Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-36834246

RESUMO

This study examined the acute effects of high-intensity resistance exercise with blood flow restriction (BFR) on performance and fatigue, metabolic stress, and markers of inflammation (interleukin-6 (IL-6)), muscle damage (myoglobin), angiogenesis (vascular endothelial growth factor (VEGF)). Thirteen resistance-trained participants (four female, 24.8 ± 4.7 years) performed four sets of barbell back-squats (75% 1RM) to failure under two conditions: blood flow restriction (BFR, bilateral 80% occlusion pressure) and control (CTRL). Completed repetitions and pre-post-exercise changes in maximal voluntary isometric contractions, countermovement jump, barbell mean propulsive velocity, and surface electromyography were recorded. Pre-post blood lactate (BLa) and venous blood samples for analysis of IL-6, myoglobin, and VEGF were collected. Ratings of perceived exertion (RPE) and pain were recorded for each set. Fewer repetitions were performed during BFR (25.5 ± 9.6 reps) compared to CTRL (43.4 ± 14.2 reps, p < 0.001), with greater repetitions performed during sets 1, 2, and 4 (p < 0.05) in CTRL. Although RPE between conditions was similar across all sets (p > 0.05), pain was greater in BFR across all sets (p < 0.05). Post-exercise fatigue was comparable between conditions. BLa was significantly greater in CTRL compared to BFR at two minutes (p = 0.001) but not four minutes post-exercise (p = 0.063). IL-6 was significantly elevated following BFR (p = 0.011). Comparable increases in myoglobin (p > 0.05) and no changes in VEGF were observed (p > 0.05). BFR increases the rate of muscular fatigue during high-intensity resistance exercise and acutely enhances IL-6 response, with significantly less total work performed, but increases pain perception, limiting implementation.


Assuntos
Treinamento Resistido , Fator A de Crescimento do Endotélio Vascular , Feminino , Humanos , Fadiga , Interleucina-6 , Músculo Esquelético/fisiologia , Mioglobina , Dor , Fluxo Sanguíneo Regional/fisiologia , Masculino
2.
Appl Physiol Nutr Metab ; 46(6): 661-668, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33337983

RESUMO

African-American (AA) women have elevated predominance of inflammatory diseases concurrent with local inflammation resulting in compromised metabolic function. The purpose of the study was 2-fold: 1) to examine the gene and protein expression of pro- and anti-inflammatory cytokine secretion by peripheral blood mononuclear cells (PBMC) obtained from AA and Caucasian-American (CA) women in response to an acute high-fat meal; and 2) to explore the influence of race (AA vs. CA) on PBMC reactivity. Ten AA and 11 CA women consumed a high-fat meal with baseline and 4 h postprandial venous blood draws. PBMCs were incubated for 3 h then messenger RNA expression and supernatant protein concentration was used to examine inflammatory profiles. All women had a postprandial increase in interleukin (IL)-8 gene expression, IL-8 protein concentration, and tumor necrosis factor alpha (TNF-α) protein concentration (P < 0.05). AA women had a postprandial increase in IL-6, IL-8, and TNF-α protein concentration (P < 0.05). AA women had higher postprandial IL-1ß protein concentration and IL-8 gene expression compared with CA women (P < 0.05). Our data uncovers the specific impact of race and time on pro-inflammatory PBMC (IL-1ß, IL-6, IL-8, and TNF-α) expression profiles in response to an acute high-fat meal challenge. Novelty: African Americans have higher predominance of inflammatory disease. We explored the potential race impact on peripheral blood mononuclear cell reactivity in response to a meal. A pro-inflammatory response to an acute high-fat meal with race impact was observed possibly contributing to health disparities impacting African-American women.


Assuntos
Negro ou Afro-Americano , Citocinas/sangue , Gorduras na Dieta/administração & dosagem , Leucócitos Mononucleares/metabolismo , Adolescente , Adulto , Citocinas/genética , Feminino , Expressão Gênica , Humanos , Interleucina-1beta/sangue , Interleucina-6/sangue , Interleucina-8/sangue , Interleucina-8/genética , Kentucky , Pessoa de Meia-Idade , Período Pós-Prandial , RNA Mensageiro/sangue , Fator de Necrose Tumoral alfa/sangue
3.
Can J Physiol Pharmacol ; 93(9): 755-63, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26176406

RESUMO

Paraoxanase-1 (PON1) is an HDL-associated enzyme that contributes to the antioxidant and antiatherosclerotic properties of HDL. Lack of PON1 results in dysfunctional HDL. HHcy is a risk factor for cardiovascular disorders, and instigates vascular dysfunction and ECM remodeling. Although studies have reported HHcy during atherosclerosis, the exact mechanism is unclear. Here, we hypothesize that dysfunctional HDL due to lack of PON1 contributes to endothelial impairment and atherogenesis through HHcy-induced ECM re-modeling. To verify this hypothesis, we used C57BL6/J and PON1 knockout mice (KO) and fed them an atherogenic diet. The expression of Akt, ADMA, and DDAH, as well as endothelial gap junction proteins such as Cx-37 and Cx-40 and eNOS was measured for vascular dysfunction and inflammation. We observed that cardiac function was decreased and plasma Hcy levels were increased in PON1 KO mice fed the atherogenic diet compared with the controls. Expression of Akt, eNOS, DDAH, Cx-37, and Cx-40 was decreased, and the expression of MMP-9 and ADMA was increased in PON1 KO mice fed an atherogenic diet compared with the controls. Our results suggest that HHcy plays an intricate role in dysfunctional HDL, owing to the lack of PON1. This contributes to vascular endothelial impairment and atherosclerosis through MMP-9-induced vascular remodeling.


Assuntos
Arildialquilfosfatase/metabolismo , Aterosclerose/fisiopatologia , Hiper-Homocisteinemia/sangue , Lipoproteínas HDL/sangue , Amidoidrolases/biossíntese , Animais , Arginina/análogos & derivados , Arginina/biossíntese , Arildialquilfosfatase/deficiência , Arildialquilfosfatase/genética , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/patologia , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Conexinas/biossíntese , Dieta Aterogênica , Endotélio Vascular/metabolismo , Fibrose/induzido quimicamente , Fibrose/patologia , Masculino , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/biossíntese , Proteínas Proto-Oncogênicas c-akt/biossíntese , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/fisiopatologia , Proteína alfa-5 de Junções Comunicantes , Proteína alfa-4 de Junções Comunicantes
4.
Can J Physiol Pharmacol ; 93(7): 577-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26118387

RESUMO

INTRODUCTION: Hyperhomocysteinemia (HHcy) is associated with inflammatory diseases and is known to increase the production of reactive oxygen species (ROS), matrix metalloproteinase (MMP)-9, and inducible nitric oxide synthase, and to decrease endothelial nitric oxide production. However, the impact of HHcy on macrophage phenotype differentiation is not well-established. It has been documented that macrophages have 2 distinct phenotypes: the "classically activated/destructive" (M1), and the "alternatively activated/constructive" (M2) subtypes. We hypothesize that HHcy increases M1 macrophage differentiation through extracellular matrix metalloproteinase inducer (EMMPRIN), a known inducer of matrix metalloproteinases. METHODS: murine J774A.1 and Raw 264.7 macrophages were treated with 100 and 500 µmol/L Hcy, respectively, for 24 h. Samples were analyzed using Western blotting and immunocytochemistry. RESULTS: Homocysteine treatment increased cluster of differentiation 40 (CD40; M1 marker) in J774A.1 and Raw 264.7 macrophages. MMP-9 was induced in both cell lines. EMMPRIN protein expression was also increased in both cell lines. Blocking EMMPRIN function by pre-treating cells with anti-EMMPRIN antibody, with or without Hcy, resulted in significantly lower expression of CD40 in both cell lines by comparison with the controls. A DCFDA assay demonstrated increased ROS production in both cell lines with Hcy treatment when compared with the controls. CONCLUSION: Our results suggest that HHcy results in an increase of the M1 macrophage phenotype. This effect seems to be at least partially mediated by EMMPRIN induction.


Assuntos
Basigina/biossíntese , Diferenciação Celular/efeitos dos fármacos , Homocisteína/farmacologia , Macrófagos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Western Blotting , Antígenos CD40/biossíntese , Técnicas de Cultura de Células , Linhagem Celular , Relação Dose-Resposta a Droga , Imuno-Histoquímica , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Transdução de Sinais
5.
Biochim Biophys Acta ; 1852(5): 732-41, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25615794

RESUMO

HHcy has been implicated in elderly frailty, but the underlying mechanisms are poorly understood. Using C57 and CBS+/- mice and C2C12 cell line, we investigated mechanisms behind HHcy induced skeletal muscle weakness and fatigability. Possible alterations in metabolic capacity (levels of LDH, CS, MM-CK and COX-IV), in structural proteins (levels of dystrophin) and in mitochondrial function (ATP production) were examined. An exercise regimen was employed to reverse HHcy induced changes. CBS+/- mice exhibited more fatigability, and generated less contraction force. No significant changes in muscle morphology were observed. However, there is a corresponding reduction in large muscle fiber number in CBS+/- mice. Excess fatigability was not due to changes in key enzymes involved in metabolism, but was due to reduced ATP levels. A marginal reduction in dystrophin levels along with a decrease in mitochondrial transcription factor A (mtTFA) were observed. There was also an increase in the mir-31, and mir-494 quantities that were implicated in dystrophin and mtTFA regulation respectively. The molecular changes elevated during HHcy, with the exception of dystrophin levels, were reversed after exercise. In addition, the amount of NRF-1, one of the transcriptional regulators of mtTFA, was significantly decreased. Furthermore, there was enhancement in mir-494 levels and a concomitant decline in mtTFA protein quantity in homocysteine treated cells. These changes in C2C12 cells were also accompanied by an increase in DNMT3a and DNMT3b proteins and global DNA methylation levels. Together, these results suggest that HHcy plays a causal role in enhanced fatigability through mitochondrial dysfunction which involves epigenetic changes.


Assuntos
Epigênese Genética , Hiper-Homocisteinemia/fisiopatologia , Mitocôndrias/metabolismo , Debilidade Muscular/fisiopatologia , Músculo Esquelético/fisiopatologia , Trifosfato de Adenosina/metabolismo , Animais , Western Blotting , Linhagem Celular , Creatina Quinase Forma MM/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Proteínas de Ligação a DNA/metabolismo , Feminino , Expressão Gênica , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Técnicas In Vitro , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Proteínas Mitocondriais/metabolismo , Contração Muscular/genética , Contração Muscular/fisiologia , Debilidade Muscular/genética , Debilidade Muscular/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Fator 1 Nuclear Respiratório/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Natação , Fatores de Transcrição/metabolismo , DNA Metiltransferase 3B
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA