Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dev Biol ; 504: 98-112, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37778717

RESUMEN

Severe muscle injury causes distress and difficulty in humans. Studying the high regenerative ability of the axolotls may provide hints for the development of an effective treatment for severe injuries to muscle tissue. Here, we examined the regenerative process in response to a muscle injury in axolotls. We found that axolotls are capable of complete regeneration in response to a partial muscle resection called volumetric muscle loss (VML), which mammals cannot perfectly regenerate. We investigated the mechanisms underlying this high regenerative capacity in response to VML, focusing on the migration of muscle satellite cells and the extracellular matrix (ECM) formed during VML injury. Axolotls form tenascin-C (TN-C)-enriched ECM after VML injury. This TN-C-enriched ECM promotes the satellite cell migration. We confirmed the importance of TN-C in successful axolotl muscle regeneration by creating TN-C mutant animals. Our results suggest that the maintenance of a TN-C-enriched ECM environment after muscle injury promotes the release of muscle satellite cells and supports eventually high muscle regenerative capacity. In the future, better muscle regeneration may be achieved in mammals through the maintenance of TN-C expression.


Asunto(s)
Ambystoma mexicanum , Tenascina , Animales , Humanos , Tenascina/genética , Tenascina/metabolismo , Ambystoma mexicanum/metabolismo , Matriz Extracelular/metabolismo , Músculos/metabolismo , Mamíferos/metabolismo , Músculo Esquelético/metabolismo
2.
Semin Cell Dev Biol ; 119: 61-69, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33994095

RESUMEN

Volumetric muscle loss (VML) VML is defined as the loss of a critical mass of skeletal muscle that overwhelms the muscle's natural healing mechanisms, leaving patients with permanent functional deficits and deformity. The treatment of these defects is complex, as skeletal muscle is a composite structure that relies closely on the action of supporting tissues such as tendons, vasculature, nerves, and bone. The gold standard of treatment for VML injuries, an autologous muscle flap transfer, suffers from many shortcomings but nevertheless remains the best clinically available avenue to restore function. This review will consider the use of composite tissue engineered constructs, with multiple components that act together to replicate the function of an intact muscle, as an alternative to autologous muscle flaps. We will discuss recent advances in the field of tissue engineering that enable skeletal muscle constructs to more closely reproduce the functionality of an autologous muscle flap by incorporating vasculature, promoting innervation, and reconstructing the muscle-tendon boundary. Additionally, our understanding of the cellular composition of skeletal muscle has evolved to recognize the importance of a diverse variety of cell types in muscle regeneration, including fibro/adipogenic progenitors and immune cells like macrophages and regulatory T cells. We will address recent advances in our understanding of how these cell types interact with, and can be incorporated into, implanted tissue engineered constructs.


Asunto(s)
Músculo Esquelético/fisiología , Enfermedades Musculares/fisiopatología , Enfermedades Musculares/terapia , Ingeniería de Tejidos/métodos , Animales , Humanos , Ratones
3.
Am J Physiol Cell Physiol ; 324(6): C1341-C1352, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37184231

RESUMEN

Volumetric muscle loss (VML) causes irrecoverable loss of muscle mass and strength and results in permanent disability. VML injury shows extensive fibrosis, which impedes functional tissue regeneration. Our lab has created a biosponge scaffold composed of extracellular matrix (ECM) proteins (i.e., biosponge) that can enhance muscle regeneration and function following VML. In this work, a potent small molecule inhibitor of alpha v-subunit containing integrins known as IDL-2965 was incorporated into the biosponges for localized suppression of fibrosis post-VML. Our results demonstrate that local delivery of IDL-2965 via the biosponges attenuated the deposition of fibrotic tissue preceded by a downregulation of profibrotic genes in VML-injured muscles. The reduction in fibrotic tissue had no detrimental effects on muscle mass, function, size, or vascularity. Overall, these findings suggest that the codelivery of biosponges and IDL-2965 is a safe and effective strategy for the mitigation of fibrotic tissue deposition in VML-injured muscles.


Asunto(s)
Músculo Esquelético , Enfermedades Musculares , Humanos , Músculo Esquelético/metabolismo , Enfermedades Musculares/patología , Cicatrización de Heridas , Proteínas de la Matriz Extracelular/metabolismo , Fibrosis
4.
J Surg Res ; 291: 683-690, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37562230

RESUMEN

INTRODUCTION: The clinical characterization of the functional status of active wounds in terms of their driving cellular and molecular biology remains a considerable challenge that currently requires excision via a tissue biopsy. In this pilot study, we use convolutional Siamese neural network (SNN) architecture to predict the functional state of a wound using digital photographs of wounds in a canine model of volumetric muscle loss (VML). METHODS: Digital images of VML injuries and tissue biopsies were obtained in a standardized fashion from an established canine model of VML. Gene expression profiles for each biopsy site were obtained using RNA sequencing. These profiles were converted to functional profiles by a manual review of validated gene ontology databases in which we determined a hierarchical representation of gene functions based on functional specificity. An SNN was trained to regress functional profile expression values, informed by an image segment showing the surface of a small tissue biopsy. RESULTS: The SNN was able to predict the functional expression of a range of functions based with error ranging from ∼5% to ∼30%, with functions that are most closely associated with the early state of wound healing to be those best-predicted. CONCLUSIONS: These initial results suggest promise for further research regarding this novel use of machine learning regression on medical images. The regression of functional profiles, as opposed to specific genes, both addresses the challenge of genetic redundancy and gives a deeper insight into the mechanistic configuration of a region of tissue in wounds.


Asunto(s)
Inteligencia Artificial , Cicatrización de Heridas , Animales , Perros , Proyectos Piloto , Redes Neurales de la Computación , Biopsia , Músculo Esquelético/patología
5.
J Surg Res ; 270: 547-554, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34826690

RESUMEN

BACKGROUND: The clinical characterization of the biological status of complex wounds remains a considerable challenge. Digital photography provides a non-invasive means of obtaining wound information and is currently employed to assess wounds qualitatively. Advances in machine learning (ML) image processing provide a means of identifying "hidden" features in pictures. This pilot study trains a convolutional neural network (CNN) to predict gene expression based on digital photographs of wounds in a canine model of volumetric muscle loss (VML). MATERIALS AND METHODS: Images of volumetric muscle loss injuries and tissue biopsies were obtained in a canine model of VML. A CNN was trained to regress gene expression values as a function of the extracted image segment (color and spatial distribution). Performance of the CNN was assessed in a held-back test set of images using Mean Absolute Percentage Error (MAPE). RESULTS: The CNN was able to predict the gene expression of certain genes based on digital images, with a MAPE ranging from ∼10% to ∼30%, indicating the presence and identification of distinct, and identifiable patterns in gene expression throughout the wound. CONCLUSIONS: These initial results suggest promise for further research regarding this novel use of ML regression on medical images. Specifically, the use of CNNs to determine the mechanistic biological state of a VML wound could aid both the design of future mechanistic interventions and the design of trials to test those therapies. Future work will expand the CNN training and/or test set, with potential expansion to predicting functional gene modules.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Redes Neurales de la Computación , Animales , Biopsia , Perros , Expresión Génica , Procesamiento de Imagen Asistido por Computador/métodos , Proyectos Piloto
6.
Mol Ther ; 29(1): 121-131, 2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-32966776

RESUMEN

Volumetric muscle loss injury is a common health problem with long-term disabilities. One common treatment is using muscle flaps from donor site, which has limited potentials due to donor site availability and morbidity. Although several stem cell therapies have been evaluated so far, most suffer from limited availability, immune incompatibility, or differentiation potential. Therefore, induced pluripotent stem cells (iPSCs) have a great promise for this purpose due to their unique differentiation, self-renewal, and immunocompatibility. Current study was designed to determine therapeutic potential of human iPSCs (hiPSCs) in a mouse model of volumetric muscle loss. Muscles were subjected to excision to generate 30%-40% muscle loss. Next, hiPSCs were differentiated toward skeletal myogenic progenitors and used with fibrin hydrogel to reconstruct the lost muscle. Histologic evaluation of the treated muscles indicated abundant engraftment of donor-derived mature fibers expressing human markers. Donor-derived fibers were also positive for the presence of neuromuscular junction (NMJ), indicating their proper innervation. Evaluation of the engrafted region indicated the presence of donor-derived satellite cells expressing human markers and Pax7. Finally, in situ muscle function analysis demonstrated significant improvement of the muscle contractility in muscles treated with hiPSCs. These results therefore provide key evidence for the therapeutic potential of human iPSCs in volumetric muscle loss injuries.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedades Musculares/patología , Enfermedades Musculares/terapia , Trasplante de Células Madre , Animales , Atrofia , Modelos Animales de Enfermedad , Supervivencia de Injerto , Ratones , Músculo Esquelético/patología , Enfermedades Musculares/etiología , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/métodos , Resultado del Tratamiento
7.
Connect Tissue Res ; 62(1): 83-98, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32552156

RESUMEN

AIM OF THE REVIEW: This review aims to describe the current state of regenerative rehabilitation of severe military extremity injuries, and promising new therapies on the horizon. DISCUSSION: The nature of warfare is rapidly shifting with information operations, autonomous weapons, and the threat of full-scale peer adversary conflicts threatening to create contested environments with delayed medical evacuation to definitive care. More destructive weapons will lead to more devastating injuries, creating new challenges for limb repair and restoration. Current paradigms of delayed rehabilitation following initial stabilization, damage control surgery, and prolonged antibiotic therapy will need to shift. Advances in regenerative medicine technologies offer the possibility of treatment along the continuum of care. Regenerative rehabilitation will begin at the point of injury and require a holistic, organ-systems approach. CONCLUSIONS: Both technological improvements and a rapidly advancing understanding of injury pathophysiology will contribute to improved limb-salvage outcomes, and shift the calculus away from early limb amputation.


Asunto(s)
Personal Militar , Amputación Quirúrgica , Extremidades , Humanos , Recuperación del Miembro , Estudios Retrospectivos
8.
Oral Dis ; 27(6): 1580-1589, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33031620

RESUMEN

OBJECTIVES: To use tissue engineering muscle repair (TEMR) for regenerating the lingual musculature of hemiglossectomized rats using neonatal myoblasts (NM) on porcine acellular urinary bladder matrix (AUBM). MATERIAL AND METHODS: The study used 80 male rats. A volumetric muscle loss (VML) injury was created on the left side of the tongue. The rats were randomized into four groups: Group 1 (AUBM + myoblasts); Group 2 (AUBM); Group 3 (myoblasts); and Group 4 (control). NM were obtained from neonatal rats. The animals were weighed on day 0 and just before euthanasia. Five rats in each group were euthanized at days 2, 14, 28, and 42; the tongues were prepared for morphometric analysis, postoperative left hemitongue weight, and immunohistochemical analysis (desmin, CD-31, and anti-neurofilament). RESULTS: The weight gain from greatest to least was as follows: AUBM + myoblasts > myoblasts > AUBM > control. The tongue dorsum occupied by VML, and difference in mg between control side and intervened side from least to great was as follows: AUBM + myoblasts < myoblasts < AUBM < control. The order from highest to lowest antibody positivity was as follows: AUBM + myoblasts > myoblasts > AUBM > control. CONCLUSION: The use of porcine AUBM and NM for the regeneration of lingual musculature was found to be an effective TEMR treatment for repairing tongue VML injury.


Asunto(s)
Ingeniería de Tejidos , Vejiga Urinaria , Animales , Masculino , Mioblastos , Ratas , Regeneración , Porcinos , Lengua , Vejiga Urinaria/cirugía
9.
Int J Mol Sci ; 22(13)2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206557

RESUMEN

Different types of scaffolds are used to reconstruct muscle volume loss injuries. In this experimental study, we correlated ultrasound observations with histological findings in a muscle volume loss injury reconstructed with autologous adipose tissue. The outcome is compared with decellularized and porous matrix implants. Autologous adipose tissue, decellularized matrix, and a porous collagen matrix were implanted in volumetric muscle loss (VML) injuries generated on the anterior tibial muscles of Wistar rats. Sixty days after implantation, ultrasound findings were compared with histological and histomorphometric analysis. The muscles with an autologous adipose tissue implant exhibited an ultrasound pattern that was quite similar to that of the regenerative control muscles. From a histological point of view, the defects had been occupied by newly formed muscle tissue with certain structural abnormalities that would explain the differences between the ultrasound patterns of the normal control muscles and the regenerated ones. While the decellularized muscle matrix implant resulted in fibrosis and an inflammatory response, the porous collagen matrix implant was replaced by regenerative muscle fibers with neurogenic atrophy and fibrosis. In both cases, the ultrasound images reflected echogenic, echotextural, and vascular changes compatible with the histological findings of failed muscle regeneration. The ultrasound analysis confirmed the histological findings observed in the VML injuries reconstructed by autologous adipose tissue implantation. Ultrasound can be a useful tool for evaluating the structure of muscles reconstructed through tissue engineering.


Asunto(s)
Tejido Adiposo/cirugía , Enfermedades Musculares/diagnóstico por imagen , Enfermedades Musculares/terapia , Procedimientos de Cirugía Plástica , Animales , Biopsia , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Enfermedades Musculares/etiología , Enfermedades Musculares/patología , Tamaño de los Órganos , Ratas , Procedimientos de Cirugía Plástica/métodos , Regeneración , Ingeniería de Tejidos , Resultado del Tratamiento , Ultrasonografía
10.
Int J Mol Sci ; 22(2)2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33419056

RESUMEN

Volumetric muscle loss injuries overwhelm the endogenous regenerative capacity of skeletal muscle, and the associated oxidative damage can delay regeneration and prolong recovery. This study aimed to investigate the effect of silicon-ions on C2C12 skeletal muscle cells under normal and excessive oxidative stress conditions to gain insights into its role on myogenesis during the early stages of muscle regeneration. In vitro studies indicated that 0.1 mM Si-ions into cell culture media significantly increased cell viability, proliferation, migration, and myotube formation compared to control. Additionally, MyoG, MyoD, Neurturin, and GABA expression were significantly increased with addition of 0.1, 0.5, and 1.0 mM of Si-ion for 1 and 5 days of C2C12 myoblast differentiation. Furthermore, 0.1-2.0 mM Si-ions attenuated the toxic effects of H2O2 within 24 h resulting in increased cell viability and differentiation. Addition of 1.0 mM of Si-ions significantly aid cell recovery and protected from the toxic effect of 0.4 mM H2O2 on cell migration. These results suggest that ionic silicon may have a potential effect in unfavorable situations where reactive oxygen species is predominant affecting cell viability, proliferation, migration, and differentiation. Furthermore, this study provides a guide for designing Si-containing biomaterials with desirable Si-ion release for skeletal muscle regeneration.


Asunto(s)
Músculo Esquelético/fisiología , Mioblastos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Regeneración/efectos de los fármacos , Silicio/farmacología , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Regulación de la Expresión Génica/efectos de los fármacos , Iones/química , Iones/farmacología , Ratones , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/genética , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/metabolismo , Mioblastos/fisiología , Especies Reactivas de Oxígeno/metabolismo , Regeneración/genética
11.
Molecules ; 26(3)2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33572728

RESUMEN

Volumetric Muscle Loss (VML) is associated with muscle loss function and often untreated and considered part of the natural sequelae of trauma. Various types of biomaterials with different physical and properties have been developed to treat VML. However, much work remains yet to be done before the scaffolds can pass from the bench to the bedside. The present review aims to provide a comprehensive summary of the latest developments in the construction and application of natural polymers-based tissue scaffolding for volumetric muscle injury. Here, the tissue engineering approaches for treating volumetric muscle loss injury are highlighted and recent advances in cell-based therapies using various sources of stem cells are elaborated in detail. An overview of different strategies of tissue scaffolding and their efficacy on skeletal muscle cells regeneration and migration are presented. Furthermore, the present paper discusses a wide range of natural polymers with a special focus on proteins and polysaccharides that are major components of the extracellular matrices. The natural polymers are biologically active and excellently promote cell adhesion and growth. These bio-characteristics justify natural polymers as one of the most attractive options for developing scaffolds for muscle cell regeneration.


Asunto(s)
Músculo Esquelético/efectos de los fármacos , Enfermedades Musculares/tratamiento farmacológico , Regeneración/efectos de los fármacos , Andamios del Tejido/química , Materiales Biocompatibles/uso terapéutico , Matriz Extracelular/efectos de los fármacos , Humanos , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Enfermedades Musculares/patología , Polímeros/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos
12.
Connect Tissue Res ; 61(2): 216-228, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31899969

RESUMEN

Purpose: This report explores the overlooked potential of bioprinting to automate biomanufacturing of simple tissue structures, such as the uniform deposition of (mono)layers of progenitor cells on sheetlike decellularized extracellular matrices (dECM). In this scenario, dECM serves as a biodegradable celldelivery matrix to provide enhanced regenerative microenvironments for tissue repair. The Tissue-Engineered Muscle Repair (TEMR) technology-where muscle progenitor cells are seeded onto a porcine bladder acellular matrix (BAM), serves as a representative testbed for bioprinting applications. Previous work demonstrated that TEMR implantation improved functional outcomes following VML injury in biologically relevant rodent models.Materials and Methods: In the described bioprinting system, a cell-laden hydrogel bioink is used to deposit high cell densities (1.4 × 105-3.5 × 105 cells/cm2), onto both sides of the bladder acellular matrix as proof-of-concept.Results: These bioprinting methods achieve a reproducible and homogeneous distribution of cells, on both sides of the BAM scaffold, after just 24hrs, with cell viability as high as 98%. These preliminary results suggest bioprinting allows for improved dual-sided cell coverage compared to manual-seeding.Conclusions: Bioprinting can enable automated fabrication of TEMR constructs with high fidelity and scalability, while reducing biomanufacturing costs and timelines. Such bioprinting applications are underappreciated, yet critical, to expand the overall biomanufacturing paradigm for tissue engineered medical products. In addition, biofabrication of sheet-like implantable constructs, with cells deposited on both sides, is a process that is both scaffold and cell-type agnostic, and furthermore, is amenable to many geometries, and thus, additional tissue engineering applications beyond skeletal muscle.


Asunto(s)
Implantes Absorbibles , Bioimpresión , Músculo Esquelético , Impresión Tridimensional , Regeneración , Ingeniería de Tejidos , Andamios del Tejido/química , Humanos , Músculo Esquelético/lesiones , Músculo Esquelético/fisiología
13.
J Cell Mol Med ; 23(6): 3784-3794, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30933431

RESUMEN

Both volumetric muscle loss (VML) and muscle degenerative diseases lead to an important decrease in skeletal muscle mass, condition that nowadays lacks an optimal treatment. This issue has driven towards an increasing interest in new strategies in tissue engineering, an emerging field that can offer very promising approaches. In addition, the discovery of induced pluripotent stem cells (iPSCs) has completely revolutionized the actual view of personalized medicine, and their utilization in skeletal muscle tissue engineering could, undoubtedly, add myriad benefits. In this review, we want to provide a general vision of the basic aspects to consider when engineering skeletal muscle tissue using iPSCs. Specifically, we will focus on the three main pillars of tissue engineering: the scaffold designing, the selection of the ideal cell source and the addition of factors that can enhance the resemblance with the native tissue.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Músculo Esquelético/citología , Regeneración/fisiología , Ingeniería de Tejidos/métodos , Animales , Técnicas de Cultivo de Célula/métodos , Humanos , Células Madre Pluripotentes Inducidas/citología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Medicina de Precisión/métodos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Andamios del Tejido
14.
Exp Physiol ; 109(6): 889-891, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38493319
16.
Int J Mol Sci ; 19(8)2018 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-30110909

RESUMEN

Several acquired or congenital pathological conditions can affect skeletal muscle leading to volumetric muscle loss (VML), i.e., an irreversible loss of muscle mass and function. Decellularized tissues are natural scaffolds derived from tissues or organs, in which the cellular and nuclear contents are eliminated, but the tridimensional (3D) structure and composition of the extracellular matrix (ECM) are preserved. Such scaffolds retain biological activity, are biocompatible and do not show immune rejection upon allogeneic or xenogeneic transplantation. An increase number of reports suggest that decellularized tissues/organs are promising candidates for clinical application in patients affected by VML. Here we explore the different strategies used to generate decellularized matrix and their therapeutic outcome when applied to treat VML conditions, both in patients and in animal models. The wide variety of VML models, source of tissue and methods of decellularization have led to discrepant results. Our review study evaluates the biological and clinical significance of reported studies, with the final aim to clarify the main aspects that should be taken into consideration for the future application of decellularized tissues in the treatment of VML conditions.


Asunto(s)
Matriz Extracelular/trasplante , Músculos/fisiología , Regeneración , Aloinjertos , Animales , Xenoinjertos , Humanos
17.
BMC Musculoskelet Disord ; 18(1): 253, 2017 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-28606129

RESUMEN

BACKGROUND: Heightened local inflammation due to muscle trauma or disease is associated with impaired bone regeneration. METHODS: We hypothesized that FK506, an FDA approved immunomodulatory compound with neurotrophic and osteogenic effects, will rescue the early phase of fracture healing which is impaired by concomitant muscle trauma in male (~4 months old) Lewis rats. FK506 (1 mg/kg; i.p.) or saline was administered systemically for 14 days after an endogenously healing tibia osteotomy was created and fixed with an intermedullary pin, and the overlying tibialis anterior (TA) muscle was either left uninjured or incurred volumetric muscle loss injury (6 mm full thickness biopsy from middle third of the muscle). RESULTS: The salient observations of this study were that 1) concomitant TA muscle trauma impaired recovery of tibia mechanical properties 28 days post-injury, 2) FK506 administration rescued the recovery of tibia mechanical properties in the presence of concomitant TA muscle trauma but did not augment mechanical recovery of an isolated osteotomy (no muscle trauma), 3) T lymphocytes and macrophage presence within the traumatized musculature were heightened by trauma and attenuated by FK506 3 days post-injury, and 4) T lymphocyte but not macrophage presence within the fracture callus were attenuated by FK506 at 14 days post-injury. FK506 did not improve TA muscle isometric torque production CONCLUSION: Collectively, these findings support the administration of FK506 to ameliorate healing of fractures with severe muscle trauma comorbidity. The results suggest one potential mechanism of action is a reduction in local T lymphocytes within the injured musculoskeletal tissue, though other mechanisms to include direct osteogenic effects of FK506 require further investigation.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Curación de Fractura/efectos de los fármacos , Inmunosupresores/uso terapéutico , Músculo Esquelético/lesiones , Tacrolimus/uso terapéutico , Fracturas de la Tibia/tratamiento farmacológico , Inmunidad Adaptativa/efectos de los fármacos , Animales , Biopsia , Clavos Ortopédicos , Callo Óseo/efectos de los fármacos , Callo Óseo/inmunología , Callo Óseo/patología , Modelos Animales de Enfermedad , Fijación Intramedular de Fracturas/instrumentación , Fijación Intramedular de Fracturas/métodos , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunosupresores/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Enfermedades Musculares/complicaciones , Enfermedades Musculares/tratamiento farmacológico , Enfermedades Musculares/inmunología , Enfermedades Musculares/patología , Osteotomía , Ratas , Ratas Endogámicas Lew , Traumatismos de los Tejidos Blandos/complicaciones , Traumatismos de los Tejidos Blandos/tratamiento farmacológico , Traumatismos de los Tejidos Blandos/inmunología , Traumatismos de los Tejidos Blandos/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Tacrolimus/farmacología , Fracturas de la Tibia/fisiopatología , Fracturas de la Tibia/cirugía , Torque
18.
Annu Rev Biomed Eng ; 17: 217-42, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26643021

RESUMEN

Although skeletal muscle is one of the most regenerative organs in our body, various genetic defects, alterations in extrinsic signaling, or substantial tissue damage can impair muscle function and the capacity for self-repair. The diversity and complexity of muscle disorders have attracted much interest from both cell biologists and, more recently, bioengineers, leading to concentrated efforts to better understand muscle pathology and develop more efficient therapies. This review describes the biological underpinnings of muscle development, repair, and disease, and discusses recent bioengineering efforts to design and control myomimetic environments, both to study muscle biology and function and to aid in the development of new drug, cell, and gene therapies for muscle disorders. The synergy between engineering-aided biological discovery and biology-inspired engineering solutions will be the path forward for translating laboratory results into clinical practice.


Asunto(s)
Modelos Biológicos , Músculo Esquelético/lesiones , Animales , Ingeniería Biomédica , Biomimética , Humanos , Desarrollo de Músculos , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , Enfermedades Musculares/terapia , Regeneración , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología
19.
Tissue Eng Part A ; 30(5-6): 181-191, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37658842

RESUMEN

There are currently no surgical procedures that effectively address the treatment of volumetric muscle loss (VML) injuries that has motivated the development of implantable scaffolding. In this study, the effectiveness of an allogenic scaffold fabricated using fibers built from the extracellular matrix (ECM) collected from muscle fibroblast cells during growth in culture was explored using a hindlimb VML injury (tibialis anterior muscle) in a rat model. Recovery outcomes (8 weeks) were explored in comparison with unrepaired controls as well previously examined allogenic scaffolds prepared from decellularized skeletal muscle (DSM) tissue (n = 9/sample group). At 8-week follow-up, we found that the repair of VML injuries using ECM fiber scaffolds in combination with an autogenic mince muscle (MM) paste significantly improved the recovery of peak contractile torque (79% ± 13% of uninjured contralateral muscle) when compared with unrepaired VML controls (57% ± 13%). Similar significant improvements were measured for muscle mass restoration (93% ± 10%) in response to ECM fiber+MM repair when compared with unrepaired VML controls (73% ± 13%). Of note, mass and contractile strength recovery outcomes for ECM fiber scaffolds were not significantly different from DSM+MM repair controls. These in vivo findings support the further exploration of cell-derived ECM fiber scaffolds as a promising strategy for the repair of VML injury with recovery outcomes that compare favorably with current tissue-sourced ECM scaffolds. Furthermore, although the therapeutic potential of ECM fibers as a treatment strategy for muscle injury was explored in this study, they could be adapted for high-throughput fabrication methods developed and routinely used by the textile industry to create a broad range of woven implants (e.g., hernia meshes) for even greater clinical impact.


Asunto(s)
Músculo Esquelético , Enfermedades Musculares , Ratas , Animales , Músculo Esquelético/lesiones , Matriz Extracelular , Andamios del Tejido , Fibras Musculares Esqueléticas , Regeneración
20.
Matrix Biol ; 127: 38-47, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38325441

RESUMEN

Volumetric muscle loss (VML) represents a clinical challenge due to the limited regenerative capacity of skeletal muscle. Most often, it results in scar tissue formation and loss of function, which cannot be prevented by current therapies. Decellularized extracellular matrix (DEM) has emerged as a native biomaterial for the enhancement of tissue repair. Here, we report the generation and characterization of hydrogels derived from DEM prepared from WT or thrombospondin (TSP)-2 null muscle tissue. TSP2-null hydrogels, when compared to WT, displayed altered architecture, protein composition, and biomechanical properties and allowed enhanced invasion of C2C12 myocytes and chord formation by endothelial cells. They also displayed enhanced cell invasion, innervation, and angiogenesis following subcutaneous implantation. To evaluate their regenerative capacity, WT or TSP2 null hydrogels were used to treat VML injury to tibialis anterior muscles and the latter induced greater recruitment of repair cells, innervation, and blood vessel formation and reduced inflammation. Taken together, these observations indicate that TSP2-null hydrogels enhance angiogenesis and promote muscle repair in a VML model.


Asunto(s)
Células Endoteliales , Hidrogeles , Hidrogeles/farmacología , Angiogénesis , Matriz Extracelular/metabolismo , Músculo Esquelético , Neurogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA