Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Mol Neurosci ; 16: 1263369, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38035265

RESUMO

Neuroinflammation is a central mechanism of brain aging and Alzheimer's disease (AD), but the exact causes of age- and AD-related neuroinflammation are incompletely understood. One potential modulator of neuroinflammation is the enzyme adenosine deaminase acting on RNA 1 (ADAR1), which regulates the accumulation of endogenous double-stranded RNA (dsRNA), a pro-inflammatory/innate immune activator. However, the role of ADAR1 and its transcriptomic targets in astrocytes, key mediators of neuroinflammation, have not been comprehensively investigated. Here, we knock down ADAR1 in primary human astrocytes via siRNA transfection and use transcriptomics (RNA-seq) to show that this results in: (1) increased expression of type I interferon and pro-inflammatory signaling pathways and (2) an accumulation of transposable element (TE) transcripts with the potential to form dsRNA. We also show that our findings may be clinically relevant, as ADAR1 gene expression declines with brain aging and AD in humans, and this is associated with a similar increase in TE transcripts. Together, our results suggest an important role for ADAR1 in preventing pro-inflammatory activation of astrocytes in response to endogenous dsRNA with aging and AD.

2.
J Gerontol A Biol Sci Med Sci ; 77(4): 659-663, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-34687299

RESUMO

Extracellular vesicles like exosomes are secreted by numerous cell types in a variety of tissues. Exosomes have been implicated in both aging and age-related disorders like Alzheimer's disease (AD). However, how aging and AD affect exosome biogenesis within and across cell types is poorly understood. Moreover, cells acquire characteristics based on tissue niche, but the impact of tissue residence on cell type exosome biogenesis is unknown. We explored the Tabula Muris Senis, Mayo RNA-seq and Rush Religious Order Study/Memory and Aging Project data sets to characterize the cell and tissue-specific effects of aging and AD on genes involved in exosome biogenesis. Specifically, we examined the age-dependent expression (age coefficient) of genes involved in exosome biogenesis (22 genes), exosome cargo (3 genes), and senescence (5 genes). Of the 131 cell populations (cell type × tissue) studied, 95 had at least 1 exosome biogenesis gene affected by age. The most common gene/transcript increased by age was charged multivesicular body protein 2A (CHMP2A) (54 cell populations). The most common gene/transcript decreased by age was syndecan-binding protein (SDCBP) (58 cell populations). The senescence-associated genes cyclin-dependent kinase 1A (CDKN1A) and CDKN2A were not related to changes in CHMP2A and SDCBP and were altered by age in fewer cell populations. Finally, individuals with AD had decreased CHMP2A and increased SDCBP expression, opposite of what is observed during mouse aging in the absence of disease. These findings indicate that exosome biogenesis gene expression is modified by age in many cell populations mostly independent of senescence, and may be further altered in AD.


Assuntos
Doença de Alzheimer , Exossomos , Vesículas Extracelulares , Envelhecimento/genética , Envelhecimento/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Exossomos/genética , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Camundongos , Sinteninas/metabolismo
3.
Neurobiol Dis ; 159: 105493, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34464705

RESUMO

The amyloid beta (Aß) peptide is believed to play a central role in Alzheimer's disease (AD), the most common age-related neurodegenerative disorder. However, the natural, evolutionarily selected functions of Aß are incompletely understood. Here, we report that nanomolar concentrations of Aß act synergistically with known cytokines to promote pro-inflammatory activation in primary human astrocytes (a cell type increasingly implicated in brain aging and AD). Using transcriptomics (RNA-seq), we show that Aß can directly substitute for the complement component C1q in a cytokine cocktail previously shown to induce astrocyte immune activation. Furthermore, we show that astrocytes synergistically activated by Aß have a transcriptional signature similar to neurotoxic "A1" astrocytes known to accumulate with age and in AD. Interestingly, we find that this biological action of Aß at low concentrations is distinct from the transcriptome changes induced by the high/supraphysiological doses of Aß often used in in vitro studies. Collectively, our results suggest an important, cytokine-like function for Aß and a novel mechanism by which it may directly contribute to the neuroinflammation associated with brain aging and AD.


Assuntos
Envelhecimento/imunologia , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Astrócitos/imunologia , Encéfalo/imunologia , Citocinas/imunologia , Doenças Neuroinflamatórias/imunologia , Peptídeos beta-Amiloides/farmacologia , Astrócitos/efeitos dos fármacos , Complemento C1q/imunologia , Complemento C1q/farmacologia , Citocinas/farmacologia , Perfilação da Expressão Gênica , Humanos , Interleucina-1alfa/imunologia , Interleucina-1alfa/farmacologia , Fragmentos de Peptídeos/farmacologia , Cultura Primária de Células , RNA-Seq , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/farmacologia
4.
Front Nutr ; 8: 712129, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34447778

RESUMO

Aging is the greatest risk factor most diseases, including cardiovascular disorders, cancers, diabetes, and neurodegeneration, but select nutritional interventions may profoundly reduce the risk for these conditions. These interventions include calorie restriction, intermittent fasting, protein restriction, and reducing intake of certain amino acids. Certain ad libitum diets, including the Mediterranean, Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability, and Okinawan diets also promote healthy aging. Evidence indicates that these dietary strategies influence aging and healthspan by acting on the biological "hallmarks of aging" and especially upstream nutrient sensing pathways. Recent advances in "omics" technologies, including RNA-sequencing (transcriptomics), have increased our understanding of how such nutritional interventions may influence gene expression related to these biological mediators of aging, primarily in pre-clinical studies. However, whether these effects are also reflected in the human transcriptome, which may provide insight on other downstream/related cellular processes with aging, is an emerging topic. Broadly, the investigation of how these nutritional interventions influence the transcriptome may provide novel insight into pathways associated with aging, and potential targets to treat age-associated disease and increase healthspan. Therefore, the purpose of this mini review is to summarize what is known about the transcriptomic effects of key dietary/nutritional interventions in both pre-clinical models and humans, address gaps in the literature, and provide insight into future research directions.

5.
Exp Gerontol ; 152: 111451, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34147619

RESUMO

Cancer is one of the most common age-related diseases, and over one-third of cancer patients will receive chemotherapy. One frequently reported side effect of chemotherapeutic agents like doxorubicin (Doxo) is impaired cognitive function, commonly known as "chemotherapy-induced cognitive impairment (CICI)", which may mimic accelerated brain aging. The biological mechanisms underlying the adverse effects of Doxo on the brain are unclear but could involve mitochondrial dysfunction. Here, we characterized brain (hippocampal) transcriptome and cognitive/behavioral changes in young mice treated with Doxo +/- the mitochondrial therapeutic MitoQ. We found that Doxo altered transcriptome/biological processes related to synaptic transmission and neurotransmitter function, neuronal health and behavior, and that these gene expression changes were: 1) similar to key differences observed in transcriptome data on brain aging; and 2) associated with related, aging-like behavioral differences, such as decreased exploration time and impaired novel object recognition test (NOR, an index of learning/memory) performance. Interestingly, MitoQ partially prevented Doxo-induced transcriptome changes in the brain, but it had no effect on behavior or cognitive function. Collectively, our findings are consistent with the idea that chemotherapeutic agents could induce neuronal/gene expression and behavioral changes similar to those that occur during brain aging. In this context, mitochondrial therapeutics may have potential as treatments for CICI at the biological level, but their effects on behavior/cognitive function require further investigation.


Assuntos
Disfunção Cognitiva , Transcriptoma , Envelhecimento/genética , Animais , Encéfalo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/genética , Doxorrubicina , Humanos , Camundongos
6.
J Gerontol A Biol Sci Med Sci ; 76(5): 805-810, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33257951

RESUMO

Transcripts from noncoding repetitive elements (REs) in the genome may be involved in aging. However, they are often ignored in transcriptome studies on healthspan and lifespan, and their role in healthy aging interventions has not been characterized. Here, we analyze REs in RNA-seq datasets from mice subjected to robust healthspan- and lifespan-increasing interventions including calorie restriction, rapamycin, acarbose, 17-α-estradiol, and Protandim. We also examine RE transcripts in long-lived transgenic mice, and in mice subjected to a high-fat diet, and we use RNA-seq to investigate the influence of aerobic exercise on RE transcripts with aging in humans. We find that (a) healthy aging interventions/behaviors globally reduce RE transcripts, whereas aging and high-fat diet (an age-accelerating treatment) increase RE expression; and (b) reduced RE expression with healthy aging interventions is associated with biological/physiological processes mechanistically linked with aging. Our results suggest that RE transcript dysregulation and suppression are likely novel mechanisms underlying aging and healthy aging interventions, respectively.


Assuntos
Elementos de DNA Transponíveis , Envelhecimento Saudável , RNA não Traduzido , Sequências Repetitivas de Ácido Nucleico , Acarbose/farmacologia , Adolescente , Idoso , Animais , Restrição Calórica , Dieta Hiperlipídica , Medicamentos de Ervas Chinesas/farmacologia , Estradiol/farmacologia , Humanos , Camundongos , Pessoa de Meia-Idade , Sirolimo/farmacologia , Adulto Jovem
7.
Aging Cell ; 16(1): 17-26, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27660040

RESUMO

Inhibition of mammalian target of rapamycin, mTOR, extends lifespan and reduces age-related disease. It is not known what role mTOR plays in the arterial aging phenotype or if mTOR inhibition by dietary rapamycin ameliorates age-related arterial dysfunction. To explore this, young (3.8 ± 0.6 months) and old (30.3 ± 0.2 months) male B6D2F1 mice were fed a rapamycin supplemented or control diet for 6-8 weeks. Although there were few other notable changes in animal characteristics after rapamycin treatment, we found that glucose tolerance improved in old mice, but was impaired in young mice, after rapamycin supplementation (both P < 0.05). Aging increased mTOR activation in arteries evidenced by elevated S6K phosphorylation (P < 0.01), and this was reversed after rapamycin treatment in old mice (P < 0.05). Aging was also associated with impaired endothelium-dependent dilation (EDD) in the carotid artery (P < 0.05). Rapamycin improved EDD in old mice (P < 0.05). Superoxide production and NADPH oxidase expression were higher in arteries from old compared to young mice (P < 0.05), and rapamycin normalized these (P < 0.05) to levels not different from young mice. Scavenging superoxide improved carotid artery EDD in untreated (P < 0.05), but not rapamycin-treated, old mice. While aging increased large artery stiffness evidenced by increased aortic pulse-wave velocity (PWV) (P < 0.01), rapamycin treatment reduced aortic PWV (P < 0.05) and collagen content (P < 0.05) in old mice. Aortic adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and expression of the cell cycle-related proteins PTEN and p27kip were increased with rapamycin treatment in old mice (all P < 0.05). Lastly, aging resulted in augmentation of the arterial senescence marker, p19 (P < 0.05), and this was ameliorated by rapamycin treatment (P < 0.05). These results demonstrate beneficial effects of rapamycin treatment on arterial function in old mice and suggest these improvements are associated with reduced oxidative stress, AMPK activation and increased expression of proteins involved in the control of the cell cycle.


Assuntos
Envelhecimento/patologia , Ciclo Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Suplementos Nutricionais , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Sirolimo/farmacologia , Adenilato Quinase/metabolismo , Animais , Artérias/efeitos dos fármacos , Artérias/patologia , Artérias/fisiopatologia , Biomarcadores/metabolismo , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Endotélio Vascular/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Insulina/sangue , Resistência à Insulina , Masculino , Camundongos Endogâmicos C57BL , Tamanho do Órgão/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Rigidez Vascular/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
8.
J Am Coll Cardiol ; 67(13): 1556-1568, 2016 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-27150688

RESUMO

BACKGROUND: Cardiac fibrosis (CF) is associated with increased ventricular stiffness and diastolic dysfunction and is an independent predictor of long-term clinical outcomes of patients with heart failure (HF). We previously showed that the matricellular CCN5 protein is cardioprotective via its ability to inhibit CF and preserve cardiac contractility. OBJECTIVES: This study examined the role of CCN5 in human heart failure and tested whether CCN5 can reverse established CF in an experimental model of HF induced by pressure overload. METHODS: Human hearts were obtained from patients with end-stage heart failure. Extensive CF was induced by applying transverse aortic constriction for 8 weeks, which was followed by adeno-associated virus-mediated transfer of CCN5 to the heart. Eight weeks following gene transfer, cellular and molecular effects were examined. RESULTS: Expression of CCN5 was significantly decreased in failing hearts from patients with end-stage heart failure compared to nonfailing hearts. Trichrome staining and myofibroblast content measurements revealed that the established CF had been reversed by CCN5 gene transfer. Anti-CF effects of CCN5 were associated with inhibition of the transforming growth factor beta signaling pathway. CCN5 significantly inhibited endothelial-mesenchymal transition and fibroblast-to-myofibroblast transdifferentiation, which are 2 critical processes for CF progression, both in vivo and in vitro. In addition, CCN5 induced apoptosis in myofibroblasts, but not in cardiomyocytes or fibroblasts, both in vivo and in vitro. CCN5 provoked the intrinsic apoptotic pathway specifically in myofibroblasts, which may have been due the ability of CCN5 to inhibit the activity of NFκB, an antiapoptotic molecule. CONCLUSIONS: CCN5 can reverse established CF by inhibiting the generation of and enhancing apoptosis of myofibroblasts in the myocardium. CCN5 may provide a novel platform for the development of targeted anti-CF therapies.


Assuntos
Proteínas de Sinalização Intercelular CCN/metabolismo , Miocárdio/patologia , Proteínas Repressoras/metabolismo , Animais , Apoptose , Proteínas de Sinalização Intercelular CCN/genética , Transdiferenciação Celular , Dependovirus , Regulação para Baixo , Transição Epitelial-Mesenquimal , Fibrose , Terapia Genética , Vetores Genéticos , Insuficiência Cardíaca/metabolismo , Humanos , Camundongos Transgênicos , Miocárdio/metabolismo , Miofibroblastos/patologia , Proteínas Repressoras/genética , Fator de Crescimento Transformador beta/metabolismo
9.
J Mol Cell Cardiol ; 53(2): 223-32, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22668785

RESUMO

Stem cell and gene therapies are being pursued as strategies for repairing damaged cardiac tissue following myocardial infarction in an attempt to prevent heart failure. The chemokine receptor-4 (CXCR4) and its ligand, CXCL12, play a critical role in stem cell recruitment post-acute myocardial infarction. Whereas progenitor cell migration via the CXCL12/CXCR4 axis is well characterized, little is known about the molecular mechanisms of CXCR4 mediated modulation of cardiac hypertrophy and failure. We used gene therapy to test the effects of CXCR4 gene delivery on adverse ventricular remodeling due to pressure overload. We assessed the effect of cardiac overexpression of CXCR4 during trans-aortic constriction (TAC) using a cardiotropic adeno-associated viral vector (AAV9) carrying the CXCR4 gene. Cardiac overexpression of CXCR4 in mice with pressure overload prevented ventricular remodeling, preserved capillary density and maintained function as determined by echocardiography and in vivo hemodynamics. In isolated adult rat cardiac myocytes, CXCL12 treatment prevented isoproterenol induced hypertrophy and interrupted the calcineurin/NFAT pathway. Finally, a complex involving the L-type calcium channel, ß2-adrenoceptor, and CXCR4 (Cav1.2/ß2AR/CXCR4) was identified in healthy cardiac myocytes and was shown to dissociate as a consequence of heart failure. CXCR4 administered to the heart via gene transfer prevents pressure overload induced heart failure. The identification of CXCR4 participation in a Cav1.2-ß2AR regulatory complex provides further insight into the mechanism by which CXCR4 modulates calcium homeostasis and chronic pressure overload responses in the cardiac myocyte. Together these results suggest that AAV9.CXCR4 gene therapy is a potential therapeutic approach for congestive heart failure.


Assuntos
Quimiocina CXCL12/farmacologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/terapia , Receptores CXCR4/metabolismo , Animais , Western Blotting , Calcineurina/metabolismo , Canais de Cálcio Tipo L/metabolismo , Cardiomegalia/genética , Cardiomegalia/metabolismo , Ensaio de Imunoadsorção Enzimática , Insuficiência Cardíaca/genética , Hemodinâmica/efeitos dos fármacos , Imunoprecipitação , Isoproterenol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Receptores Adrenérgicos beta 3/metabolismo , Receptores CXCR4/genética
10.
Am J Physiol Heart Circ Physiol ; 303(3): H353-67, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22610174

RESUMO

Diabetic cardiomyopathy is characterized, in part, by calcium handling imbalances associated with ventricular dysfunction. The cardiac Na(+)/Ca(2+) exchanger 1 (NCX1) has been implicated as a compensatory mechanism in response to reduced contractility in the heart; however, its role in diabetic cardiomyopathy remains unknown. We aimed to fully characterize the Akita(ins2) murine model of type 1 diabetes through assessing cardiac function and NCX1 regulation. The CXCL12/CXCR4 chemokine axis is well described in its cardioprotective effects via progenitor cell recruitment postacute myocardial infarction; however, it also functions in regulating calcium dependent processes in the cardiac myocyte. We therefore investigated the potential impact of CXCR4 in diabetic cardiomyopathy. Cardiac performance in the Akita(ins2) mouse was monitored using echocardiography and in vivo pressure-volume analysis. The Akita(ins2) mouse is protected against ventricular systolic failure evident at both 5 and 12 mo of age. However, the preserved contractility was associated with a decreased sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA2a)/phospholamban ratio and increased NCX1 content. Direct myocardial injection of adenovirus encoding anti-sense NCX1 significantly decreased NCX1 expression and induced systolic failure in the Akita(ins2) mouse. CXCL12 and CXCR4 were both upregulated in the Akita(ins2) heart, along with an increase in IκB-α and NF-κB p65 phosphorylation. We demonstrated that CXCR4 activation upregulates NCX1 expression through a NF-κB-dependent signaling pathway in the cardiac myocyte. In conclusion, the Akita(ins2) type 1 diabetic model is protected against systolic failure due to increased NCX1 expression. In addition, our studies reveal a novel role of CXCR4 in the diabetic heart by regulating NCX1 expression via a NF-κB-dependent mechanism.


Assuntos
Diabetes Mellitus Tipo 1/complicações , Cardiomiopatias Diabéticas/etiologia , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Receptores CXCR4/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Disfunção Ventricular Esquerda/prevenção & controle , Potenciais de Ação , Animais , Cálcio/metabolismo , Células Cultivadas , Quimiocina CXCL12/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Cardiomiopatias Diabéticas/diagnóstico por imagem , Cardiomiopatias Diabéticas/genética , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/fisiopatologia , Cardiomiopatias Diabéticas/prevenção & controle , Diástole , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Hemodinâmica , Insulina/genética , Masculino , Camundongos , Fosforilação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Trocador de Sódio e Cálcio/genética , Sístole , Ultrassonografia , Regulação para Cima , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda
11.
Exp Gerontol ; 47(1): 45-51, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22037549

RESUMO

A senescent phenotype in endothelial cells is associated with increased apoptosis, reduced endothelial nitric oxide synthase (eNOS) and inflammation, which are implicated in arterial dysfunction and disease in humans. We tested the hypothesis that changes in microRNAs are associated with a senescent phenotype in human aortic endothelial cells (HAEC). Compared with early-passage HAEC, late-passage HAEC had a reduced proliferation rate and increased staining for senescence-associated beta-galactosidase and the tumor suppressor p16(INK4a). Late-passage senescent HAEC had reduced expression of proliferation-stimulating/apoptosis-suppressing miR-21, miR-214 and miR-92 and increased expression of tumor suppressors and apoptotic markers. eNOS-suppressing miR-221 and miR-222 were increased and eNOS protein and eNOS activation (phosphorylation at serine1177) were lower in senescent HAEC. Caveolin-1 inhibiting miR-133a was reduced and caveolin-1, a negative regulator of eNOS activity, was elevated in senescent HAEC. Inflammation-repressing miR-126 was reduced and inflammation-stimulating miR-125b was increased, whereas inflammatory proteins were greater in senescent HAEC. Development of a senescent arterial endothelial cell phenotype featuring reduced cell proliferation, enhanced apoptosis and inflammation and reduced eNOS is associated with changes in miRNAs linked to the regulation of these processes. Our results support the hypothesis that miRNAs could play a critical role in arterial endothelial cell senescence.


Assuntos
Aortite/patologia , Apoptose/fisiologia , Senescência Celular/fisiologia , Células Endoteliais/patologia , MicroRNAs/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Caspases/metabolismo , Proliferação de Células , Células Cultivadas , Quimiocina CCL2/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Molécula 1 de Adesão de Célula Vascular/metabolismo
12.
Aging Cell ; 10(6): 1032-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21943306

RESUMO

We tested the hypothesis that older men who perform habitual aerobic exercise do not demonstrate age-associated vascular endothelial oxidative stress compared with their sedentary peers. Older exercising men (n=13, 62±2 years) had higher (P<0.05) physical activity (79±7 vs. 30±6 MET hours per week) and maximal exercise oxygen consumption (42±1 vs. 29±1 mL kg(-1) per minute) vs. sedentary men (n=28, 63±1 years). Brachial artery flow-mediated dilation (FMD), a measure of vascular endothelial function, was greater (P<0.05) in the exercising vs. sedentary older men (6.3±0.5 vs. 4.9±0.4%Δ) and not different than young controls (n=20, 25±1 years, 7.1±0.5%Δ). In vascular endothelial cells sampled from the brachial artery, nitrotyrosine, a marker of oxidative stress, was 51% lower in the exercising vs. sedentary older men (0.38±0.06 vs. 0.77±0.10 AU). This was associated with lower endothelial expression of the oxidant enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (p47(phox) subunit, 0.33±0.05 vs. 0.61±0.09 AU) and the redox-sensitive transcription factor nuclear factor kappa B (NFκB) (p65 subunit, 0.36±0.05 vs. 0.72±0.09 AU). Expression of the antioxidant enzyme manganese superoxide dismutase (SOD) (0.57±0.13 vs. 0.30±0.04 AU) and activity of endothelium-bound extracellular SOD were greater (6.4±0.5 vs. 5.0±0.6 U mL(-1) per minute) in the exercising men (both P<0.05), but differences no longer were significant after correcting for adiposity and circulating metabolic factors. Overall, values for the young controls differed with those for the sedentary, but not the exercising older men. Older men who exercise regularly do not demonstrate vascular endothelial oxidative stress, and this may be a key molecular mechanism underlying their reduced risk of cardiovascular diseases.


Assuntos
Envelhecimento , Artéria Braquial/metabolismo , Doenças Cardiovasculares/prevenção & controle , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Exercício Físico/fisiologia , Adulto , Artéria Braquial/citologia , Células Endoteliais/citologia , Endotélio Vascular/citologia , Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Estresse Oxidativo , Consumo de Oxigênio , Fatores de Risco , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Tirosina/análogos & derivados , Tirosina/análise , Vasodilatação
13.
J Cardiovasc Pharmacol ; 56(5): 548-59, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20729750

RESUMO

Chemokines are small secreted proteins with chemoattractant properties that play a key role in inflammation, metastasis, and embryonic development. We previously demonstrated a nonchemotactic role for one such chemokine pair, stromal cell-derived factor-1α and its G-protein coupled receptor, CXCR4. Stromal cell-derived factor-1/CXCR4 are expressed on cardiac myocytes and have direct consequences on cardiac myocyte physiology by inhibiting contractility in response to the nonselective ß-adrenergic receptor (ßAR) agonist, isoproterenol. As a result of the importance of ß-adrenergic signaling in heart failure pathophysiology, we investigated the underlying mechanism involved in CXCR4 modulation of ßAR signaling. Our studies demonstrate activation of CXCR4 by stromal cell-derived factor-1 leads to a decrease in ßAR-induced PKA activity as assessed by cAMP accumulation and PKA-dependent phosphorylation of phospholamban, an inhibitor of SERCA2a. We determined CXCR4 regulation of ßAR downstream targets is ß2AR-dependent. We demonstrated a physical interaction between CXCR4 and ß2AR as determined by coimmunoprecipitation, confocal microscopy, and BRET techniques. The CXCR4-ß2AR interaction leads to G-protein signal modulation and suggests the interaction is a novel mechanism for regulating cardiac myocyte contractility. Chemokines are physiologically and developmentally relevant to myocardial biology and represent a novel receptor class of cardiac modulators. The CXCR4-ß2AR complex could represent a hitherto unknown target for therapeutic intervention.


Assuntos
Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 2/fisiologia , Receptores CXCR4/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Técnicas In Vitro , Isoproterenol/farmacologia , Fosforilação , Ratos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA