Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biotechniques ; 76(4): 153-160, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38334498

RESUMO

Modern approaches to discovering molecular mechanisms and validating treatments for age-related neuromusculoskeletal dysfunction typically rely on high-throughput transcriptome analysis. Previously harvested and fixed tissues offer an incredible reservoir of untapped molecular information. However, obtaining RNA from such formaldehyde-fixed neuromusculoskeletal tissues, especially fibrotic aged tissues, is technically challenging and often results in RNA degradation, chemical modification and yield reduction, prohibiting further analysis. Therefore, we developed a protocol to extract high-quality RNA from formaldehyde-fixed brain, cartilage, muscle and peripheral nerve isolated from naturally aged mice. Isolated RNA produced reliable gene expression data comparable to fresh and flash-frozen tissues and was sensitive enough to detect age-related changes, making our protocol valuable to researchers in the field of aging.


Assuntos
Formaldeído , RNA , Camundongos , Animais , Fixação de Tecidos/métodos , Transcriptoma , Encéfalo , Inclusão em Parafina/métodos , Perfilação da Expressão Gênica/métodos
2.
Geroscience ; 46(1): 1285-1302, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37535205

RESUMO

Onset and rates of sarcopenia, a disease characterized by a loss of muscle mass and function with age, vary greatly between sexes. Currently, no clinical interventions successfully arrest age-related muscle impairments since the decline is frequently multifactorial. Previously, we found that systemic transplantation of our unique adult multipotent muscle-derived stem/progenitor cells (MDSPCs) isolated from young mice-but not old-extends the health-span in DNA damage mouse models of progeria, a disease of accelerated aging. Additionally, induced neovascularization in the muscles and brain-where no transplanted cells were detected-strongly suggests a systemic therapeutic mechanism, possibly activated through circulating secreted factors. Herein, we used ZMPSTE24-deficient mice, a lamin A defect progeria model, to investigate the ability of young MDSPCs to preserve neuromuscular tissue structure and function. We show that progeroid ZMPST24-deficient mice faithfully exhibit sarcopenia and age-related metabolic dysfunction. However, systemic transplantation of young MDSPCs into ZMPSTE24-deficient progeroid mice sustained healthy function and histopathology of muscular tissues throughout their 6-month life span in a sex-specific manner. Indeed, female-but not male-mice systemically transplanted with young MDSPCs demonstrated significant preservation of muscle endurance, muscle fiber size, mitochondrial respirometry, and neuromuscular junction morphometrics. These novel findings strongly suggest that young MDSPCs modulate the systemic environment of aged animals by secreted rejuvenating factors to maintain a healthy homeostasis in a sex-specific manner and that the female muscle microenvironment remains responsive to exogenous regenerative cues in older age. This work highlights the age- and sex-related differences in neuromuscular tissue degeneration and the future prospect of preserving health in older adults with systemic regenerative treatments.


Assuntos
Células-Tronco Adultas , Progéria , Sarcopenia , Masculino , Camundongos , Feminino , Animais , Progéria/genética , Modelos Animais de Doenças , Células-Tronco Adultas/metabolismo , Músculos/metabolismo
3.
Aging Cell ; 22(4): e13782, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36734200

RESUMO

Cardiomyopathy is a progressive disease of the myocardium leading to impaired contractility. Genotoxic cancer therapies are known to be potent drivers of cardiomyopathy, whereas causes of spontaneous disease remain unclear. To test the hypothesis that endogenous genotoxic stress contributes to cardiomyopathy, we deleted the DNA repair gene Ercc1 specifically in striated muscle using a floxed allele of Ercc1 and mice expressing Cre under control of the muscle-specific creatinine kinase (Ckmm) promoter or depleted systemically (Ercc1-/D mice). Ckmm-Cre+/- ;Ercc1-/fl mice expired suddenly of heart disease by 7 months of age. As young adults, the hearts of Ckmm-Cre+/- ;Ercc1-/fl mice were structurally and functionally normal, but by 6-months-of-age, there was significant ventricular dilation, wall thinning, interstitial fibrosis, and systolic dysfunction indicative of dilated cardiomyopathy. Cardiac tissue from the tissue-specific or systemic model showed increased apoptosis and cardiac myocytes from Ckmm-Cre+/- ;Ercc1-/fl mice were hypersensitive to genotoxins, resulting in apoptosis. p53 levels and target gene expression, including several antioxidants, were increased in cardiac tissue from Ckmm-Cre+/- ;Ercc1-/fl and Ercc1-/D mice. Despite this, cardiac tissue from older mutant mice showed evidence of increased oxidative stress. Genetic or pharmacologic inhibition of p53 attenuated apoptosis and improved disease markers. Similarly, overexpression of mitochondrial-targeted catalase improved disease markers. Together, these data support the conclusion that DNA damage produced endogenously can drive cardiac disease and does so mechanistically via chronic activation of p53 and increased oxidative stress, driving cardiac myocyte apoptosis, dilated cardiomyopathy, and sudden death.


Assuntos
Cardiomiopatia Dilatada , Miócitos Cardíacos , Camundongos , Animais , Miócitos Cardíacos/metabolismo , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Miocárdio/metabolismo , Reparo do DNA
4.
Immun Ageing ; 18(1): 27, 2021 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-34098983

RESUMO

BACKGROUND: Osteoarthritis (OA) is one of the most prevalent joint diseases of advanced age and is a leading cause of disability worldwide. Ageing is a major risk factor for the articular cartilage (AC) degeneration that leads to OA, and the age-related decline in regenerative capacity accelerates OA progression. Here we demonstrate that systemic transplantation of a unique population of adult multipotent muscle-derived stem/progenitor cells (MDSPCs), isolated from young wild-type mice, into Zmpste24-/- mice (a model of Hutchinson-Gilford progeria syndrome, a condition marked by accelerated ageing), prevents ageing-related homeostatic decline of AC. RESULTS: MDSPC treatment inhibited expression of cartilage-degrading factors such as pro-inflammatory cytokines and extracellular matrix-proteinases, whereas pro-regenerative markers associated with cartilage mechanical support and tensile strength, cartilage resilience, chondrocyte proliferation and differentiation, and cartilage growth, were increased. Notably, MDSPC transplantation also increased the expression level of genes known for their key roles in immunomodulation, autophagy, stress resistance, pro-longevity, and telomere protection. Our findings also indicate that MDSPC transplantation increased proteoglycan content by regulating chondrocyte proliferation. CONCLUSIONS: Together, these findings demonstrate the ability of systemically transplanted young MDSPCs to preserve a healthy homeostasis and promote tissue regeneration at the molecular and tissue level in progeroid AC. These results highlight the therapeutic potential of systemically delivered multipotent adult stem cells to prevent age-associated AC degeneration.

5.
Aging Dis ; 12(3): 726-731, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34094638

RESUMO

Osteoarthritis (OA) is the most common and debilitating joint disease of advanced age and has no universally effective therapy. Here, we demonstrate that systemic transplantation of adult multipotent muscle-derived stem/progenitor cells (MDSPCs)-isolated from young mice-rejuvenates the knee articular cartilage (AC) of naturally aged mice. This intervention reduced expression of pro-inflammatory cytokines (Tnf and Il1a) and catabolic matrix-degrading proteinases (Mmp3 and Mmp13) in aged cartilage. Treatment with young MDSPCs also increased expression of pro-regenerative (Col2a1 and Acan) and prolongevity genes (Pot1b), including those associated with chondrocyte proliferation and differentiation, cartilage growth, and telomere protection. Indeed, the AC of MDSPC-treated mice exhibited reduced age-related histological pathologies. Importantly, the reduced mobility and arthritis-related gait dysfunctions of aged mice were also ameliorated by this treatment. Together, our findings demonstrate the rejuvenating effects of systemic transplantation of young MDSPCs on aging AC-at the molecular, tissue, and functional levels. This suggests that MDSPCs, or their secreted factors, may represent a novel therapy that can increase mobility and function in aged or OA patients.

6.
Aging Cell ; 20(4): e13337, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33728821

RESUMO

Aging drives progressive loss of the ability of tissues to recover from stress, partly through loss of somatic stem cell function and increased senescent burden. We demonstrate that bone marrow-derived mesenchymal stem cells (BM-MSCs) rapidly senescence and become dysfunctional in culture. Injection of BM-MSCs from young mice prolonged life span and health span, and conditioned media (CM) from young BM-MSCs rescued the function of aged stem cells and senescent fibroblasts. Extracellular vesicles (EVs) from young BM-MSC CM extended life span of Ercc1-/- mice similarly to injection of young BM-MSCs. Finally, treatment with EVs from MSCs generated from human ES cells reduced senescence in culture and in vivo, and improved health span. Thus, MSC EVs represent an effective and safe approach for conferring the therapeutic effects of adult stem cells, avoiding the risks of tumor development and donor cell rejection. These results demonstrate that MSC-derived EVs are highly effective senotherapeutics, slowing the progression of aging, and diseases driven by cellular senescence.


Assuntos
Envelhecimento/metabolismo , Senescência Celular/fisiologia , Vesículas Extracelulares/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Longevidade , Células-Tronco Mesenquimais/citologia , Senoterapia/metabolismo , Animais , Meios de Cultivo Condicionados/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endonucleases/genética , Endonucleases/metabolismo , Fibroblastos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Transdução de Sinais/fisiologia
7.
J Orthop Res ; 35(7): 1375-1382, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27572850

RESUMO

Mice expressing reduced levels of ERCC1-XPF (Ercc1-/Δ mice) demonstrate premature onset of age-related changes due to decreased repair of DNA damage. Muscle-derived stem/progenitor cells (MDSPCs) isolated from Ercc1-/Δ mice have an impaired capacity for cell differentiation. The mammalian target of rapamycin (mTOR) is a critical regulator of cell growth in response to nutrient, hormone, and oxygen levels. Inhibition of the mTOR pathway extends the lifespan of several species. Here, we examined the role of mTOR in regulating the MDSPC dysfunction that occurs with accelerated aging. We show that mTOR signaling pathways are activated in Ercc1-/Δ MDSPCs compared with wild-type (WT) MDSPCs. Additionally, inhibiting mTOR with rapamycin promoted autophagy and improved the myogenic differentiation capacity of the Ercc1-/Δ MDSPCs. The percent of apoptotic and senescent cells in Ercc1-/Δ MDSPC cultures was decreased upon mTOR inhibition. These results establish that mTOR signaling contributes to stem cell dysfunction and cell fate decisions in response to endogenous DNA damage. Therefore, mTOR represents a potential therapeutic target for improving defective, aged stem cells. © 2016 The Authors. Journal of Orthopaedic Research Published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 35:1375-1382, 2017.


Assuntos
Envelhecimento/fisiologia , Células-Tronco Multipotentes/fisiologia , Progéria/fisiopatologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Apoptose , Autofagia , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Endonucleases/genética , Camundongos , Desenvolvimento Muscular , Progéria/genética , Progéria/metabolismo , Transdução de Sinais , Sirolimo , Serina-Treonina Quinases TOR/antagonistas & inibidores
8.
Mol Cell Endocrinol ; 439: 328-336, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-27663076

RESUMO

The aim of the current work was to evaluate the ovarian follicle reserve and the ovarian transcriptome in Ames dwarf (df/df) mice. The results suggest a delayed ovarian aging in df/df mice compared to normal (N) mice. Although a high number of genes were differentially expressed during aging of N mice, only a small fraction of these changed with aging in df/df mice. These alterations involved more than 500 categorized biological processes. The majority of these biological processes, including inflammatory/immune responses, were up-regulated with aging in N mice, while old df/df mice were characterized by down-regulation of these same processes in comparison to age matched N mice. However, biological processes related to DNA damage and repairing were commonly down-regulated with aging in both genotypes. In conclusion, delayed ovarian aging in long-living df/df mice was associated with reduced expression of genes related to the inflammatory and immune responses.


Assuntos
Nanismo/genética , Longevidade/genética , Ovário/metabolismo , Reprodução/genética , Transcriptoma/genética , Animais , Núcleo Celular/metabolismo , Feminino , Perfilação da Expressão Gênica , Camundongos Mutantes , Modelos Biológicos , Oócitos/metabolismo , Folículo Ovariano/metabolismo , Análise de Componente Principal , Análise de Sequência de RNA
9.
J Reconstr Microsurg ; 31(4): 243-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25503421

RESUMO

BACKGROUND: Peripheral nerve injuries remain a challenging problem for microsurgeons. Direct repair is the gold standard, but often the surgeon is left with a gap that prevents tension-free repair. The use of empty tubes/conduits/allograft has resulted in regeneration of some sensory and motor function, but the results remain suboptimal compared with autograft. However, the use of nerve autograft has associated donor site morbidity and limited availability. METHODS: A review of the literature was performed to determine current biologic strategies to improve nerve regeneration after nerve repair. RESULTS: Nerve conduits, various neurotrophic factors, and stem cells are currently being studied as alternatives to the use of nerve autograft. CONCLUSIONS: Sensory and motor recovery after peripheral nerve regeneration remains suboptimal, especially in cases where primary nerve repair is not possible. Current strategies to augment nerve regeneration have focused on modulating the presence and activity of Schwann cells, either through direct implantation or by stimulating stem cells to differentiate toward Schwann cells, and through the use of neurotrophic factors to enhance the speed and quality of axon growth. Clinical studies will be necessary to determine the benefit of these strategies.


Assuntos
Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/terapia , Humanos , Microcirurgia/métodos , Fatores de Crescimento Neural/uso terapêutico , Transferência de Nervo/métodos , Procedimentos Neurocirúrgicos/métodos , Procedimentos de Cirurgia Plástica/métodos , Recuperação de Função Fisiológica , Células de Schwann/fisiologia , Transplante de Células-Tronco
10.
J Orthop Res ; 32(10): 1326-32, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24964749

RESUMO

DNA damage is a cause of age related pathologies, including osteoarthritis (OA). Excision repair cross complementation group 1 (ERCC1) is an endonuclease required for DNA damage repair. In this study we investigated the function of ERCC1 in chondrocytes and its association with the pathophysiology of OA. ERCC1 expression in normal and osteoarthritic cartilage was assessed, as were changes in ERCC1 expression in chondrocytes under catabolic stress. Inhibiting ERCC1 in chondrocytes under interleukin-1ß stimulation using small interfering RNA (siRNA) was also evaluated. Finally, cellular senescence and apoptosis were examined in relation to ERCC1 function. ERCC1 expression was decreased in OA cartilage and increased within 4 h of exposure to interleukin (IL)-1ß, but decreased after 12 h. The inhibition of ERCC1 by siRNA increased the expression of matrix metallopeptidase 13 and decreased collagen type II. ERCC1 inhibition also increased the number of apoptotic and senescent cells. The inhibition of ERCC1 in chondrocytes increased their expression of OA related proteins, apoptosis, cellular senescence, and hypertrophic-like changes which suggest that ERCC1 is critical for protecting human chondrocytes (HCs) from catabolic stresses and provides insights into the pathophysiology of OA and a potential target for its treatment. (191)


Assuntos
Apoptose , Senescência Celular , Proteínas de Ligação a DNA/fisiologia , Endonucleases/fisiologia , Osteoartrite/etiologia , Osteoartrite/fisiopatologia , Animais , Apoptose/genética , Cartilagem Articular/fisiologia , Cartilagem Articular/fisiopatologia , Senescência Celular/genética , Condrócitos/patologia , Condrócitos/fisiologia , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Endonucleases/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Osteoartrite/genética
11.
J Clin Invest ; 124(4): 1745-56, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24642464

RESUMO

Peripheral nerve injuries and neuropathies lead to profound functional deficits. Here, we have demonstrated that muscle-derived stem/progenitor cells (MDSPCs) isolated from adult human skeletal muscle (hMDSPCs) can adopt neuronal and glial phenotypes in vitro and ameliorate a critical-sized sciatic nerve injury and its associated defects in a murine model. Transplanted hMDSPCs surrounded the axonal growth cone, while hMDSPCs infiltrating the regenerating nerve differentiated into myelinating Schwann cells. Engraftment of hMDSPCs into the area of the damaged nerve promoted axonal regeneration, which led to functional recovery as measured by sustained gait improvement. Furthermore, no adverse effects were observed in these animals up to 18 months after transplantation. Following hMDSPC therapy, gastrocnemius muscles from mice exhibited substantially less muscle atrophy, an increase in muscle mass after denervation, and reorganization of motor endplates at the postsynaptic sites compared with those from PBS-treated mice. Evaluation of nerve defects in animals transplanted with vehicle-only or myoblast-like cells did not reveal histological or functional recovery. These data demonstrate the efficacy of hMDSPC-based therapy for peripheral nerve injury and suggest that hMDSPC transplantation has potential to be translated for use in human neuropathies.


Assuntos
Células-Tronco Adultas/transplante , Músculo Esquelético/citologia , Regeneração Nervosa/fisiologia , Células-Tronco Neurais/transplante , Adulto , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Xenoenxertos , Humanos , Camundongos , Atrofia Muscular/patologia , Atrofia Muscular/terapia , Regeneração Nervosa/genética , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células de Schwann/citologia , Células de Schwann/metabolismo , Nervo Isquiático/lesões , Nervo Isquiático/patologia , Nervo Isquiático/fisiopatologia , Transcriptoma , Adulto Jovem
12.
PLoS One ; 8(12): e82173, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24349213

RESUMO

Here, we demonstrated the differentiation potential of murine muscle-derived stem/progenitor cells (MDSPCs) toward myogenic, neuronal, and glial lineages. MDSPCs, following transplantation into a critical-sized sciatic nerve defect in mice, showed full regeneration with complete functional recovery of the injured peripheral nerve at 6 weeks post-implantation. However, several weeks after regeneration of the sciatic nerve, neoplastic growths were observed. The resulting tumors were malignant peripheral nerve sheath tumors (MPNSTs) with rhabdomyoblastic differentiation, expressing myogenic, neurogenic, and glial markers, common markers of human malignant triton tumors (MTTs). No signs of tumorigenesis were observed 17 weeks post-implantation of MDSPCs into the gastrocnemius muscles of dystrophic/mdx mice, or 1 year following subcutaneous or intravenous injection. While MDSPCs were not oncogenic in nature, the neoplasias were composed almost entirely of donor cells. Furthermore, cells isolated from the tumors were serially transplantable, generating tumors when reimplanted into mice. However, this transformation could be abrogated by differentiation of the cells toward the neurogenic lineage prior to implantation. These results establish that MDSPCs participated in the regeneration of the injured peripheral nerve but transformed in a microenvironment- and time-dependent manner, when they likely received concomitant neurogenic and myogenic differentiation signals. This microenvironment-specific transformation provides a useful mouse model for human MTTs and potentially some insight into the origins of this disease.


Assuntos
Células-Tronco Adultas/patologia , Transformação Celular Neoplásica/patologia , Microambiente Celular , Neurilemoma/patologia , Adulto , Animais , Diferenciação Celular , Linhagem da Célula , Separação Celular , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular , Músculo Esquelético/patologia , Regeneração Nervosa , Neurilemoma/fisiopatologia , Neurogênese , Neuroglia/citologia , Recuperação de Função Fisiológica , Células de Schwann/citologia , Nervo Isquiático/patologia , Nervo Isquiático/fisiopatologia , Nicho de Células-Tronco , Transplante de Células-Tronco
13.
PLoS One ; 8(6): e64923, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23762264

RESUMO

Human muscle-derived progenitor cells (hMDPCs) offer great promise for muscle cell-based regenerative medicine; however, prolonged ex-vivo expansion using animal sera is necessary to acquire sufficient cells for transplantation. Due to the risks associated with the use of animal sera, the development of a strategy for the ex vivo expansion of hMDPCs is required. The purpose of this study was to investigate the efficacy of using platelet-rich plasma (PRP) for the ex-vivo expansion of hMDPCs. Pre-plated MDPCs, myoendothelial cells, and pericytes are three populations of hMDPCs that we isolated by the modified pre-plate technique and Fluorescence Activated Cell Sorting (FACS), respectively. Pooled allogeneic human PRP was obtained from a local blood bank, and the effect that thrombin-activated PRP-releasate supplemented media had on the ex-vivo expansion of the hMDPCs was tested against FBS supplemented media, both in vitro and in vivo. PRP significantly enhanced short and long-term cell proliferation, with or without FBS supplementation. Antibody-neutralization of PDGF significantly blocked the mitogenic/proliferative effects that PRP had on the hMDPCs. A more stable and sustained expression of markers associated with stemness, and a decreased expression of lineage specific markers was observed in the PRP-expanded cells when compared with the FBS-expanded cells. The in vitro osteogenic, chondrogenic, and myogenic differentiation capacities of the hMDPCs were not altered when expanded in media supplemented with PRP. All populations of hMDPCs that were expanded in PRP supplemented media retained their ability to regenerate myofibers in vivo. Our data demonstrated that PRP promoted the proliferation and maintained the multi-differentiation capacities of the hMDPCs during ex-vivo expansion by maintaining the cells in an undifferentiated state. Moreover, PDGF appears to be a key contributing factor to the beneficial effect that PRP has on the proliferation of hMDPCs.


Assuntos
Células Musculares/citologia , Plasma Rico em Plaquetas/metabolismo , Células-Tronco/citologia , Idoso , Animais , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Mitógenos/farmacologia , Células Musculares/efeitos dos fármacos , Células Musculares/metabolismo , Desenvolvimento Muscular/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
14.
Stem Cell Res Ther ; 4(2): 33, 2013 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-23531345

RESUMO

INTRODUCTION: Loss of adult stem cell function during aging contributes to impaired tissue regeneration. Here, we tested the aging-related decline in regeneration potential of adult stem cells residing in the skeletal muscle. METHODS: We isolated muscle-derived stem/progenitor cells (MDSPCs) from progeroid Zmpste24-deficient mice (Zmpste24(-/-)) with accelerated aging phenotypes to investigate whether mutation in lamin A has an adverse effect on muscle stem/progenitor cell function. RESULTS: Our results indicate that MDSPCs isolated from Zmpste24(-/-) mice show reduced proliferation and myogenic differentiation. In addition, Zmpste24(-/-) MDSPCs showed impaired muscle regeneration, with a limited engraftment potential when transplanted into dystrophic muscle, compared with wild-type (WT) MDSPCs. Exposure of progeroid Zmpste24(-/-) MDSPCs to WT MDSPCs rescued the myogenic differentiation defect in vitro. CONCLUSIONS: These results demonstrate that adult stem/progenitor cell dysfunction contributes to impairment of tissue regeneration and suggest that factors secreted by functional cells are indeed important for the therapeutic effect of adult stem cells.


Assuntos
Proteínas de Membrana/metabolismo , Metaloendopeptidases/metabolismo , Músculo Esquelético/fisiologia , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Técnicas de Cocultura , Lamina Tipo A/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Metaloendopeptidases/deficiência , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Músculo Esquelético/citologia , Fenótipo , Regeneração , Transplante de Células-Tronco , Células-Tronco/citologia , Transplante Homólogo
15.
Methods Mol Biol ; 976: 53-65, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23400434

RESUMO

Our lab developed and optimized a method, known as the modified pre-plate technique, to isolate stem/progenitor cells from skeletal muscle. This method separates different populations of myogenic cells based on their propensity to adhere to a collagen I-coated surface. Based on their surface markers and stem-like properties, including self-renewal, multi-lineage differentiation, and ability to promote tissue regeneration, the last cell fraction or slowest to adhere to the collagen-coated surface (pre-plate 6; pp6) appears to be early, quiescent progenitor cells termed muscle-derived stem/progenitor cells (MDSPCs). The cell fractions preceding pp6 (pp1-5) are likely populations of more committed (differentiated) cells, including fibroblast- and myoblast-like cells. This technique may be used to isolate MDSPCs from skeletal muscle of humans or mice regardless of age, sex or disease state, although the yield of MDSPCs varies with age and health. MDSPCs can be used for regeneration of a variety of tissues including bone, articular cartilage, skeletal and cardiac muscle, and nerve. MDSPCs are currently being tested in clinical trials for treatment of urinary incontinence and myocardial infarction. MDSPCs from young mice have also been demonstrated to extend life span and healthspan in mouse models of accelerated aging through an apparent paracrine/endocrine mechanism. Here we detail methods for isolation and characterization of MDSPCs.


Assuntos
Adesão Celular/fisiologia , Separação Celular/métodos , Colágeno/metabolismo , Músculo Esquelético/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Células Cultivadas , Camundongos , Músculo Esquelético/fisiologia , Células-Tronco/fisiologia
16.
Nat Commun ; 3: 608, 2012 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-22215083

RESUMO

With ageing, there is a loss of adult stem cell function. However, there is no direct evidence that this has a causal role in ageing-related decline. We tested this using muscle-derived stem/progenitor cells (MDSPCs) in a murine progeria model. Here we show that MDSPCs from old and progeroid mice are defective in proliferation and multilineage differentiation. Intraperitoneal administration of MDSPCs, isolated from young wild-type mice, to progeroid mice confer significant lifespan and healthspan extension. The transplanted MDSPCs improve degenerative changes and vascularization in tissues where donor cells are not detected, suggesting that their therapeutic effect may be mediated by secreted factor(s). Indeed, young wild-type-MDSPCs rescue proliferation and differentiation defects of aged MDSPCs when co-cultured. These results establish that adult stem/progenitor cell dysfunction contributes to ageing-related degeneration and suggests a therapeutic potential of post-natal stem cells to extend health.


Assuntos
Músculos/metabolismo , Progéria/genética , Células-Tronco/citologia , Animais , Antígenos CD34/biossíntese , Antígenos Ly/metabolismo , Diferenciação Celular , Proliferação de Células , Técnicas de Cocultura , Colágeno/metabolismo , Reparo do DNA , Modelos Animais de Doenças , Genótipo , Humanos , Longevidade , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Osteócitos/citologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Progéria/patologia
17.
Mol Ther ; 20(3): 661-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22158056

RESUMO

Inhibition of the inhibitor of kappa B kinase (IKK)/nuclear factor-kappa B (NF-κB) pathway enhances muscle regeneration in injured and diseased skeletal muscle, but it is unclear exactly how this pathway contributes to the regeneration process. In this study, we examined the role of NF-κB in regulating the proliferation and differentiation of muscle-derived stem cells (MDSCs). MDSCs isolated from the skeletal muscles of p65(+/-) mice (haploinsufficient for the p65 subunit of NF-κB) had enhanced proliferation and myogenic differentiation compared to MDSCs isolated from wild-type (wt) littermates. In addition, selective pharmacological inhibition of IKKß, an upstream activator of NF-κB, enhanced wt MDSC differentiation into myotubes in vitro. The p65(+/-) MDSCs also displayed a higher muscle regeneration index than wt MDSCs following implantation into adult mice with muscular dystrophy. Additionally, using a muscle injury model, we observed that p65(+/-) MDSC engraftments were associated with reduced inflammation and necrosis. These results suggest that inhibition of the IKK/NF-κB pathway represents an effective approach to improve the myogenic regenerative potential of MDSCs and possibly other adult stem cell populations. Moreover, our results suggest that the improved muscle regeneration observed following inhibition of IKK/NF-κB, is mediated, at least in part, through enhanced stem cell proliferation and myogenic potential.


Assuntos
Desenvolvimento Muscular/genética , Mioblastos/metabolismo , Células-Tronco/metabolismo , Fator de Transcrição RelA/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Perfilação da Expressão Gênica , Heterozigoto , Quinase I-kappa B/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Camundongos SCID , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Mioblastos/citologia , Miosite/genética , Necrose/genética , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Células-Tronco/citologia , Fator de Transcrição RelA/metabolismo
18.
PLoS One ; 6(9): e24801, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21966370

RESUMO

BACKGROUND: Based on growing evidence that some adult multipotent cells necessary for tissue regeneration reside in the walls of blood vessels and the clinical success of vein wrapping for functional repair of nerve damage, we hypothesized that the repair of nerves via vein wrapping is mediated by cells migrating from the implanted venous grafts into the nerve bundle. METHODOLOGY/PRINCIPAL FINDINGS: To test the hypothesis, severed femoral nerves of rats were grafted with venous grafts from animals of the opposite sex. Nerve regeneration was impaired when decellularized or irradiated venous grafts were used in comparison to untreated grafts, supporting the involvement of venous graft-derived cells in peripheral nerve repair. Donor cells bearing Y chromosomes integrated into the area of the host injured nerve and participated in remyelination and nerve regeneration. The regenerated nerve exhibited proper axonal myelination, and expressed neuronal and glial cell markers. CONCLUSIONS/SIGNIFICANCE: These novel findings identify the mechanism by which vein wrapping promotes nerve regeneration.


Assuntos
Nervo Femoral/fisiopatologia , Nervo Femoral/cirurgia , Veia Femoral/transplante , Regeneração Nervosa , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Movimento Celular , Proliferação de Células , Células Cultivadas , Feminino , Nervo Femoral/lesões , Veia Femoral/citologia , Veia Femoral/efeitos da radiação , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Masculino , Microscopia Eletrônica de Transmissão , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/ultraestrutura , Proteínas de Neurofilamentos/metabolismo , Ratos , Ratos Endogâmicos F344 , Células de Schwann/metabolismo , Células de Schwann/ultraestrutura , Fatores Sexuais , Transplante Homólogo
19.
Stem Cell Res Ther ; 2(4): 31, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21745421

RESUMO

We have found that when muscle-derived stem cells (MDSCs) are implanted into a variety of tissues only a small fraction of the donor cells can be found within the regenerated tissues and the vast majority of cells are host derived. This observation has also been documented by other investigators using a variety of different stem cell types. It is speculated that the transplanted stem cells release factors that modulate repair indirectly by mobilizing the host's cells and attracting them to the injury site in a paracrine manner. This process is loosely called a 'paracrine mechanism', but its effects are not necessarily restricted to the injury site. In support of this speculation, it has been reported that increasing angiogenesis leads to an improvement of cardiac function, while inhibiting angiogenesis reduces the regeneration capacity of the stem cells in the injured vascularized tissues. This observation supports the finding that most of the cells that contribute to the repair process are indeed chemo-attracted to the injury site, potentially through host neo-angiogenesis. Since it has recently been observed that cells residing within the walls of blood vessels (endothelial cells and pericytes) appear to represent an origin for post-natal stem cells, it is tempting to hypothesize that the promotion of tissue repair, via neo-angiogenesis, involves these blood vessel-derived stem cells. For non-vascularized tissues, such as articular cartilage, the regenerative property of the injected stem cells still promotes a paracrine, or bystander, effect, which involves the resident cells found within the injured microenvironment, albeit not through the promotion of angiogenesis. In this paper, we review the current knowledge of post-natal stem cell therapy and demonstrate the influence that implanted stem cells have on the tissue regeneration and repair process. We argue that the terminal differentiation capacity of implanted stem cells is not the major determinant of the cells regenerative potential and that the paracrine effect imparted by the transplanted cells plays a greater role in the regeneration process.


Assuntos
Diferenciação Celular , Transplante de Células-Tronco , Animais , Doenças das Cartilagens/patologia , Doenças das Cartilagens/terapia , Terapia Baseada em Transplante de Células e Tecidos , Microambiente Celular , Cardiopatias/patologia , Cardiopatias/terapia , Humanos , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/fisiologia , Neovascularização Patológica , Comunicação Parácrina , Regeneração , Células-Tronco/citologia , Células-Tronco/fisiologia
20.
J Biol Chem ; 282(35): 25852-63, 2007 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-17597062

RESUMO

Recent studies have shown that myostatin, first identified as a negative regulator of skeletal muscle growth, may also be involved in the formation of fibrosis within skeletal muscle. In this study, we further explored the potential role of myostatin in skeletal muscle fibrosis, as well as its interaction with both transforming growth factor-beta1 and decorin. We discovered that myostatin stimulated fibroblast proliferation in vitro and induced its differentiation into myofibroblasts. We further found that transforming growth factor-beta1 stimulated myostatin expression, and conversely, myostatin stimulated transforming growth factor-beta1 secretion in C2C12 myoblasts. Decorin, a small leucine-rich proteoglycan, was found to neutralize the effects of myostatin in both fibroblasts and myoblasts. Moreover, decorin up-regulated the expression of follistatin, an antagonist of myostatin. The results of in vivo experiments showed that myostatin knock-out mice developed significantly less fibrosis and displayed better skeletal muscle regeneration when compared with wild-type mice at 2 and 4 weeks following gastrocnemius muscle laceration injury. In wild-type mice, we found that transforming growth factor-beta1 and myostatin co-localize in myofibers in the early stages of injury. Recombinant myostatin protein stimulated myofibers to express transforming growth factor-beta1 in skeletal muscles at early time points following injection. In summary, these findings define a fibrogenic property of myostatin and suggest the existence of co-regulatory relationships between transforming growth factor-beta1, myostatin, and decorin.


Assuntos
Diferenciação Celular , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Músculo Esquelético/lesões , Músculo Esquelético/metabolismo , Doenças Musculares/metabolismo , Mioblastos/metabolismo , Proteoglicanas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Decorina , Proteínas da Matriz Extracelular/farmacologia , Feminino , Fibroblastos/patologia , Fibrose , Folistatina/biossíntese , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Doenças Musculares/patologia , Mioblastos/patologia , Miostatina , Células NIH 3T3 , Proteoglicanas/farmacologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/deficiência , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA