Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Blood ; 133(19): 2043-2055, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-30803992

RESUMO

The proinflammatory cytokine interferon-γ (IFN-γ) has been implicated in human hematopoietic stem and progenitor cell (HSPC) depletion in immune-mediated bone marrow failure syndromes. We show that IFN-γ specifically prevents full engagement of thrombopoietin (TPO), a primary positive regulator of HSPC survival, to its receptor (c-MPL) via steric occlusion of the low-affinity binding site, contributing to perturbation of TPO-induced signaling pathways and decreased survival of human HSPCs. Eltrombopag, a synthetic small molecule mimetic of TPO that interacts with c-MPL at a position distinct from the extracellular binding site of TPO, bypasses this inhibition, providing an explanation for its clinical activity in bone marrow failure, despite already elevated endogenous TPO levels. Thus, IFN-γ-mediated perturbation of TPO:c-MPL complex formation and the resulting inhibition of a critical pathway of growth factor cell signaling may represent a general mechanism by which IFN-γ impairs the function of human HSPCs. This understanding could have broad therapeutic implications for various disorders of chronic inflammation.


Assuntos
Benzoatos/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Hidrazinas/farmacologia , Interferon gama/metabolismo , Pirazóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Feminino , Células-Tronco Hematopoéticas/metabolismo , Xenoenxertos , Humanos , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Trombopoetina/metabolismo , Transdução de Sinais/fisiologia , Trombopoetina/metabolismo
2.
Exp Gerontol ; 103: 35-46, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29269268

RESUMO

Perivascular stromal cells, including mesenchymal stem/stromal cells (MSCs), secrete paracrine factor in response to exercise training that can facilitate improvements in muscle remodeling. This study was designed to test the capacity for muscle-resident MSCs (mMSCs) isolated from young mice to release regenerative proteins in response to mechanical strain in vitro, and subsequently determine the extent to which strain-stimulated mMSCs can enhance skeletal muscle and cognitive performance in a mouse model of uncomplicated aging. Protein arrays confirmed a robust increase in protein release at 24h following an acute bout of mechanical strain in vitro (10%, 1Hz, 5h) compared to non-strain controls. Aged (24month old), C57BL/6 mice were provided bilateral intramuscular injection of saline, non-strain control mMSCs, or mMSCs subjected to a single bout of mechanical strain in vitro (4×104). No significant changes were observed in muscle weight, myofiber size, maximal force, or satellite cell quantity at 1 or 4wks between groups. Peripheral perfusion was significantly increased in muscle at 4wks post-mMSC injection (p<0.05), yet no difference was noted between control and preconditioned mMSCs. Intramuscular injection of preconditioned mMSCs increased the number of new neurons and astrocytes in the dentate gyrus of the hippocampus compared to both control groups (p<0.05), with a trend toward an increase in water maze performance noted (p=0.07). Results from this study demonstrate that acute injection of exogenously stimulated muscle-resident stromal cells do not robustly impact aged muscle structure and function, yet increase the survival of new neurons in the hippocampus.


Assuntos
Envelhecimento/fisiologia , Transplante de Células-Tronco Mesenquimais , Músculo Esquelético/fisiologia , Neurônios/fisiologia , Animais , Feminino , Hipocampo/patologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia , Condicionamento Físico Animal , Estresse Mecânico
3.
Theranostics ; 7(16): 3876-3888, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29109784

RESUMO

Peripheral arterial disease (PAD) is a debilitating complication of diabetes mellitus (DM) that leads to thousands of injuries, amputations, and deaths each year. The use of mesenchymal stem cells (MSCs) as a regenerative therapy holds the promise of regrowing injured vasculature, helping DM patients live healthier and longer lives. We report the use of muscle-derived MSCs to treat surgically-induced hindlimb ischemia in a mouse model of type 1 diabetes (DM-1). We serially evaluate several facets of the recovery process, including αVß3 -integrin expression (a marker of angiogenesis), blood perfusion, and muscle function. We also perform microarray transcriptomics experiments to characterize the gene expression states of the MSC-treated is- chemic tissues, and compare the results with those of non-ischemic tissues, as well as ischemic tissues from a saline-treated control group. The results show a multifaceted impact of mMSCs on hindlimb ischemia. We determined that the angiogenic activity one week after mMSC treatment was enhanced by approximately 80% relative to the saline group, which resulted in relative increases in blood perfusion and muscle strength of approximately 42% and 1.7-fold, respectively. At the transcriptomics level, we found that several classes of genes were affected by mMSC treatment. The mMSCs appeared to enhance both pro-angiogenic and metabolic genes, while suppressing anti-angiogenic genes and certain genes involved in the inflammatory response. All told, mMSC treatment appears to exert far-reaching effects on the microenvironment of ischemic tissue, enabling faster and more complete recovery from vascular occlusion.


Assuntos
Angiopatias Diabéticas/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Angiopatias Diabéticas/complicações , Angiopatias Diabéticas/diagnóstico por imagem , Angiopatias Diabéticas/fisiopatologia , Regulação da Expressão Gênica , Processamento de Imagem Assistida por Computador , Integrina alfaVbeta3/metabolismo , Isquemia/patologia , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Músculos/fisiopatologia , Neovascularização Fisiológica , Perfusão , Doença Arterial Periférica/complicações , Doença Arterial Periférica/patologia , Doença Arterial Periférica/terapia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Mudanças Depois da Morte , Proteoma/metabolismo , Distribuição Tecidual , Transcriptoma/genética
5.
Med Sci Sports Exerc ; 47(2): 315-25, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24905768

RESUMO

PURPOSE: The α7ß1 integrin is a transmembrane protein expressed in the skeletal muscle that can link the actin cytoskeleton to the surrounding basal lamina. We have previously demonstrated that transgenic mice overexpressing the α7B integrin in the skeletal muscle (MCK:α7B; α7Tg) mount an enhanced satellite cell and growth response to single or multiple bouts of eccentric exercise. In addition, interstitial stem cells characterized as mesenchymal stem cells (MSCs) accumulate in α7Tg muscle (mMSCs) in the sedentary state and after exercise. The results from these studies prompted us to determine the extent to which mMSC underlie the beneficial adaptive responses observed in α7Tg skeletal muscle after exercise. METHODS: mMSCs (Sca-1CD45) were isolated from α7Tg mice, dye-labeled, and intramuscularly injected into adult wild type recipient mice. After injection of mMSCs or saline, mice remained sedentary (SED) or were subjected to eccentric exercise training (TR) (downhill running) on a treadmill (three times per week) for 2 or 4 wk. Gastrocnemius-soleus complexes were collected 24 h after the last bout of exercise. RESULTS: mMSCs did not directly fuse with existing fibers; however, mMSCs injection enhanced Pax7 satellite cell number and myonuclear content compared with all other groups at 2 wk after exercise. Mean CSA, percentage of larger caliber fibers (>3000 µm), and grip strength were increased in mMSCs/TR compared with saline/SED and mMSCs/SED at 4 wk. mMSC transplantation did not enhance repair or growth in the absence of exercise. CONCLUSIONS: The results from this study demonstrate that mMSCs contribute to beneficial changes in satellite cell expansion and growth in α7Tg muscle after eccentric exercise. Thus, MSCs that naturally accumulate in the muscle after eccentric contractions may enhance the adaptive response to exercise.


Assuntos
Adaptação Fisiológica , Integrinas/metabolismo , Células-Tronco Mesenquimais/fisiologia , Músculo Esquelético/fisiologia , Condicionamento Físico Animal/fisiologia , Animais , Antígenos Ly , Feminino , Antígenos Comuns de Leucócito , Proteínas de Membrana , Transplante de Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos C57BL , Força Muscular , Músculo Esquelético/metabolismo
6.
Stem Cell Res Ther ; 5(3): 74, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24906706

RESUMO

INTRODUCTION: Mesenchymal stem cells (MSCs) reside in a variety of tissues and provide a stromal role in regulating progenitor cell function. Current studies focus on identifying the specific factors in the niche that can alter the MSC secretome, ultimately determining the effectiveness and timing of tissue repair. The purpose of the present study was to evaluate the extent to which substrate and mechanical strain simultaneously regulate MSC quantity, gene expression, and secretome. METHODS: MSCs (Sca-1+CD45-) isolated from murine skeletal muscle (muscle-derived MSCs, or mMSCs) via fluorescence-activated cell sorting were seeded onto laminin (LAM)- or collagen type 1 (COL)-coated membranes and exposed to a single bout of mechanical strain (10%, 1 Hz, 5 hours). RESULTS: mMSC proliferation was not directly affected by substrate or strain; however, gene expression of growth and inflammatory factors and extracellular matrix (ECM) proteins was downregulated in mMSCs grown on COL in a manner independent of strain. Focal adhesion kinase (FAK) may be involved in substrate regulation of mMSC secretome as FAK phosphorylation was significantly elevated 24 hours post-strain in mMSCs plated on LAM but not COL (P <0.05). Conditioned media (CM) from mMSCs exposed to both LAM and strain increased myoblast quantity 5.6-fold 24 hours post-treatment compared with myoblasts treated with serum-free media (P <0.05). This response was delayed in myoblasts treated with CM from mMSCs grown on COL. CONCLUSIONS: Here, we demonstrate that exposure to COL, the primary ECM component associated with tissue fibrosis, downregulates genes associated with growth and inflammation in mMSCs and delays the ability for mMSCs to stimulate myoblast proliferation.


Assuntos
Células-Tronco Mesenquimais/citologia , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/citologia , Estresse Mecânico , Animais , Western Blotting , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Proliferação de Células , Separação Celular , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Citometria de Fluxo , Laminina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Reação em Cadeia da Polimerase , Transcriptoma
7.
Am J Physiol Heart Circ Physiol ; 304(1): H72-81, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23280781

RESUMO

The α(7)ß(1)-integrin is an adhesion molecule highly expressed in skeletal muscle that can enhance regeneration in response to eccentric exercise. We have demonstrated that mesenchymal stem cells (MSCs), predominantly pericytes, accumulate in muscle (mMSCs) overexpressing the α(7B)-integrin (MCK:α(7B); α(7)Tg) and contribute to new fiber formation following exercise. Since vascularization is a common event that supports tissue remodeling, we hypothesized that the α(7)-integrin and/or mMSCs may stimulate vessel growth following eccentric exercise. Wild-type (WT) and α(7)Tg mice were subjected to single or multiple (3 times/wk, 4 wk) bouts of downhill running exercise. Additionally, 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI) -labeled mMSCs were intramuscularly injected into WT recipients. A subset of recipient mice were run downhill before injection to recapitulate the exercised microenvironment. While total number of CD31(+) vessels declined in both WT and α(7)Tg muscle following a single bout of exercise, the number of larger CD31(+) vessels with a visible lumen was preferentially increased in α(7)Tg mice following eccentric exercise training (P < 0.05). mMSC transplantation similarly increased vessel diameter and the total number of neuron-glial antigen-2 (NG2(+)) arterioles postexercise. Secretion of arteriogenic factors from mMSCs in response to mechanical strain, including epidermal growth factor and granulocyte macrophage-colony stimulating factor, may account for vessel remodeling. In conclusion, this study demonstrates that the α(7)-integrin and mMSCs contribute to increased vessel diameter size and arteriolar density in muscle in response to eccentric exercise. The information in this study has implications for the therapeutic treatment of injured muscle and disorders that result in vessel occlusion, including peripheral artery disease.


Assuntos
Capilares/fisiologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Contração Muscular , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Proteínas Angiogênicas/metabolismo , Animais , Antígenos/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Arteríolas/metabolismo , Arteríolas/fisiologia , Biomarcadores/metabolismo , Capilares/metabolismo , Feminino , Injeções Intramusculares , Cadeias alfa de Integrinas/genética , Cadeias alfa de Integrinas/metabolismo , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteoglicanas/metabolismo , Corrida , Estresse Mecânico , Fatores de Tempo
8.
PLoS One ; 7(1): e29760, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22253772

RESUMO

Eccentric, or lengthening, contractions result in injury and subsequently stimulate the activation and proliferation of satellite stem cells which are important for skeletal muscle regeneration. The discovery of alternative myogenic progenitors in skeletal muscle raises the question as to whether stem cells other than satellite cells accumulate in muscle in response to exercise and contribute to post-exercise repair and/or growth. In this study, stem cell antigen-1 (Sca-1) positive, non-hematopoetic (CD45⁻) cells were evaluated in wild type (WT) and α7 integrin transgenic (α7Tg) mouse muscle, which is resistant to injury yet liable to strain, 24 hr following a single bout of eccentric exercise. Sca-1⁺CD45⁻ stem cells were increased 2-fold in WT muscle post-exercise. The α7 integrin regulated the presence of Sca-1⁺ cells, with expansion occurring in α7Tg muscle and minimal cells present in muscle lacking the α7 integrin. Sca-1⁺CD45⁻ cells isolated from α7Tg muscle following exercise were characterized as mesenchymal-like stem cells (mMSCs), predominantly pericytes. In vitro multiaxial strain upregulated mMSC stem cells markers in the presence of laminin, but not gelatin, identifying a potential mechanistic basis for the accumulation of these cells in muscle following exercise. Transplantation of DiI-labeled mMSCs into WT muscle increased Pax7⁺ cells and facilitated formation of eMHC⁺DiI⁻ fibers. This study provides the first demonstration that mMSCs rapidly appear in skeletal muscle in an α7 integrin dependent manner post-exercise, revealing an early event that may be necessary for effective repair and/or growth following exercise. The results from this study also support a role for the α7 integrin and/or mMSCs in molecular- and cellular-based therapeutic strategies that can effectively combat disuse muscle atrophy.


Assuntos
Células-Tronco Mesenquimais/citologia , Músculo Esquelético/citologia , Condicionamento Físico Animal , Animais , Antígenos CD/metabolismo , Ataxina-1 , Ataxinas , Biomarcadores/metabolismo , Proliferação de Células/efeitos dos fármacos , Separação Celular , Células do Tecido Conjuntivo/citologia , Feminino , Gelatina/farmacologia , Cadeias alfa de Integrinas/metabolismo , Laminina/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/efeitos dos fármacos , Desenvolvimento Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fator de Transcrição PAX7/metabolismo , Pericitos/citologia , Pericitos/efeitos dos fármacos , Transplante de Células-Tronco , Estresse Mecânico , Regulação para Cima/efeitos dos fármacos
9.
J Appl Physiol (1985) ; 111(4): 1134-41, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21817112

RESUMO

Mechanical stimuli increase skeletal muscle growth in a mammalian target of rapamycin (mTOR)- and p70(S6K)-dependent manner. It has been proposed that costameric proteins at Z bands may sense and transfer tension to these initiators of protein translation, but few candidates have been identified. The purpose of this study was to determine whether a role exists for the α(7)-integrin in the activation of hypertrophic signaling and growth following eccentric exercise training. Five-week-old, wild-type (WT) and α(7)BX2-integrin transgenic (α(7)Tg) mice were randomly assigned to one of two groups: 1) sedentary (SED), or 2) exercise training (EX). Exercise training consisted of downhill running 3 sessions/wk for 4 wk (-20°, 17 m/min, 30 min). Downhill running was used to induce physiological mechanical strain. Twenty-four hours following the final training session, maximal isometric hindlimb plantar flexor force was measured. Gastrocnemius-soleus complexes were collected for further analysis of signaling changes, which included AKT, mTOR and p70(S6K), and muscle growth. Despite increased p70(S6K) activity in WT/EX, no significant changes in cross-sectional area or force were observed in WT/EX compared with WT/SED. AKT, mTOR, and p70(S6K) activation was higher, and whole muscle hypertrophy, relative muscle weight, myofibrillar protein, and force were significantly elevated in α(7)Tg/EX compared with α(7)Tg/SED. A marked increase in average myofiber cross-sectional area was observed in α(7)Tg/EX compared with all groups. Our findings demonstrate that the α(7)ß(1)-integrin sensitizes skeletal muscle to mechanical strain and subsequent growth. Thus the α(7)ß(1)-integrin may represent a novel molecular therapy for the treatment of disuse muscle atrophy.


Assuntos
Integrinas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Condicionamento Físico Animal , Animais , Feminino , Hipertrofia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miofibrilas/metabolismo , Miofibrilas/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Corrida/fisiologia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA