidade , Nitrato de Prata
10.
Cells ; 10(8)2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34440882

RESUMO

Dysregulation of autophagy is an important underlying cause in the onset and progression of many metabolic diseases, including diabetes. Studies in animal models and humans show that impairment in the removal and the recycling of organelles, in particular, contributes to cellular damage, functional failure, and the onset of metabolic diseases. Interestingly, in certain contexts, inhibition of autophagy can be protective. While the inability to upregulate autophagy can play a critical role in the development of diseases, excessive autophagy can also be detrimental, making autophagy an intricately regulated process, the altering of which can adversely affect organismal health. Autophagy is indispensable for maintaining normal cardiac and vascular structure and function. Patients with diabetes are at a higher risk of developing and dying from vascular complications. Autophagy dysregulation is associated with the development of heart failure, many forms of cardiomyopathy, atherosclerosis, myocardial infarction, and microvascular complications in diabetic patients. Here, we review the recent findings on selective autophagy in hyperglycemia and diabetes-associated microvascular and macrovascular complications.


Assuntos
Autofagia/fisiologia , Hiperglicemia/patologia , Doenças Vasculares/patologia , Animais , Complicações do Diabetes/metabolismo , Complicações do Diabetes/patologia , Humanos , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Organelas/metabolismo , Organelas/patologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Doenças Vasculares/etiologia , Doenças Vasculares/metabolismo
11.
Am J Physiol Cell Physiol ; 320(5): C873-C879, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33689478

RESUMO

The biguanide metformin is the most commonly used antidiabetic drug. Recent studies show that metformin not only improves chronic inflammation by improving metabolic parameters but also has a direct anti-inflammatory effect. In light of these findings, it is essential to identify the inflammatory pathways targeted by metformin to develop a comprehensive understanding of the mechanisms of action of this drug. Commonly accepted mechanisms of metformin action include AMPK activation and inhibition of mTOR pathways, which are evaluated in multiple diseases. Additionally, metformin's action on mitochondrial function and cellular homeostasis processes such as autophagy is of particular interest because of the importance of these mechanisms in maintaining cellular health. Both dysregulated mitochondria and failure of the autophagy pathways, the latter of which impair clearance of dysfunctional, damaged, or excess organelles, affect cellular health drastically and can trigger the onset of metabolic and age-related diseases. Immune cells are the fundamental cell types that govern the health of an organism. Thus, dysregulation of autophagy or mitochondrial function in immune cells has a remarkable effect on susceptibility to infections, response to vaccination, tumor onset, and the development of inflammatory and autoimmune conditions. In this study, we summarize the latest research on metformin's regulation of immune cell mitochondrial function and autophagy as evidence that new clinical trials on metformin with primary outcomes related to the immune system should be considered to treat immune-mediated diseases over the near term.


Assuntos
Anti-Inflamatórios/uso terapêutico , Inflamação/tratamento farmacológico , Metformina/uso terapêutico , Mitocôndrias/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Animais , Proteínas Relacionadas à Autofagia/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Transdução de Sinais
13.
Autophagy ; 16(12): 2285-2286, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32960694

RESUMO

Striking age-related changes occur in the human immune system, beginning in the sixth decade of life. Age is a non-modifiable, universal risk factor that results in the dysregulation of many cellular homeostatic processes. The decline in immune cell macroautophagy/autophagy and the increased generation of proinflammatory cytokines during agingfuels the development of diseases in the elderly. We reported that higher Th17 inflammation during aging was secondary to dysregulation in T cell autophagy. However, the mechanism underlying lower anti-CD3 and anti-CD28 activation-induced T cell autophagy during aging remain unknown. Our data fuel the speculation that dysregulation of the glutathione (GSH) system might cause the decline in T cell autophagy in aging, additionally provoked by reactive oxygen species signaling emanating from the mitochondria.


Assuntos
Autofagia , Metformina , Idoso , Envelhecimento , Humanos , Inflamação , Mitocôndrias , Espécies Reativas de Oxigênio
14.
Cell Metab ; 32(1): 44-55.e6, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32402267

RESUMO

Age is a non-modifiable risk factor for the inflammation that underlies age-associated diseases; thus, anti-inflammaging drugs hold promise for increasing health span. Cytokine profiling and bioinformatic analyses showed that Th17 cytokine production differentiates CD4+ T cells from lean, normoglycemic older and younger subjects, and mimics a diabetes-associated Th17 profile. T cells from older compared to younger subjects also had defects in autophagy and mitochondrial bioenergetics that associate with redox imbalance. Metformin ameliorated the Th17 inflammaging profile by increasing autophagy and improving mitochondrial bioenergetics. By contrast, autophagy-targeting siRNA disrupted redox balance in T cells from young subjects and activated the Th17 profile by activating the Th17 master regulator, STAT3, which in turn bound IL-17A and F promoters. Mitophagy-targeting siRNA failed to activate the Th17 profile. We conclude that metformin improves autophagy and mitochondrial function largely in parallel to ameliorate a newly defined inflammaging profile that echoes inflammation in diabetes.


Assuntos
Envelhecimento/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Inflamação/metabolismo , Metformina/farmacologia , Mitocôndrias/efeitos dos fármacos , Adulto , Envelhecimento/metabolismo , Humanos , Pessoa de Meia-Idade , Mitocôndrias/metabolismo
15.
Biomolecules ; 9(8)2019 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-31344923

RESUMO

Elesclomol ((N-malonyl-bis(N'-methyl-N'-thiobenzoylhydrazide)); formerly STA-4783) is a mitochondria-targeted chemotherapeutic agent that has demonstrated efficacy in selective cancer cell killing in pre-clinical and clinical testing. The biologically active form of elesclomol is a deprotonated copper chelate (elesclomol:copper; E:C), which has been shown to enhance reactive oxygen species (ROS) production and induce a transcriptional gene profile characteristic of an oxidative stress response in vitro. Previous studies suggest that E:C interacts with the electron transport chain (ETC) to generate high levels of ROS within the organelle and ultimately induce cell death. The purpose of this study was to further explore the mechanism of cellular and mitochondrial toxicity of E:C by examining its direct effect on mitochondrial bioenergetic function. The results obtained indicate that E:C treatment in whole cells of non-tumorigenic origin at high concentrations (40 M and higher) induces a rapid and substantial increase in mitochondrial superoxide levels and dissipation of mitochondrial membrane potential. Furthermore, similar higher concentrations of E:C act as a direct uncoupler of oxidative phosphorylation and generalized inhibitor of electron transport activity in isolated, intact mitochondria, and induce a dose-dependent inhibition of mitochondrial NADH-ubiquinone oxidoreductase activity in freeze-thawed mitochondrial preparations. The results of this study are important in that they are the first to demonstrate a direct effect of the E:C chelate on bioenergetic function in isolated mammalian mitochondria, and suggest the possibility that the increase in ROS production and cytotoxicity induced by E:C may in part be due to uncoupling of mitochondrial oxidative phosphorylation and/or inhibition of electron transport activity. These results also provide important information about the mechanisms of mitochondrial and cellular toxicity induced by E:C and will ultimately contribute to a better understanding of the therapeutic potential of elesclomol as an anticancer compound.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Hidrazinas/farmacologia , Mitocôndrias/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Regulação para Baixo , Transporte de Elétrons/efeitos dos fármacos , Complexo I de Transporte de Elétrons/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Fosforilação Oxidativa/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
16.
Biomolecules ; 9(2)2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30823587

RESUMO

Circulating fatty acids (FAs) increase with obesity and can drive mitochondrial damage and inflammation. Nicotinamide nucleotide transhydrogenase (NNT) is a mitochondrial protein that positively regulates nicotinamide adenine dinucleotide phosphate (NADPH), a key mediator of energy transduction and redox homeostasis. The role that NNT-regulated bioenergetics play in the inflammatory response of immune cells in obesity is untested. Our objective was to determine how free fatty acids (FFAs) regulate inflammation through impacts on mitochondria and redox homeostasis of peripheral blood mononuclear cells (PBMCs). PBMCs from lean subjects were activated with a T cell-specific stimulus in the presence or absence of generally pro-inflammatory palmitate and/or non-inflammatory oleate. Palmitate decreased immune cell expression of NNT, NADPH, and anti-oxidant glutathione, but increased reactive oxygen and proinflammatory Th17 cytokines. Oleate had no effect on these outcomes. Genetic inhibition of NNT recapitulated the effects of palmitate. PBMCs from obese (BMI >30) compared to lean subjects had lower NNT and glutathione expression, and higher Th17 cytokine expression, none of which were changed by exogenous palmitate. Our data identify NNT as a palmitate-regulated rheostat of redox balance that regulates immune cell function in obesity and suggest that dietary or therapeutic strategies aimed at increasing NNT expression may restore redox balance to ameliorate obesity-associated inflammation.


Assuntos
Ácidos Graxos/farmacologia , Inflamação/tratamento farmacológico , NADP Trans-Hidrogenases/antagonistas & inibidores , Linfócitos T/efeitos dos fármacos , Adulto , Feminino , Humanos , Inflamação/metabolismo , Masculino , Pessoa de Meia-Idade , NADP Trans-Hidrogenases/genética , NADP Trans-Hidrogenases/metabolismo , Linfócitos T/metabolismo
17.
Eur J Appl Physiol ; 118(8): 1653-1660, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29846794

RESUMO

INTRODUCTION: Exercise training is recommended for improving health and protecting against the development of metabolic and cardiovascular pathologies. Combined resistance and aerobic exercise training (CRAE) has been shown to provide unique benefits in older adults with cardiovascular diseases. PURPOSE: We sought to determine the beneficial effects of CRAE in adolescent girls who are obese and hyperinsulinemic. METHODS: Forty adolescent girls who are obese (age 14.7 ± 1 years; BMI 30 ± 2) were randomly assigned to a "no exercise" (CON n = 20) or combined exercise group (EX n = 20). The EX group performed resistance and aerobic exercise for 12 weeks, 5 times per week. Exercise intensity was increased gradually, from 40 to 70% of heart rate reserve (HRR), every 4 weeks. The brachial-ankle pulse wave velocity (BaPWV), blood pressure (BP), heart rate (HR), blood leptin, adiponectin levels, and body composition were measured before and after the 12-week intervention. RESULTS: We observed that CRAE effectively reduced the body fat percentage, body weight, and waist circumference in the EX group (p < 0.05). After 12 weeks of training, subjects in the CRAE group maintained appropriate leptin and adiponectin levels and showed positive improvements of blood insulin, glucose, and insulin resistance parameters relative to baseline and to the CON group (p < 0.05). CONCLUSION: CRAE is a useful therapeutic method to alleviate metabolic risk factors in adolescent girls who are obese and hyperinsulinemic.


Assuntos
Adiposidade , Resistência à Insulina , Obesidade/terapia , Treinamento Resistido/métodos , Adiponectina/sangue , Adolescente , Glicemia/metabolismo , Pressão Sanguínea , Feminino , Frequência Cardíaca , Humanos , Insulina/sangue , Leptina/sangue
18.
Mol Nutr Food Res ; 62(2)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29024402

RESUMO

SCOPE: Lipotoxicity-induced endothelial dysfunction is an important vascular complication associated with diabetes. Clinical studies support the vascular benefits of blueberry anthocyanins, but the underlying mechanism is unclear. The hypothesis that metabolites of blueberry anthocyanins attenuate lipotoxicity-induced endothelial dysfunction was tested. METHODS AND RESULTS: Human aortic endothelial cells (HAECs) were treated for 6 h with either: (i) the parent anthocyanins (malvidin-3-glucoside and cyanidin-3-glucoside); or (ii) the blueberry metabolites (hydroxyhippuric acid, hippuric acid, benzoic acid-4-sulfate, isovanillic acid-3-sulfate, and vanillic acid-4-sulfate), at concentrations known to circulate in humans following blueberry consumption. For the last 5 h HAECs were treated with palmitate or vehicle. HAECs treated with palmitate displayed elevated reactive oxygen species generation, increased mRNA expression of NOX4, chemokines, adhesion molecules, and IκBα, exaggerated monocyte binding, and suppressed nitric oxide production. Of note, the damaging effects of palmitate were ameliorated in HAECs treated with blueberry metabolites but not parent anthocyanins. Further, important translational relevance of these results was provided by our observation that palmitate-induced endothelial dysfunction was lessened in arterial segments that incubated concurrently with blueberry metabolites. CONCLUSION: The presented findings indicate that the vascular benefits of blueberry anthocyanins are mediated by their metabolites. Blueberries might complement existing therapies to lessen vascular complications.


Assuntos
Antocianinas/farmacologia , Mirtilos Azuis (Planta)/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Ácido Palmítico/toxicidade , Animais , Aorta/citologia , Mirtilos Azuis (Planta)/química , Células Cultivadas , Células Endoteliais , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Insulina/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Espécies Reativas de Oxigênio/metabolismo
19.
Compr Physiol ; 7(4): 1307-1337, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-28915326

RESUMO

Adipose tissue (AT) is the primary energy reservoir organ, and thereby plays a critical role in energy homeostasis and regulation of metabolism. AT expands in response to chronic overnutrition or aging and becomes a major source of inflammation that has marked influence on systemic metabolism. The chronic, sterile inflammation that occurs in the AT during the development of obesity or in aging contributes to onset of devastating diseases such as insulin resistance, diabetes, and cardiovascular pathologies. Numerous studies have shown that inflammation in the visceral AT of humans and animals is a critical trigger for the development of metabolic syndrome. This work underscores the well-supported conclusion that the inflammatory immune response and metabolic pathways in the AT are tightly interwoven by multiple layers of relatively conserved mechanisms. During the development of diet-induced obesity or age-associated adiposity, cells of the innate and the adaptive immune systems infiltrate and proliferate in the AT. Macrophages, which dominate AT-associated immune cells in mouse models of obesity, but are less dominant in obese people, have been studied extensively. However, cells of the adaptive immune system, including T cells and B cells, contribute significantly to AT inflammation, perhaps more in humans than in mice. Lymphocytes regulate recruitment of innate immune cells into AT, and produce cytokines that influence the helpful-to-harmful inflammatory balance that, in turn, regulates organismal metabolism. This review describes inflammation, or more precisely, metabolic inflammation (metaflammation) with an eye toward the AT and the roles lymphocytes play in regulation of systemic metabolism during obesity and aging. © 2017 American Physiological Society. Compr Physiol 7:1307-1337, 2017.


Assuntos
Imunidade Adaptativa , Tecido Adiposo/metabolismo , Envelhecimento/imunologia , Obesidade/imunologia , Tecido Adiposo/crescimento & desenvolvimento , Tecido Adiposo/imunologia , Envelhecimento/metabolismo , Animais , Citocinas/genética , Citocinas/metabolismo , Humanos , Linfócitos/imunologia , Linfócitos/metabolismo , Obesidade/metabolismo
20.
Arterioscler Thromb Vasc Biol ; 37(9): 1646-1656, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28684613

RESUMO

OBJECTIVE: Impaired endothelial cell (EC) autophagy compromises shear stress-induced nitric oxide (NO) generation. We determined the responsible mechanism. APPROACH AND RESULTS: On autophagy compromise in bovine aortic ECs exposed to shear stress, a decrease in glucose uptake and EC glycolysis attenuated ATP production. We hypothesized that decreased glycolysis-dependent purinergic signaling via P2Y1 (P2Y purinoceptor 1) receptors, secondary to impaired autophagy in ECs, prevents shear-induced phosphorylation of eNOS (endothelial nitric oxide synthase) at its positive regulatory site S1117 (p-eNOSS1177) and NO generation. Maneuvers that restore glucose transport and glycolysis (eg, overexpression of GLUT1 [glucose transporter 1]) or purinergic signaling (eg, addition of exogenous ADP) rescue shear-induced p-eNOSS1177 and NO production in ECs with impaired autophagy. Conversely, inhibiting glucose transport via GLUT1 small interfering RNA, blocking purinergic signaling via ectonucleotidase-mediated ATP/ADP degradation (eg, apyrase), or inhibiting P2Y1 receptors using pharmacological (eg, MRS2179 [2'-deoxy-N6-methyladenosine 3',5'-bisphosphate tetrasodium salt]) or genetic (eg, P2Y1-receptor small interfering RNA) procedures inhibit shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Supporting a central role for PKCδT505 (protein kinase C delta T505) in relaying the autophagy-dependent purinergic-mediated signal to eNOS, we find that (1) shear stress-induced activating phosphorylation of PKCδT505 is negated by inhibiting autophagy, (2) shear-induced p-eNOSS1177 and NO generation are restored in autophagy-impaired ECs via pharmacological (eg, bryostatin) or genetic (eg, constitutively active PKCδ) activation of PKCδT505, and (3) pharmacological (eg, rottlerin) and genetic (eg, PKCδ small interfering RNA) PKCδ inhibition prevents shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Key nodes of dysregulation in this pathway on autophagy compromise were revealed in human arterial ECs. CONCLUSIONS: Targeted reactivation of purinergic signaling and PKCδ has strategic potential to restore compromised NO generation in pathologies associated with suppressed EC autophagy.


Assuntos
Trifosfato de Adenosina/metabolismo , Autofagia , Células Endoteliais/enzimologia , Glicólise , Mecanotransdução Celular , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Animais , Autofagia/efeitos dos fármacos , Proteínas Relacionadas à Autofagia/deficiência , Proteínas Relacionadas à Autofagia/genética , Bovinos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Mecanotransdução Celular/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Receptores Purinérgicos P2Y1/efeitos dos fármacos , Receptores Purinérgicos P2Y1/genética , Serina , Estresse Mecânico , Transfecção , Enzimas de Conjugação de Ubiquitina/deficiência , Enzimas de Conjugação de Ubiquitina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...