Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Osteoarthritis Cartilage ; 32(7): 912-921, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38642879

RESUMEN

OBJECTIVE: Exercise remains a hallmark treatment for post-traumatic osteoarthritis (PTOA) and may maintain joint homeostasis in part by clearing inflammatory cytokines, cells, and particles. It remains largely unknown whether exercise-induced joint clearance can provide therapeutic relief of PTOA. In this study, we hypothesized that exercise could slow the progression of preclinical PTOA in part by enhancing knee joint clearance. DESIGN: Surgical medial meniscal transection was used to induce PTOA in 3-month-old male Lewis rats. A sham surgery was used as a control. Mild treadmill walking was introduced 3 weeks post-surgery and maintained to 6 weeks post-surgery. Gait and isometric muscle torque were measured at the study endpoint. Near-infrared imaging tracked how exercise altered lymphatic and venous knee joint clearance during discrete time points of PTOA progression. RESULTS: Exercise mitigated joint degradation associated with PTOA by preserving glycosaminoglycan content and reducing osteophyte volume (effect size (95% Confidence Interval (CI)); 1.74 (0.71-2.26)). PTOA increased hind step widths (0.57 (0.18-0.95) cm), but exercise corrected this gait dysfunction (0.54 (0.16-0.93) cm), potentially indicating pain relief. Venous, but not lymphatic, clearance was quicker 1-, 3-, and 6-weeks post-surgery compared to baseline. The mild treadmill walking protocol expedited lymphatic clearance rate in moderate PTOA (3.39 (0.20-6.59) hrs), suggesting exercise may play a critical role in restoring joint homeostasis. CONCLUSIONS: We conclude that mild exercise has the potential to slow disease progression in part by expediting joint clearance in moderate PTOA.


Asunto(s)
Inestabilidad de la Articulación , Osteoartritis de la Rodilla , Condicionamiento Físico Animal , Ratas Endogámicas Lew , Animales , Masculino , Ratas , Condicionamiento Físico Animal/fisiología , Inestabilidad de la Articulación/fisiopatología , Osteoartritis de la Rodilla/fisiopatología , Modelos Animales de Enfermedad , Marcha/fisiología , Articulación de la Rodilla/fisiopatología , Glicosaminoglicanos/metabolismo , Osteoartritis/fisiopatología , Osteoartritis/metabolismo , Osteofito , Progresión de la Enfermedad
2.
Commun Biol ; 6(1): 749, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37468760

RESUMEN

Volumetric muscle loss (VML) results in permanent functional deficits and remains a substantial regenerative medicine challenge. A coordinated immune response is crucial for timely myofiber regeneration, however the immune response following VML has yet to be fully characterized. Here, we leveraged dimensionality reduction and pseudo-time analysis techniques to elucidate the cellular players underlying a functional or pathological outcome as a result of subcritical injury or critical VML in the murine quadriceps, respectively. We found that critical VML resulted in a sustained presence of M2-like and CD206hiLy6Chi 'hybrid' macrophages whereas subcritical defects resolved these populations. Notably, the retained M2-like macrophages from critical VML injuries presented with aberrant cytokine production which may contribute to fibrogenesis, as indicated by their co-localization with fibroadipogenic progenitors (FAPs) in areas of collagen deposition within the defect. Furthermore, several T cell subpopulations were significantly elevated in critical VML compared to subcritical injuries. These results demonstrate a dysregulated immune response in critical VML that is unable to fully resolve the chronic inflammatory state and transition to a pro-regenerative microenvironment within the first week after injury. These data provide important insights into potential therapeutic strategies which could reduce the immune cell burden and pro-fibrotic signaling characteristic of VML.


Asunto(s)
Músculo Esquelético , Enfermedades Musculares , Ratones , Animales , Músculo Esquelético/patología , Regeneración , Enfermedades Musculares/patología , Enfermedades Musculares/terapia , Medicina Regenerativa , Colágeno
3.
J Biomed Mater Res A ; 109(5): 695-712, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32608188

RESUMEN

Regeneration of skeletal muscle after volumetric injury is thought to be impaired by a dysregulated immune microenvironment that hinders endogenous repair mechanisms. Such defects result in fatty infiltration, tissue scarring, chronic inflammation, and debilitating functional deficits. Here, we evaluated the key cellular processes driving dysregulation in the injury niche through localized modulation of sphingosine-1-phosphate (S1P) receptor signaling. We employ dimensionality reduction and pseudotime analysis on single cell cytometry data to reveal heterogeneous immune cell subsets infiltrating preclinical muscle defects due to S1P receptor inhibition. We show that global knockout of S1P receptor 3 (S1PR3) is marked by an increase of muscle stem cells within injured tissue, a reduction in classically activated relative to alternatively activated macrophages, and increased bridging of regenerating myofibers across the defect. We found that local S1PR3 antagonism via nanofiber delivery of VPC01091 replicated key features of pseudotime immune cell recruitment dynamics and enhanced regeneration characteristic of global S1PR3 knockout. Our results indicate that local S1P receptor modulation may provide an effective immunotherapy for promoting a proreparative environment leading to improved regeneration following muscle injury.


Asunto(s)
Ciclopentanos/uso terapéutico , Inmunoterapia/métodos , Músculo Esquelético/lesiones , Regeneración/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/fisiología , Animales , Ciclopentanos/farmacología , Liberación de Fármacos , Citometría de Flujo , Leucopenia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía de Fuerza Atómica , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Células Mieloides/inmunología , Nanofibras , Tamaño de los Órganos , Músculo Cuádriceps/inmunología , Músculo Cuádriceps/lesiones , Músculo Cuádriceps/metabolismo , Músculo Cuádriceps/patología , Transducción de Señal/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/deficiencia , Receptores de Esfingosina-1-Fosfato/genética , Subgrupos de Linfocitos T/inmunología , Andamios del Tejido
4.
Sci Rep ; 9(1): 9551, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31266969

RESUMEN

Critical limb ischemia, the most severe form of peripheral artery disease, leads to extensive damage and alterations to skeletal muscle homeostasis. Although recent research has investigated the tissue-specific responses to ischemia, the role of the muscle stem cell in the regeneration of its niche components within skeletal muscle has been limited. To elucidate the regenerative mechanism of the muscle stem cell in response to ischemic insults, we explored cellular interactions between the vasculature, neural network, and muscle fiber within the muscle stem cell niche. Using a surgical murine hindlimb ischemia model, we first discovered a significant increase in subsynaptic nuclei and remodeling of the neuromuscular junction following ischemia-induced denervation. In addition, ischemic injury causes significant alterations to the myofiber through a muscle stem cell-mediated accumulation of total myonuclei and a concomitant decrease in myonuclear domain size, possibly to enhance the transcriptional and translation output and restore muscle mass. Results also revealed an accumulation of total mitochondrial content per myonucleus in ischemic myofibers to compensate for impaired mitochondrial function and high turnover rate. Taken together, the findings from this study suggest that the muscle stem cell plays a role in motor neuron reinnervation, myonuclear accretion, and mitochondrial biogenesis for skeletal muscle regeneration following ischemic injury.


Asunto(s)
Extremidades/irrigación sanguínea , Isquemia/metabolismo , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Unión Neuromuscular , Animales , Modelos Animales de Enfermedad , Isquemia/etiología , Ratones , Mitocondrias Musculares/metabolismo , Mioblastos/metabolismo , Regeneración
5.
Acta Biomater ; 94: 243-252, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31228633

RESUMEN

Skeletal muscle possesses efficient ability to regenerate upon minor injuries, but its capacity to regenerate is severely compromised with traumatic injuries and muscle-associated diseases. Recent evidence suggests that skeletal muscle regeneration can be enhanced by transplantation of muscle satellite cells (MuSCs) or treatment with pro-myogenic factors, such as Wingless-type MMTV Integrated 7a (Wnt7a) protein. Although direct intramuscular injection is the simplest method to deliver MuSCs and Wnt7a for regenerative therapy, direct injections are not viable in many clinical cases where structural integrity is severely compromised. To address this challenge, we evaluated the feasibility of co-delivering pro-myogenic factors, such as Wnt7a, and MuSCs using a synthetic poly(ethylene glycol) (PEG)-based hydrogel to the affected skeletal muscles. The Wnt7a release rate can be controlled by modulating the polymer density of the hydrogel, and this release rate can be further accelerated through the proteolytic degradation of the hydrogel. Treating cryo-injured tibialis anterior (TA) muscles with Wnt7a-loaded hydrogels resulted in an improved regenerative response by day 14, measured by increased muscle fiber cross-sectional area, bulk TA mass, and the number of Pax7+ MuSCs at the injury site, compared to the TA muscles treated with Wnt7a-free hydrogels. Co-delivery of Wnt7a and primary MuSCs using the synthetic hydrogel to the cryo-injured TA muscles significantly increased cellular migration during the engraftment process. This work provides a synthetic biomaterial platform for advancing treatment strategies of skeletal muscle conditions where direct intramuscular injection may be challenging. Finally, the current outcomes establish an important foundation for future applications in treating severe muscle trauma and diseases, where the endogenous repair capacity is critically impaired. STATEMENT OF SIGNIFICANCE: Skeletal muscle injuries and diseases cause debilitating health consequences, including disability and diminished quality of life. Treatment using protein and stem cell-based therapeutics may help regenerate the affected muscles, but direct intramuscular injection may not be feasible in severe muscle injuries due to the gravely damaged tissue structure. In chronic muscle diseases, such as Duchenne muscular dystrophy, local treatment of the diaphragm, a muscle critical for respiration, may be necessary but direct injection is difficult due to its thin dimensions. To address this challenge, this work presents a synthetic and bioactive muscle "patch" that enables concurrent administration of proteins and muscle stem cells for accelerated muscle healing.


Asunto(s)
Hidrogeles/química , Músculo Esquelético/fisiología , Regeneración/fisiología , Proteínas Wnt/genética , Animales , Línea Celular , Movimiento Celular , Proliferación Celular , Cruzamientos Genéticos , Femenino , Proteínas Fluorescentes Verdes/genética , Humanos , Masculino , Maleimidas/química , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/fisiología , Polietilenglicoles/química , Proteínas Recombinantes/genética , Células Satélite del Músculo Esquelético/fisiología
6.
Tissue Eng Part C Methods ; 25(2): 59-70, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30648479

RESUMEN

IMPACT STATEMENT: The goal of this study was to determine the threshold for a critically sized, nonhealing muscle defect by characterizing key components in the balance between fibrosis and regeneration as a function of injury size in the mouse quadriceps. There is currently limited understanding of what leads to a critically sized muscle defect and which muscle regenerative components are functionally impaired. With the substantial increase in preclinical VML models as testbeds for tissue engineering therapeutics, defining the critical threshold for VML injuries will be instrumental in characterizing therapeutic efficacy and potential for subsequent translation.


Asunto(s)
Enfermedades Musculares/patología , Enfermedades Musculares/terapia , Miofibrillas/fisiología , Unión Neuromuscular/citología , Músculo Cuádriceps/citología , Músculo Cuádriceps/lesiones , Ingeniería de Tejidos , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Músculo Cuádriceps/fisiología , Andamios del Tejido , Cicatrización de Heridas
7.
Proc Natl Acad Sci U S A ; 115(42): E9944-E9952, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30275336

RESUMEN

Dysfunctional endothelium causes more disease than any other cell type. Systemically administered RNA delivery to nonliver tissues remains challenging, in large part because there is no high-throughput method to identify nanoparticles that deliver functional mRNA to cells in vivo. Here we report a system capable of simultaneously quantifying how >100 lipid nanoparticles (LNPs) deliver mRNA that is translated into functional protein. Using this system (named FIND), we measured how >250 LNPs delivered mRNA to multiple cell types in vivo and identified 7C2 and 7C3, two LNPs that efficiently deliver siRNA, single-guide RNA (sgRNA), and mRNA to endothelial cells. The 7C3 delivered Cas9 mRNA and sgRNA to splenic endothelial cells as efficiently as hepatocytes, distinguishing it from LNPs that deliver Cas9 mRNA and sgRNA to hepatocytes more than other cell types. These data demonstrate that FIND can identify nanoparticles with novel tropisms in vivo.


Asunto(s)
Sistemas CRISPR-Cas , Células Endoteliales/metabolismo , Edición Génica , Técnicas de Transferencia de Gen , Lípidos/química , Nanopartículas/administración & dosificación , ARN Guía de Kinetoplastida/genética , ARN Mensajero/genética , Animales , Células Cultivadas , Células Endoteliales/citología , Células HEK293 , Hepatocitos/citología , Hepatocitos/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , ARN Guía de Kinetoplastida/química , ARN Mensajero/química
8.
Sci Adv ; 4(8): eaar4008, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30116776

RESUMEN

Muscle satellite cells (MuSCs) play a central role in muscle regeneration, but their quantity and function decline with comorbidity of trauma, aging, and muscle diseases. Although transplantation of MuSCs in traumatically injured muscle in the comorbid context of aging or pathology is a strategy to boost muscle regeneration, an effective cell delivery strategy in these contexts has not been developed. We engineered a synthetic hydrogel-based matrix with optimal mechanical, cell-adhesive, and protease-degradable properties that promotes MuSC survival, proliferation, and differentiation. Furthermore, we establish a biomaterial-mediated cell delivery strategy for treating muscle trauma, where intramuscular injections may not be applicable. Delivery of MuSCs in the engineered matrix significantly improved in vivo cell survival, proliferation, and engraftment in nonirradiated and immunocompetent muscles of aged and dystrophic mice compared to collagen gels and cell-only controls. This platform may be suitable for treating craniofacial and limb muscle trauma, as well as postoperative wounds of elderly and dystrophic patients.


Asunto(s)
Envejecimiento , Hidrogeles/química , Músculo Esquelético/citología , Distrofias Musculares/terapia , Células Satélite del Músculo Esquelético/trasplante , Heridas y Lesiones/terapia , Animales , Diferenciación Celular , Comorbilidad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Regeneración , Células Satélite del Músculo Esquelético/citología , Ingeniería de Tejidos , Heridas y Lesiones/metabolismo , Heridas y Lesiones/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...