Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Muscle Res Cell Motil ; 41(4): 297-311, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-31392564

RESUMO

Xenografts of skeletal muscle are used to study muscle repair and regeneration, mechanisms of muscular dystrophies, and potential cell therapies for musculoskeletal disorders. Typically, xenografting involves using an immunodeficient host that is pre-injured to create a niche for human cell engraftment. Cell type and method of delivery to muscle depend on the specific application, but can include myoblasts, satellite cells, induced pluripotent stem cells, mesangioblasts, immortalized muscle precursor cells, and other multipotent cell lines delivered locally or systemically. Some studies follow cell engraftment with interventions to enhance cell proliferation, migration, and differentiation into mature muscle fibers. Recently, several advances in xenografting human-derived muscle cells have been applied to study and treat Duchenne muscular dystrophy and Facioscapulohumeral muscular dystrophy. Here, we review the vast array of techniques available to aid researchers in designing future experiments aimed at creating robust muscle xenografts in rodent hosts.


Assuntos
Transplante de Células/métodos , Músculo Esquelético/metabolismo , Animais , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
2.
Am J Pathol ; 185(6): 1686-98, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25920768

RESUMO

Mutations in the dysferlin gene (DYSF) lead to human muscular dystrophies known as dysferlinopathies. The dysferlin-deficient A/J mouse develops a mild myopathy after 6 months of age, and when younger models the subclinical phase of the human disease. We subjected the tibialis anterior muscle of 3- to 4-month-old A/J mice to in vivo large-strain injury (LSI) from lengthening contractions and studied the progression of torque loss, myofiber damage, and inflammation afterward. We report that myofiber damage in A/J mice occurs before inflammatory cell infiltration. Peak edema and inflammation, monitored by magnetic resonance imaging and by immunofluorescence labeling of neutrophils and macrophages, respectively, develop 24 to 72 hours after LSI, well after the appearance of damaged myofibers. Cytokine profiles 72 hours after injury are consistent with extensive macrophage infiltration. Dysferlin-sufficient A/WySnJ mice show much less myofiber damage and inflammation and lesser cytokine levels after LSI than do A/J mice. Partial suppression of macrophage infiltration by systemic administration of clodronate-incorporated liposomes fails to suppress LSI-induced damage or to accelerate torque recovery in A/J mice. The findings from our studies suggest that, although macrophage infiltration is prominent in dysferlin-deficient A/J muscle after LSI, it is the consequence and not the cause of progressive myofiber damage.


Assuntos
Inflamação/patologia , Macrófagos/patologia , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/patologia , Animais , Modelos Animais de Doenças , Disferlina , Inflamação/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Músculo Esquelético/lesões , Músculo Esquelético/metabolismo , Distrofia Muscular do Cíngulo dos Membros/metabolismo
3.
Proc Natl Acad Sci U S A ; 110(51): 20831-6, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24302765

RESUMO

Dysferlinopathies, most commonly limb girdle muscular dystrophy 2B and Miyoshi myopathy, are degenerative myopathies caused by mutations in the DYSF gene encoding the protein dysferlin. Studies of dysferlin have focused on its role in the repair of the sarcolemma of skeletal muscle, but dysferlin's association with calcium (Ca(2+)) signaling proteins in the transverse (t-) tubules suggests additional roles. Here, we reveal that dysferlin is enriched in the t-tubule membrane of mature skeletal muscle fibers. Following experimental membrane stress in vitro, dysferlin-deficient muscle fibers undergo extensive functional and structural disruption of the t-tubules that is ameliorated by reducing external [Ca(2+)] or blocking L-type Ca(2+) channels with diltiazem. Furthermore, we demonstrate that diltiazem treatment of dysferlin-deficient mice significantly reduces eccentric contraction-induced t-tubule damage, inflammation, and necrosis, which resulted in a concomitant increase in postinjury functional recovery. Our discovery of dysferlin as a t-tubule protein that stabilizes stress-induced Ca(2+) signaling offers a therapeutic avenue for limb girdle muscular dystrophy 2B and Miyoshi myopathy patients.


Assuntos
Sinalização do Cálcio , Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Estresse Fisiológico , Animais , Anti-Hipertensivos/farmacologia , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Membrana Celular/patologia , Diltiazem/farmacologia , Disferlina , Proteínas de Membrana/genética , Camundongos , Camundongos Mutantes , Contração Muscular/efeitos dos fármacos , Contração Muscular/genética , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/patologia , Necrose/genética , Necrose/metabolismo , Necrose/patologia
4.
Muscle Nerve ; 50(2): 286-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24639380

RESUMO

INTRODUCTION: We adopted a proteomics-based approach to gain insights into phenotypic differences between A/J and B10.SJL murine dysferlinopathy models. METHODS: We optimized immunoblotting of dysferlin by preparing homogenates of the tibialis anterior (TA) muscle under several different conditions. We compared TA muscles of control, A/J, and B10.SJL mice for levels of dysferlin; dysferlin's partners MG53, annexin-A2, and caveolin-3; and the endoplasmic reticulum (ER) stress marker CHOP. We performed immunoelectron microscopy on control rat TA muscle to determine the precise location of dysferlin. RESULTS: RIPA (radioimmunoprecipitation assay) buffer and sonication improves immunoblotting of dysferlin. The ER stress marker CHOP is elevated in A/J muscle. Dysferlin is localized mostly to membranes close to the Z-disk that have been reported to be part of the Golgi, ER, and sarcoplasmic reticulum (SR) networks. CONCLUSIONS: ER stress might underlie phenotypic differences between A/J and B10.SJL mice and play a role in human dysferlinopathies.


Assuntos
Immunoblotting , Distrofia Muscular do Cíngulo dos Membros/diagnóstico , Distrofia Muscular do Cíngulo dos Membros/fisiopatologia , Fenótipo , Animais , Anexina A2/metabolismo , Proteínas de Transporte/metabolismo , Caveolina 3/metabolismo , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/fisiologia , Proteínas de Membrana , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Ensaio de Radioimunoprecipitação , Especificidade da Espécie , Fator de Transcrição CHOP/metabolismo
5.
Cell Transplant ; 33: 9636897241242624, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38600801

RESUMO

Xenografts of human skeletal muscle generated in mice can be used to study muscle pathology and to test drugs designed to treat myopathies and muscular dystrophies for their efficacy and specificity in human tissue. We previously developed methods to generate mature human skeletal muscles in immunocompromised mice starting with human myogenic precursor cells (hMPCs) from healthy individuals and individuals with facioscapulohumeral muscular dystrophy (FSHD). Here, we examine a series of alternative treatments at each stage in order to optimize engraftment. We show that (i) X-irradiation at 25Gy is optimal in preventing regeneration of murine muscle while supporting robust engraftment and the formation of human fibers without significant murine contamination; (ii) hMPC lines differ in their capacity to engraft; (iii) some hMPC lines yield grafts that respond better to intermittent neuromuscular electrical stimulation (iNMES) than others; (iv) some lines engraft better in male than in female mice; (v) coinjection of hMPCs with laminin, gelatin, Matrigel, or Growdex does not improve engraftment; (vi) BaCl2 is an acceptable replacement for cardiotoxin, but other snake venom preparations and toxins, including the major component of cardiotoxin, cytotoxin 5, are not; and (vii) generating grafts in both hindlimbs followed by iNMES of each limb yields more robust grafts than housing mice in cages with running wheels. Our results suggest that replacing cardiotoxin with BaCl2 and engrafting both tibialis anterior muscles generates robust grafts of adult human muscle tissue in mice.


Assuntos
Cardiotoxinas , Distrofia Muscular Facioescapuloumeral , Adulto , Humanos , Masculino , Camundongos , Feminino , Animais , Xenoenxertos , Transplante Heterólogo , Músculo Esquelético/patologia , Distrofia Muscular Facioescapuloumeral/patologia
6.
Nat Aging ; 4(2): 261-274, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38200273

RESUMO

Epigenetic 'clocks' based on DNA methylation have emerged as the most robust and widely used aging biomarkers, but conventional methods for applying them are expensive and laborious. Here we develop tagmentation-based indexing for methylation sequencing (TIME-seq), a highly multiplexed and scalable method for low-cost epigenetic clocks. Using TIME-seq, we applied multi-tissue and tissue-specific epigenetic clocks in over 1,800 mouse DNA samples from eight tissue and cell types. We show that TIME-seq clocks are accurate and robust, enriched for polycomb repressive complex 2-regulated loci, and benchmark favorably against conventional methods despite being up to 100-fold less expensive. Using dietary treatments and gene therapy, we find that TIME-seq clocks reflect diverse interventions in multiple tissues. Finally, we develop an economical human blood clock (R > 0.96, median error = 3.39 years) in 1,056 demographically representative individuals. These methods will enable more efficient epigenetic clock measurement in larger-scale human and animal studies.


Assuntos
Metilação de DNA , Trabalho de Parto , Gravidez , Feminino , Humanos , Camundongos , Animais , Metilação de DNA/genética , Epigênese Genética , Envelhecimento/genética , Epigenômica/métodos
7.
Sci Immunol ; 8(89): eadi5377, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37922340

RESUMO

Exercise enhances physical performance and reduces the risk of many disorders such as cardiovascular disease, type 2 diabetes, dementia, and cancer. Exercise characteristically incites an inflammatory response, notably in skeletal muscles. Although some effector mechanisms have been identified, regulatory elements activated in response to exercise remain obscure. Here, we have addressed the roles of Foxp3+CD4+ regulatory T cells (Tregs) in the healthful activities of exercise via immunologic, transcriptomic, histologic, metabolic, and biochemical analyses of acute and chronic exercise models in mice. Exercise rapidly induced expansion of the muscle Treg compartment, thereby guarding against overexuberant production of interferon-γ and consequent metabolic disruptions, particularly mitochondrial aberrancies. The performance-enhancing effects of exercise training were dampened in the absence of Tregs. Thus, exercise is a natural Treg booster with therapeutic potential in disease and aging contexts.


Assuntos
Diabetes Mellitus Tipo 2 , Linfócitos T Reguladores , Camundongos , Animais , Interferon gama , Diabetes Mellitus Tipo 2/metabolismo , Fatores de Transcrição/metabolismo , Mitocôndrias Musculares
8.
Aging (Albany NY) ; 12(10): 9959-9981, 2020 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-32470948

RESUMO

The severity and outcome of coronavirus disease 2019 (COVID-19) largely depends on a patient's age. Adults over 65 years of age represent 80% of hospitalizations and have a 23-fold greater risk of death than those under 65. In the clinic, COVID-19 patients most commonly present with fever, cough and dyspnea, and from there the disease can progress to acute respiratory distress syndrome, lung consolidation, cytokine release syndrome, endotheliitis, coagulopathy, multiple organ failure and death. Comorbidities such as cardiovascular disease, diabetes and obesity increase the chances of fatal disease, but they alone do not explain why age is an independent risk factor. Here, we present the molecular differences between young, middle-aged and older people that may explain why COVID-19 is a mild illness in some but life-threatening in others. We also discuss several biological age clocks that could be used in conjunction with genetic tests to identify both the mechanisms of the disease and individuals most at risk. Finally, based on these mechanisms, we discuss treatments that could increase the survival of older people, not simply by inhibiting the virus, but by restoring patients' ability to clear the infection and effectively regulate immune responses.


Assuntos
Envelhecimento/fisiologia , Infecções por Coronavirus , Epigênese Genética/fisiologia , Imunidade/fisiologia , Pandemias , Administração dos Cuidados ao Paciente/métodos , Pneumonia Viral , Idoso , Betacoronavirus/isolamento & purificação , Betacoronavirus/fisiologia , COVID-19 , Comorbidade , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/terapia , Síndrome da Liberação de Citocina/etiologia , Síndrome da Liberação de Citocina/imunologia , Humanos , Pneumonia Viral/epidemiologia , Pneumonia Viral/imunologia , Pneumonia Viral/terapia , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/imunologia , Medição de Risco , Fatores de Risco , SARS-CoV-2 , Índice de Gravidade de Doença
9.
Exp Neurol ; 320: 113011, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31306642

RESUMO

Aberrant expression of DUX4, a gene unique to humans and primates, causes Facioscapulohumeral Muscular Dystrophy-1 (FSHD), yet the pathogenic mechanism is unknown. As transgenic overexpression models have largely failed to replicate the genetic changes seen in FSHD, many studies of endogenously expressed DUX4 have been limited to patient biopsies and myogenic cell cultures, which never fully differentiate into mature muscle fibers. We have developed a method to xenograft immortalized human muscle precursor cells from patients with FSHD and first-degree relative controls into the tibialis anterior muscle compartment of immunodeficient mice, generating human muscle xenografts. We report that FSHD cells mature into organized and innervated human muscle fibers with minimal contamination of murine myonuclei. They also reconstitute the satellite cell niche within the xenografts. FSHD xenografts express DUX4 and DUX4 downstream targets, retain the 4q35 epigenetic signature of their original donors, and express a novel protein biomarker of FSHD, SLC34A2. Ours is the first scalable, mature in vivo human model of FSHD. It should be useful for studies of the pathogenic mechanism of the disease as well as for testing therapeutic strategies targeting DUX4 expression.


Assuntos
Modelos Animais de Doenças , Xenoenxertos , Distrofia Muscular Facioescapuloumeral , Mioblastos/transplante , Animais , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Músculo Esquelético/patologia , Distrofia Muscular Facioescapuloumeral/genética
10.
Physiol Rep ; 6(11): e13727, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29890050

RESUMO

B6.A-Dysfprmd /GeneJ (BLAJ) mice model human limb-girdle muscular dystrophy 2B (LGMD2B), which is linked to mutations in the dysferlin (DYSF) gene. We tested the hypothesis that, the calcium ion (Ca2+ ) channel blocker diltiazem (DTZ), reduces contraction-induced skeletal muscle damage, in BLAJ mice. We randomly assigned mice (N = 12; 3-4 month old males) to one of two groups - DTZ (N = 6) or vehicle (VEH, distilled water, N = 6). We conditioned mice with either DTZ or VEH for 1 week, after which, their tibialis anterior (TA) muscles were tested for contractile torque and susceptibility to injury from forced eccentric contractions. We continued dosing with DTZ or VEH for 3 days following eccentric contractions, and then studied torque recovery and muscle damage. We analyzed contractile torque before eccentric contractions, immediately after eccentric contractions, and at 3 days after eccentric contractions; and counted damaged fibers in the injured and uninjured TA muscles. We found that DTZ improved contractile torque before and immediately after forced eccentric contractions, but did not reduce delayed-onset muscle damage that was observed at 3 days after eccentric contractions.


Assuntos
Bloqueadores dos Canais de Cálcio/administração & dosagem , Diltiazem/administração & dosagem , Disferlina/genética , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular do Cíngulo dos Membros/prevenção & controle , Distrofia Muscular do Cíngulo dos Membros/fisiopatologia , Animais , Modelos Animais de Doenças , Masculino , Camundongos Knockout , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular do Cíngulo dos Membros/genética
11.
Appl Physiol Nutr Metab ; 41(10): 1108-1111, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27628198

RESUMO

We performed a placebo-controlled pre-clinical study to determine if sodium 4-phenylbutyrate (4PB) can reduce contraction-induced myofiber damage in the mdx mouse model of Duchenne muscular dystrophy (DMD). At 72 h post-eccentric contractions, 4PB significantly increased contractile torque and reduced myofiber damage and macrophage infiltration. We conclude that 4PB, which is approved by Health Canada (Pheburane) and the United States Food and Drug Administration (Buphenyl) for urea cycle disorders, might modify disease severity in patients with DMD.


Assuntos
Inibidores de Histona Desacetilases/uso terapêutico , Ativação de Macrófagos/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular de Duchenne/tratamento farmacológico , Manipulações Musculoesqueléticas/efeitos adversos , Miofibrilas/efeitos dos fármacos , Fenilbutiratos/uso terapêutico , Animais , Membro Posterior , Inibidores de Histona Desacetilases/administração & dosagem , Injeções Intraperitoneais , Traumatismos da Perna/prevenção & controle , Masculino , Camundongos Endogâmicos mdx , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/imunologia , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/imunologia , Distrofia Muscular de Duchenne/patologia , Miofibrilas/imunologia , Miofibrilas/patologia , Fenilbutiratos/administração & dosagem , Torque
12.
Skelet Muscle ; 6: 4, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26925213

RESUMO

BACKGROUND: Studies of the pathogenic mechanisms underlying human myopathies and muscular dystrophies often require animal models, but models of some human diseases are not yet available. Methods to promote the engraftment and development of myogenic cells from individuals with such diseases in mice would accelerate such studies and also provide a useful tool for testing therapeutics. Here, we investigate the ability of immortalized human myogenic precursor cells (hMPCs) to form mature human myofibers following implantation into the hindlimbs of non-obese diabetic-Rag1 (null) IL2rγ (null) (NOD-Rag)-immunodeficient mice. RESULTS: We report that hindlimbs of NOD-Rag mice that are X-irradiated, treated with cardiotoxin, and then injected with immortalized control hMPCs or hMPCs from an individual with facioscapulohumeral muscular dystrophy (FSHD) develop mature human myofibers. Furthermore, intermittent neuromuscular electrical stimulation (iNMES) of the peroneal nerve of the engrafted limb enhances the development of mature fibers in the grafts formed by both immortal cell lines. With control cells, iNMES increases the number and size of the human myofibers that form and promotes closer fiber-to-fiber packing. The human myofibers in the graft are innervated, fully differentiated, and minimally contaminated with murine myonuclei. CONCLUSIONS: Our results indicate that control and FSHD human myofibers can form in mice engrafted with hMPCs and that iNMES enhances engraftment and subsequent development of mature human muscle.


Assuntos
Diferenciação Celular , Estimulação Elétrica/métodos , Desenvolvimento Muscular , Distrofia Muscular Facioescapuloumeral/patologia , Mioblastos Esqueléticos/transplante , Junção Neuromuscular , Nervo Fibular , Adulto , Animais , Biomarcadores/metabolismo , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Sobrevivência de Enxerto , Xenoenxertos , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Distrofia Muscular Facioescapuloumeral/metabolismo , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA