Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38746385

RESUMEN

BACKGROUND: There are currently no proven methods to reverse muscle loss in humans, which is caused by trauma (e.g., volumetric muscle loss, VML), genetic neuromuscular diseases (e.g., muscular dystrophies, MDs), and accelerated senescence (e.g., sarcopenia). Since muscle tissue is capable of regeneration through muscle satellite cells (MuSCs), the implantation of autologous (or other) donor MuSCs and MuSC-derived myoblasts into host muscles can promote donor-cell-derived myogenesis. Direct injection or implantation of MuSCs or MuSC-derived myoblasts into host muscles only promotes minimal donor-cell-derived myogenesis, whereas implantation of MuSCs/myoblasts along with associated muscle tissue (muscle fibers, extracellular matrix, neurovascular pathways, etc.) gives better results. METHODS: We aim to leverage the benefits of constraining donor myogenic cells within a template that resembles muscle tissue. In this paper, we present a workflow for basic and translational studies aimed at promoting donor-cell-derived myogenesis to increase functional muscle mass in mice. Our workflow involves preparing a slurry of 10% sodium alginate mixed with myogenic cells in cell culture media, extruding the cell-containing slurry into 10% calcium lactate to form tubes, and implanting the cellularized alginate tubes into host muscle. RESULTS: Our data suggest that, the extruded alginate tubes can tolerate a peak stress of 1892 ± 527 mN, that the elastic range is at ~75-125% strain beyond initial length, and that the Young's modulus (stiffness) is 14.17 ± 1.68 %/mm2. Importantly, these mechanical properties render the alginate tubes suitable for a published technique known as minimally-invasive muscle embedding (MIME) that was developed by us to implant myogenic material into host muscle. MIME involves threading donor myogenic tissue into a needle track created within a host muscle. Cellularized alginate tubes implanted into the tibialis anterior muscle of previously euthanized mice had numerous hematoxylin-stained structures similar to nuclear staining, supporting the idea that our alginate tubes can support cell seeding. Alginate tubes that were seeded with MuSCs, incubated in MuSC/myoblast growth (i.e., proliferation) media for two days, incubated in myotube differentiation media for six days, and then minced and reseeded in new dishes, were able to promote in vitro myoblast outgrowth over several days. DISCUSSION: This pilot study is limited in its translational scope because it was performed in vitro and with previously euthanized mice. Additional studies are needed to confirm that cellularized alginate tubes can promote the de novo development of donor-cell-derived muscle fibers, which can contribute to contractile force production. CONCLUSION: Alginate tubes with MuSC/myoblasts can be generated by a simple extrusion method. The alginate tubes have sufficient mechanical strength to tolerate insertion into a host muscle, in a minimally-invasive manner, through a needle track. The cellularized alginate tubes demonstrate myogenic potential since they are capable of being maintained in culture conditions for several days, after which they can still facilitate myoblast outgrowth in a dish.

2.
Microorganisms ; 11(8)2023 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-37630523

RESUMEN

Anti-SARS-CoV-2 vaccines have played a pivotal role in reducing the risk of developing severe illness from COVID-19, thus helping end the COVID-19 global public health emergency after more than three years. Intriguingly, as SARS-CoV-2 variants emerged, individuals who were fully vaccinated did get infected in high numbers, and viral loads in vaccinated individuals were as high as those in the unvaccinated. However, even with high viral loads, vaccinated individuals were significantly less likely to develop severe illness; this begs the question as to whether the main effect of anti-SARS-CoV-2 vaccines is to confer protection against severe illness or immunity against infection. The answer to this question is consequential, not only to the understanding of how anti-SARS-CoV-2 vaccines work, but also to public health efforts against existing and novel pathogens. In this review, we argue that immune system sensitization-desensitization rather than sterilizing immunity may explain vaccine-mediated protection against severe COVID-19 illness even when the SARS-CoV-2 viral load is high. Through the lessons learned from COVID-19, we make the case that in the disease's aftermath, public health agencies must revisit healthcare policies, including redefining the term "vaccine effectiveness."

3.
J Vis Exp ; (186)2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-36121282

RESUMEN

Progressive resistance training (PRT), which involves performing muscle contractions against progressively greater external loads, can increase muscle mass and strength in healthy individuals and in patient populations. There is a need for precision rehabilitation tools to test the safety and effectiveness of PRT to maintain and/or restore muscle mass and strength in preclinical studies on small and large animal models. The PRT methodology and device described in this article can be used to perform dosage-adjusted resistance training (DART). The DART device can be used as a standalone dynamometer to objectively assess the concentric contractile torque generated by the ankle dorsiflexors in mice or can be added to a pre-existing isokinetic dynamometry system. The DART device can be fabricated with a standard 3D printer based on the instructions and open-source 3D print files provided in this work. The article also describes the workflow for a study to compare contraction-induced muscle damage caused by a single bout of DART to muscle damage caused by a comparable bout of isometric contractions (ISOM) in a mouse model of limb-girdle muscular dystrophy type 2B/R2 (BLAJ mice). The data from eight BLAJ mice (four animals for each condition) suggest that less than 10% of the tibialis anterior (TA) muscle was damaged from a single bout of DART or ISOM, with DART being less damaging than ISOM.


Asunto(s)
Entrenamiento de Fuerza , Animales , Humanos , Contracción Isométrica/fisiología , Ratones , Contracción Muscular/fisiología , Músculo Esquelético/fisiología , Torque
4.
Curr Res Physiol ; 4: 47-59, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34746826

RESUMEN

µ-Crystallin, encoded by the CRYM gene, binds the thyroid hormones, T3 and T4. Because T3 and T4 are potent regulators of metabolism and gene expression, and CRYM levels in human skeletal muscle can vary widely, we investigated the effects of overexpression of Crym. We generated transgenic mice, Crym tg, that expressed Crym protein specifically in skeletal muscle at levels 2.6-147.5 fold higher than in controls. Muscular functions, Ca2+ transients, contractile force, fatigue, running on treadmills or wheels, were not significantly altered, although T3 levels in tibialis anterior (TA) muscle were elevated ~190-fold and serum T4 was decreased 1.2-fold. Serum T3 and thyroid stimulating hormone (TSH) levels were unaffected. Crym transgenic mice studied in metabolic chambers showed a significant decrease in the respiratory exchange ratio (RER) corresponding to a 13.7% increase in fat utilization as an energy source compared to controls. Female but not male Crym tg mice gained weight more rapidly than controls when fed high fat or high simple carbohydrate diets. Although labeling for myosin heavy chains showed no fiber type differences in TA or soleus muscles, application of machine learning algorithms revealed small but significant morphological differences between Crym tg and control soleus fibers. RNA-seq and gene ontology enrichment analysis showed a significant shift towards genes associated with slower muscle function and its metabolic correlate, ß-oxidation. Protein expression showed a similar shift, though with little overlap. Our study shows that µ-crystallin plays an important role in determining substrate utilization in mammalian muscle and that high levels of µ-crystallin are associated with a shift toward greater fat metabolism.

5.
Artículo en Inglés | MEDLINE | ID: mdl-34639569

RESUMEN

Physical activity (PA) is beneficial for the health and wellness of individuals and societies. During an infectious disease pandemic, such as the one caused by COVID-19, social distancing, quarantines, and lockdowns are used to reduce community spread of the disease. Unfortunately, such nonpharmacological interventions or physical risk mitigation measures also make it challenging to engage in PA. Reduced PA could then trigger physiological changes that affect both mental and physical health. In this regard, women are more likely to experience physical and psychological distress. PA is a safe and effective nonpharmacological modality that can help prevent and manage several mental and physical health problems when performed correctly. PA might even confer benefits that are directly related to decreasing COVID-19 morbidity and mortality in women. In this review, we summarize why optimal PA must be a priority for women during the COVID-19 pandemic. We then discuss chronic COVID-19 illness and its impact on women, which further underscores the need for worldwide preventive health strategies that include PA. Finally, we discuss the importance of vaccination against COVID-19 for women, as part of prioritizing preventive healthcare and an active lifestyle.


Asunto(s)
COVID-19 , Pandemias , Control de Enfermedades Transmisibles , Ejercicio Físico , Femenino , Humanos , SARS-CoV-2
6.
FASEB J ; 34(6): 7265-7269, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32359101

RESUMEN

As of April 20, 2020, over time, the COVID-19 pandemic has resulted in 157 970 deaths out of 2 319 066 confirmed cases, at a Case Fatality Rate of ~6.8%. With the pandemic rapidly spreading, and health delivery systems being overwhelmed, it is imperative that safe and effective pharmacotherapeutic strategies are rapidly explored to improve survival. In this paper, we use established and emerging evidence to propose a testable hypothesis that, a vicious positive feedback loop of des-Arg(9)-bradykinin- and bradykinin-mediated inflammation â†’ injury â†’ inflammation, likely precipitates life threatening respiratory complications in COVID-19. Through our hypothesis, we make the prediction that the FDA-approved molecule, icatibant, might be able to interrupt this feedback loop and, thereby, improve the clinical outcomes. This hypothesis could lead to basic, translational, and clinical studies aimed at reducing COVID-19 morbidity and mortality.


Asunto(s)
Betacoronavirus , Antagonistas del Receptor de Bradiquinina B2/uso terapéutico , Bradiquinina/análogos & derivados , Infecciones por Coronavirus/fisiopatología , Modelos Biológicos , Peptidil-Dipeptidasa A/fisiología , Neumonía Viral/fisiopatología , Receptores Virales/fisiología , Enzima Convertidora de Angiotensina 2 , Bradiquinina/farmacología , Bradiquinina/fisiología , Bradiquinina/uso terapéutico , Antagonistas del Receptor de Bradiquinina B2/farmacología , COVID-19 , Ensayos de Uso Compasivo , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/tratamiento farmacológico , Disnea/etiología , Disnea/fisiopatología , Retroalimentación Fisiológica/efectos de los fármacos , Humanos , Inflamación , Uso Fuera de lo Indicado , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/tratamiento farmacológico , Receptores de Bradiquinina/efectos de los fármacos , Receptores de Bradiquinina/fisiología , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19
7.
Muscle Nerve ; 62(3): 393-403, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32363622

RESUMEN

INTRODUCTION: Dysferlin-deficient murine muscle sustains severe damage after repeated eccentric contractions. METHODS: With a robotic dynamometer, we studied the response of dysferlin-sufficient and dysferlin-deficient mice to 12 weeks of concentrically or eccentrically biased contractions. We also studied whether concentric contractions before or after eccentric contractions reduced muscle damage in dysferlin-deficient mice. RESULTS: After 12 weeks of concentric training, there was no net gain in contractile force in dysferlin-sufficient or dysferlin-deficient mice, whereas eccentric training produced a net gain in force in both mouse strains. However, eccentric training induced more muscle damage in dysferlin-deficient vs dysferlin-sufficient mice. Although concentric training produced minimal muscle damage in dysferlin-deficient mice, it still led to a prominent increase in centrally nucleated fibers. Previous exposure to concentric contractions conferred slight protection on dysferlin-deficient muscle against damage from subsequent injurious eccentric contractions. DISCUSSION: Concentric contractions may help dysferlin-deficient muscle derive the benefits of exercise without inducing damage.


Asunto(s)
Disferlina/genética , Contracción Muscular/fisiología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Cinturas/rehabilitación , Condicionamiento Físico Animal/fisiología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Distrofia Muscular de Cinturas/fisiopatología
8.
Mil Med ; 185(Suppl 1): 423-429, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-32074337

RESUMEN

INTRODUCTION: The aim of this study was to quantify the extent of donor-cell-derived myogenesis achieved by a novel surgical technique known as Minimally Invasive Muscle Embedding (MIME). MATERIALS AND METHODS: Through MIME, we implanted a single extensor digitorum longus muscle from donor mice (N = 2) that expressed a red fluorescent protein (RFP), into the left tibialis anterior (TA) muscle of immunodeficient host mice (N = 4) that expressed a green fluorescent protein (GFP). Soon after MIME, we injected a myotoxin (barium chloride), into the host TA muscle, to trigger concerted muscle degeneration and regeneration. In lieu of MIME, we performed a SHAM procedure on the right TA muscle of the same set of animals. RESULTS: In MIME-treated muscles, 22% ± 7% and 78% ± 7% muscle fibers were RFP+ and GFP+, respectively (mean ± standard deviation); and all RFP+ fibers were positive for desmin and dystrophin. Conclusion. We conclude that MIME helps generate muscle fibers of donor origin, in host muscle.


Asunto(s)
Desmina/análisis , Distrofina/análisis , Fibras Musculares Esqueléticas/trasplante , Adhesión del Tejido/métodos , Animales , Modelos Animales de Enfermedad , Ratones , Ratones SCID , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Adhesión del Tejido/estadística & datos numéricos
9.
Arch Phys Med Rehabil ; 101(5): 917-923, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32035141

RESUMEN

The growing field of regenerative rehabilitation has great potential to improve clinical outcomes for individuals with disabilities. However, the science to elucidate the specific biological underpinnings of regenerative rehabilitation-based approaches is still in its infancy and critical questions regarding clinical translation and implementation still exist. In a recent roundtable discussion from International Consortium for Regenerative Rehabilitation stakeholders, key challenges to progress in the field were identified. The goal of this article is to summarize those discussions and to initiate a broader discussion among clinicians and scientists across the fields of regenerative medicine and rehabilitation science to ultimately progress regenerative rehabilitation from an emerging field to an established interdisciplinary one. Strategies and case studies from consortium institutions-including interdisciplinary research centers, formalized courses, degree programs, international symposia, and collaborative grants-are presented. We propose that these strategic directions have the potential to engage and train clinical practitioners and basic scientists, transform clinical practice, and, ultimately, optimize patient outcomes.


Asunto(s)
Medicina Regenerativa/tendencias , Rehabilitación/tendencias , Certificación , Congresos como Asunto , Curriculum , Becas , Humanos , Medicina Regenerativa/educación , Rehabilitación/educación
10.
Eur J Histochem ; 62(3)2018 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-30043594

RESUMEN

We report that, labeling mouse muscle tissue, with mouse monoclonal antibodies specific to slow or fast myosin heavy chain (sMyHC and fMyHC, respectively), can lead to artefactual labeling of damaged muscle fibers, as hybrid fibers (sMyHC+ and fMyHC+).  We demonstrate that such erroneous immunophenotyping of muscle may be avoided, by performing colabeling or serial-section-labeling, to identify damaged fibers. The quadriceps femoris muscle group (QF) in 7-month-old, male, C57BL/6J mice had: 1.21 ± 0.21%, 98.34 ± 1.06%, 0.07 ± 0.01%, and 0.53 ± 0.85% fibers, that were, sMyHC+, fMyHC+, hybrid, and damaged, respectively.  All fibers in the tibialis anterior muscle (TA) of 3-month-old, male, C57BL/6J mice were fMyHC+; and at 3 days after injurious eccentric contractions, there was no fiber-type shift, but ~ 18% fibers were damaged.


Asunto(s)
Anticuerpos Monoclonales , Cadenas Pesadas de Miosina , Marcadores de Afinidad , Animales , Anticuerpos Monoclonales/química , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético , Cadenas Pesadas de Miosina/química
11.
Physiol Rep ; 6(11): e13727, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29890050

RESUMEN

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.


Asunto(s)
Bloqueadores de los Canales de Calcio/administración & dosificación , Diltiazem/administración & dosificación , Disferlina/genética , Contracción Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Cinturas/prevención & control , Distrofia Muscular de Cinturas/fisiopatología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones Noqueados , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Cinturas/genética
12.
Regen Med ; 13(4): 371-374, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29611460

RESUMEN

The 6th International Symposium on Regenerative Rehabilitation, hosted by the Alliance for Regenerative Rehabilitation Research and Training (AR3T), included a preconference meeting of institutional representatives of the International Consortium of Regenerative Rehabilitation, keynote talks from distinguished scientists, platform and poster presentations from experts and trainees, panel discussions and postconference workshops. The following priorities were identified: increasing rigor in basic, preclinical and clinical studies, especially the use of better controls; developing better outcome measures for preclinical and clinical trials; focusing on developing more tissue-based interventions versus cell-based interventions; including regenerative rehabilitation in curricula of professional programs like occupational and physical therapy; and developing better instruments to quantify rehabilitative interventions.


Asunto(s)
Terapia Ocupacional , Modalidades de Fisioterapia/tendencias , Medicina Regenerativa , Animales , Congresos como Asunto , Humanos , Terapia Ocupacional/métodos , Terapia Ocupacional/tendencias , Medicina Regenerativa/métodos , Medicina Regenerativa/tendencias
13.
J Vis Exp ; (126)2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28872121

RESUMEN

Skeletal muscle possesses regenerative capacity due to tissue-resident, muscle-fiber-generating (myogenic) satellite cells (SCs), which can form new muscle fibers under the right conditions. Although SCs can be harvested from muscle tissue and cultured in vitro, the resulting myoblast cells are not very effective in promoting myogenesis when transplanted into host muscle. Surgically exposing the host muscle and grafting segments of donor muscle tissue, or the isolated muscle fibers with their SCs onto host muscle, promotes better myogenesis compared to myoblast transplantation. We have developed a novel technique that we call Minimally Invasive Muscle Embedding (MIME). MIME involves passing a surgical needle through the host muscle, drawing a piece of donor muscle tissue through the needle track, and then leaving the donor tissue embedded in the host muscle so that it may act as a source of SCs for the host muscle. Here we describe in detail the steps involved in performing MIME in an immunodeficient mouse model that expresses a green fluorescent protein (GFP) in all of its cells. Immunodeficiency in the host mouse reduces the risk of immune rejection of the donor tissue, and GFP expression enables easy identification of the host muscle fibers (GFP+) and donor-cell-derived muscle fibers (GFP-). Our pilot data suggest that MIME can be used to implant an extensor digitorum longus (EDL) muscle from a donor mouse into the tibialis anterior (TA) muscle of a host mouse. Our data also suggest that when a myotoxin (barium chloride, BaCl2) is injected into the host muscle after MIME, there is evidence of donor-cell-derived myogenesis in the host muscle, with approximately 5%, 26%, 26% and 43% of the fibers in a single host TA muscle showing no host contribution, minimal host contribution, moderate host contribution, and maximal host contribution, respectively.


Asunto(s)
Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Músculo Esquelético/trasplante , Regeneración/fisiología , Animales , Proteínas Fluorescentes Verdes/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/fisiología
14.
Appl Physiol Nutr Metab ; 41(10): 1108-1111, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27628198

RESUMEN

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.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Activación de Macrófagos/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Manipulaciones Musculoesqueléticas/efectos adversos , Miofibrillas/efectos de los fármacos , Fenilbutiratos/uso terapéutico , Animales , Miembro Posterior , Inhibidores de Histona Desacetilasas/administración & dosificación , Inyecciones Intraperitoneales , Traumatismos de la Pierna/prevención & control , Masculino , Ratones Endogámicos mdx , Contracción Muscular/efectos de los fármacos , Músculo Esquelético/inmunología , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/inmunología , Distrofia Muscular de Duchenne/patología , Miofibrillas/inmunología , Miofibrillas/patología , Fenilbutiratos/administración & dosificación , Torque
15.
Skelet Muscle ; 6: 4, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26925213

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Estimulación Eléctrica/métodos , Desarrollo de Músculos , Distrofia Muscular Facioescapulohumeral/patología , Mioblastos Esqueléticos/trasplante , Unión Neuromuscular , Nervio Peroneo , Adulto , Animales , Biomarcadores/metabolismo , Línea Celular , Proliferación Celular , Supervivencia Celular , Supervivencia de Injerto , Xenoinjertos , Humanos , Masculino , Ratones Endogámicos NOD , Ratones Transgénicos , Distrofia Muscular Facioescapulohumeral/metabolismo , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patología , Factores de Tiempo
16.
Am J Pathol ; 185(6): 1686-98, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25920768

RESUMEN

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.


Asunto(s)
Inflamación/patología , Macrófagos/patología , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/patología , Animales , Modelos Animales de Enfermedad , Disferlina , Inflamación/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/metabolismo
17.
Muscle Nerve ; 50(2): 286-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24639380

RESUMEN

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.


Asunto(s)
Immunoblotting , Distrofia Muscular de Cinturas/diagnóstico , Distrofia Muscular de Cinturas/fisiopatología , Fenotipo , Animales , Anexina A2/metabolismo , Proteínas Portadoras/metabolismo , Caveolina 3/metabolismo , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/fisiología , Proteínas de la Membrana , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Ensayo de Radioinmunoprecipitación , Especificidad de la Especie , Factor de Transcripción CHOP/metabolismo
18.
Front Physiol ; 5: 57, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24600403

RESUMEN

Oxidative stress is a critical disease modifier in the muscular dystrophies. Recently, we discovered a pathway by which mechanical stretch activates NADPH Oxidase 2 (Nox2) dependent ROS generation (X-ROS). Our work in dystrophic skeletal muscle revealed that X-ROS is excessive in dystrophin-deficient (mdx) skeletal muscle and contributes to muscle injury susceptibility, a hallmark of the dystrophic process. We also observed widespread alterations in the expression of genes associated with the X-ROS pathway and redox homeostasis in muscles from both Duchenne muscular dystrophy patients and mdx mice. As nuclear factor erythroid 2-related factor 2 (Nrf2) plays an essential role in the transcriptional regulation of genes involved in redox homeostasis, we hypothesized that Nrf2 deficiency may contribute to enhanced X-ROS signaling by reducing redox buffering. To directly test the effect of diminished Nrf2 activity, Nrf2 was genetically silenced in the A/J model of dysferlinopathy-a model with a mild histopathologic and functional phenotype. Nrf2-deficient A/J mice exhibited significant muscle-specific functional deficits, histopathologic abnormalities, and dramatically enhanced X-ROS compared to control A/J and WT mice, both with functional Nrf2. Having identified that reduced Nrf2 activity is a negative disease modifier, we propose that strategies targeting Nrf2 activation may address the generalized reduction in redox homeostasis to halt or slow dystrophic progression.

19.
Proc Natl Acad Sci U S A ; 110(51): 20831-6, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24302765

RESUMEN

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.


Asunto(s)
Señalización del Calcio , Membrana Celular/metabolismo , Proteínas de la Membrana/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Cinturas/metabolismo , Estrés Fisiológico , Animales , Antihipertensivos/farmacología , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Membrana Celular/patología , Diltiazem/farmacología , Disferlina , Proteínas de la Membrana/genética , Ratones , Ratones Mutantes , Contracción Muscular/efectos de los fármacos , Contracción Muscular/genética , Fibras Musculares Esqueléticas/patología , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Necrosis/genética , Necrosis/metabolismo , Necrosis/patología
20.
PLoS One ; 7(5): e38036, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22666441

RESUMEN

Mutations in the dysferlin gene are the cause of Limb-girdle Muscular Dystrophy type 2B and Miyoshi Myopathy. The dysferlin protein has been implicated in sarcolemmal resealing, leading to the idea that the pathophysiology of dysferlin deficiencies is due to a deficit in membrane repair. Here, we show using two different approaches that fulfilling membrane repair as asseyed by laser wounding assay is not sufficient for alleviating the dysferlin deficient pathology. First, we generated a transgenic mouse overexpressing myoferlin to test the hypothesis that myoferlin, which is homologous to dysferlin, can compensate for the absence of dysferlin. The myoferlin overexpressors show no skeletal muscle abnormalities, and crossing them with a dysferlin-deficient model rescues the membrane fusion defect present in dysferlin-deficient mice in vitro. However, myoferlin overexpression does not correct muscle histology in vivo. Second, we report that AAV-mediated transfer of a minidysferlin, previously shown to correct the membrane repair deficit in vitro, also fails to improve muscle histology. Furthermore, neither myoferlin nor the minidysferlin prevented myofiber degeneration following eccentric exercise. Our data suggest that the pathogenicity of dysferlin deficiency is not solely related to impairment in sarcolemmal repair and highlight the care needed in selecting assays to assess potential therapies for dysferlinopathies.


Asunto(s)
Membrana Celular/metabolismo , Membrana Celular/patología , Terapia Genética/métodos , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Animales , Efecto Espectador/genética , Dependovirus/genética , Disferlina , Femenino , Eliminación de Gen , Regulación de la Expresión Génica/genética , Humanos , Masculino , Fusión de Membrana/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Músculos/metabolismo , Músculos/patología , Músculos/fisiopatología , Distrofia Muscular de Cinturas/terapia , Fenotipo , Sarcolema/metabolismo , Sarcolema/patología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA