Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Acta Biomater ; 154: 108-122, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36272687

RESUMEN

Biological and mechanical cues are both vital for biomaterial aided tendon repair and regeneration. Here, we fabricated mechanically tendon-like (0 s UV) QHM polyurethane scaffolds (Q: Quadrol, H: Hexamethylene diisocyanate; M: Methacrylic anhydride) and immobilized them with Growth and differentiation factor-7 (GDF-7) to produce mechanically strong and tenogenic scaffolds. In this study, we assessed QHM polymer cytocompatibility, amenability to fibrin-coating, immobilization and persistence of GDF-7, and capability to support GDF-7-mediated tendon differentiation in vitro as well as in vivo in mouse subcutaneous and acute rat rotator cuff tendon resection models. Cytocompatibility studies showed that QHM facilitated cell attachment, proliferation, and viability. Fibrin-coating and GDF-7 retention studies showed that mechanically tendon-like 0 s UV QHM polymer could be immobilized with GDF-7 and retained the growth factor (GF) for at least 1-week ex vivo. In vitro differentiation studies showed that GDF-7 mediated bone marrow-derived human mesenchymal stem cell (hMSC) tendon-like differentiation on 0 s UV QHM. Subcutaneous implantation of GDF-7-immobilized, fibrin-coated, QHM polymer in mice for 2 weeks demonstrated de novo formation of tendon-like tissue while implantation of GDF-7-immobilized, fibrin-coated, QHM polymer in a rat acute rotator cuff resection injury model indicated tendon-like tissue formation in situ and the absence of heterotopic ossification. Together, our work demonstrates a promising synthetic scaffold with human tendon-like biomechanical attributes as well as immobilized tenogenic GDF-7 for tendon repair and regeneration. STATEMENT OF SIGNIFICANCE: Biological activity and mechanical robustness are key features required for tendon-promoting biomaterials. While synthetic biomaterials can be mechanically robust, they often lack bioactivity. To biologically augment synthetic biomaterials, numerous drug and GF delivery strategies exist but the large tissue space within the shoulder is constantly flushed with saline during arthroscopic surgery, hindering efficacious controlled release of therapeutic molecules. Here, we coated QHM polymer (which exhibits human tendon-to-bone-like biomechanical attributes) with fibrin for GF binding. Unlike conventional drug delivery strategies, our approach utilizes immobilized GFs as opposed to released GFs for sustained, localized tissue regeneration. Our data demonstrated that GF immobilization can be broadly applied to synthetic biomaterials for enhancing bioactivity, and GDF-7-immobilized QHM exhibit high clinical translational potential for tendon repair.


Asunto(s)
Polímeros , Lesiones del Manguito de los Rotadores , Ratas , Ratones , Humanos , Animales , Poliuretanos/farmacología , Anhídridos , Tendones , Diferenciación Celular , Materiales Biocompatibles , Lesiones del Manguito de los Rotadores/cirugía , Andamios del Tejido/química
2.
Adv Funct Mater ; 28(20)2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29785178

RESUMEN

Critical considerations in engineering biomaterials for rotator cuff repair include bone-tendon-like mechanical properties to support physiological loading and biophysicochemical attributes that stabilize the repair site over the long-term. In this study, UV-crosslinkable polyurethane based on quadrol (Q), hexamethylene diisocyante (H), and methacrylic anhydride (M; QHM polymers), which are free of solvent, catalyst, and photoinitiator, is developed. Mechanical characterization studies demonstrate that QHM polymers possesses phototunable bone- and tendon-like tensile and compressive properties (12-74 MPa tensile strength, 0.6-2.7 GPa tensile modulus, 58-121 MPa compressive strength, and 1.5-3.0 GPa compressive modulus), including the capability to withstand 10 000 cycles of physiological tensile loading and reduce stress concentrations via stiffness gradients. Biophysicochemical studies demonstrate that QHM polymers have clinically favorable attributes vital to rotator cuff repair stability, including slow degradation profiles (5-30% mass loss after 8 weeks) with little-to-no cytotoxicity in vitro, exceptional suture retention ex vivo (2.79-3.56-fold less suture migration relative to a clinically available graft), and competent tensile properties (similar ultimate load but higher normalized tensile stiffness relative to a clinically available graft) as well as good biocompatibility for augmenting rat supraspinatus tendon repair in vivo. This work demonstrates functionally graded, bone-tendon-like biomaterials for interfacial tissue engineering.

3.
Biomaterials ; 113: 31-41, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27810640

RESUMEN

A variety of techniques have been applied to generate tissue engineered constructs, where cells are combined with degradable scaffolds followed by a period of in vitro culture or direct implantation. In the current study, a cellularized scaffold was generated by concurrent deposition of electrospun biodegradable elastomer (poly(ester urethane)urea, PEUU) and electrosprayed culture medium + skeletal muscle-derived stem cells (MDSCs) or electrosprayed culture medium alone as a control. MDSCs were obtained from green fluorescent protein (GFP) transgenic rats. The created scaffolds were implanted into allogenic strain-matched rats to replace a full thickness abdominal wall defect. Both control and MDSC-integrated scaffolds showed extensive cellular infiltration at 4 and 8 wk. The number of blood vessels was higher, the area of residual scaffold was lower, number of multinucleated giant cells was lower and area of connective tissue was lower in MDSC-integrated scaffolds (p < 0.05). GFP + cells co-stained positive for VEGF. Bi-axial mechanical properties of the MDSC-microintegrated constructs better approximated the anisotropic behavior of the native abdominal wall. GFP + cells were observed throughout the scaffold at ∼5% of the cell population at 4 and 8 wk. RNA expression at 4 wk showed higher expression of early myogenic marker Pax7, and b-FGF in the MDSC group. Also, higher expression of myogenin and VEGF were seen in the MDSC group at both 4 and 8 wk time points. The paracrine effect of donor cells on host cells likely contributed to the differences found in vivo between the groups. This approach for the rapid creation of highly-cellularized constructs with soft tissue like mechanics offers an attractive methodology to impart cell-derived bioactivity into scaffolds providing mechanical support during the healing process and might find application in a variety of settings.


Asunto(s)
Pared Abdominal/fisiología , Elastómeros/química , Músculo Esquelético/citología , Regeneración , Trasplante de Células Madre , Células Madre/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Pared Abdominal/irrigación sanguínea , Pared Abdominal/patología , Animales , Materiales Biocompatibles/química , Células Cultivadas , Femenino , Poliuretanos/química , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Cicatrización de Heridas
4.
Muscle Nerve ; 51(5): 750-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25242666

RESUMEN

INTRODUCTION: Acute compartment syndrome (CS) is caused by an elevation of pressure within a muscular compartment that can be caused by numerous factors, including blunt trauma. In this study, we characterized a rodent model of CS-like injury. METHODS: Forty male athymic rats received a standardized injury of ischemia and compression to their hindlimbs, while the intracompartmental pressure (ICP) was measured using an implantable transmitter. Tetanic muscle function was evaluated, and histology was performed on the tibialis anterior (TA) muscle. RESULTS: ICPs were held at 260.70 ± 2.70 mm Hg during injury. Injured muscles recovered 59% of their total function 4 weeks after injury, and histology showed high levels of edema, inflammation (CD68(+) ), angiogenesis (CD31(+) ), and fibrosis within 72 hours after injury. CONCLUSIONS: We describe a novel CS-like injury model and a novel method to measure ICP, which could potentially be used to develop innovative therapies to manage CS injury in patients.


Asunto(s)
Síndromes Compartimentales/fisiopatología , Modelos Animales de Enfermedad , Músculo Esquelético/lesiones , Músculo Esquelético/fisiopatología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores/metabolismo , Fibrosis , Masculino , Músculo Esquelético/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Presión/efectos adversos , Ratas , Ratas Desnudas , Recuperación de la Función/fisiología
5.
Sci Rep ; 4: 4826, 2014 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-24769917

RESUMEN

Understanding stem cell (SC) population dynamics is essential for developing models that can be used in basic science and medicine, to aid in predicting cells fate. These models can be used as tools e.g. in studying patho-physiological events at the cellular and tissue level, predicting (mal)functions along the developmental course, and personalized regenerative medicine. Using time-lapsed imaging and statistical tools, we show that the dynamics of SC populations involve a heterogeneous structure consisting of multiple sub-population behaviors. Using non-Gaussian statistical approaches, we identify the co-existence of fast and slow dividing subpopulations, and quiescent cells, in stem cells from three species. The mathematical analysis also shows that, instead of developing independently, SCs exhibit a time-dependent fractal behavior as they interact with each other through molecular and tactile signals. These findings suggest that more sophisticated models of SC dynamics should view SC populations as a collective and avoid the simplifying homogeneity assumption by accounting for the presence of more than one dividing sub-population, and their multi-fractal characteristics.


Asunto(s)
Modelos Biológicos , Modelos Estadísticos , Células Madre/citología , Células Madre/fisiología , Animales , División Celular , Proliferación Celular , Humanos , Ratones , Ratas , Imagen de Lapso de Tiempo
6.
Postgrad Med J ; 89(1057): 666-70, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24129034

RESUMEN

Skeletal muscle injuries are among the most common and frequently disabling injuries sustained by athletes. Repair of injured skeletal muscle is an area that continues to present a challenge for sports medicine clinicians and researchers due, in part, to complete muscle recovery being compromised by development of fibrosis leading to loss of function and susceptibility to re-injury. Injured skeletal muscle goes through a series of coordinated and interrelated phases of healing including degeneration, inflammation, regeneration and fibrosis. Muscle regeneration initiated shortly after injury can be limited by fibrosis which affects the degree of recovery and predisposes the muscle to reinjury. It has been demonstrated in animal studies that antifibrotic agents that inactivate transforming growth factor (TGF)-ß1 have been effective at decreasing scar tissue formation. Several studies have also shown that vascular endothelial growth factor (VEGF) can increase the efficiency of skeletal muscle repair by increasing angiogenesis and, at the same time, reducing the accumulation of fibrosis. We have isolated and thoroughly characterised a population of skeletal muscle-derived stem cells (MDSCs) that enhance repair of damaged skeletal muscle fibres by directly differentiating into myofibres and secreting paracrine factors that promote tissue repair. Indeed, we have found that MDSCs transplanted into skeletal and cardiac muscles have been successful at repair probably because of their ability to secrete VEGF that works in a paracrine fashion. The application of these techniques to the study of sport-related muscle injuries awaits investigation. Other useful strategies to enhance skeletal muscle repair through increased vascularisation may include gene therapy, exercise, neuromuscular electrical stimulation and, potentially, massage therapy. Based on recent studies showing an accelerated recovery of muscle function from intense eccentric exercise through massage-based therapies, we believe that this treatment modality offers a practical and non-invasive form of therapy for skeletal muscle injuries. However, the biological mechanism(s) behind the beneficial effect of massage are still unclear and require further investigation using animal models and potentially randomised, human clinical studies.

7.
J Orthop Res ; 31(7): 1089-95, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23553740

RESUMEN

We have previously reported the high regenerative potential of murine muscle-derived stem cells (mMDSCs) that are capable of differentiating into multiple mesodermal cell lineages, including myogenic, endothelial, chondrocytic, and osteoblastic cells. Recently, we described a putative human counterpart of mMDSCs, the myogenic endothelial cells (MECs), in adult human skeletal muscle, which efficiently repair/regenerate the injured and dystrophic skeletal muscle as well as the ischemic heart in animal disease models. Nevertheless it remained unclear whether human MECs, at the clonal level, preserve mMDSC-like chondrogenic and osteogenic potentials and classic stem cell characteristics including high proliferation and resistance to stress. Herein, we demonstrated that MECs, sorted from fresh postnatal human skeletal muscle biopsies, can be grown clonally and exhibit robust resistance to oxidative stress with no tumorigeneity. MEC clones were capable of differentiating into chondrocytes and osteoblasts under inductive conditions in vitro and participated in cartilage and bone formation in vivo. Additionally, adipogenic and angiogenic potentials of clonal MECs (cMECs) were observed. Overall, our study showed that cMECs not only display typical properties of adult stem cells but also exhibit chondrogenic and osteogenic capacities in vitro and in vivo, suggesting their potential applications in articular cartilage and bone repair/regeneration.


Asunto(s)
Diferenciación Celular/fisiología , Condrogénesis/fisiología , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Osteogénesis/fisiología , Adipocitos/citología , Adulto , Animales , Biopsia , Proliferación Celular , Células Cultivadas , Condrocitos/citología , Endotelio/citología , Endotelio/fisiología , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones SCID , Osteoblastos/citología , Estrés Oxidativo/fisiología , Trasplante Heterólogo
8.
Mol Ther ; 21(3): 662-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23319053

RESUMEN

Direct intracardiac cell injection for heart repair is hindered by numerous limitations including: cell death, poor spreading of the injected cells, arrhythmia, needle injury, etc. Tissue-engineered cell sheet implantation has the potential to overcome some of these limitations. We evaluated whether the transplantation of a muscle-derived stem cell (MDSC) sheet could improve the regenerative capacity of MDSCs in a chronic model of myocardial infarction. MDSC sheet-implanted mice displayed a reduction in left ventricle (LV) dilation and sustained LV contraction compared with the other groups. The MDSC sheet formed aligned myotubes and produced a significant increase in capillary density and a reduction of myocardial fibrosis compared with the other groups. Hearts transplanted with the MDSC sheets did not display any significant arrhythmias and the donor MDSC survival rate was higher than the direct myocardial MDSC injection group. MDSC sheet implantation yielded better functional recovery of chronic infarcted myocardium without any significant arrhythmic events compared with direct MDSC injection, suggesting this cell sheet delivery system could significantly improve the myocardial regenerative potential of the MDSCs.


Asunto(s)
Arritmias Cardíacas/prevención & control , Células Musculares/citología , Infarto del Miocardio/fisiopatología , Trasplante de Células Madre , Células Madre/citología , Ingeniería de Tejidos , Animales , Arritmias Cardíacas/fisiopatología , Capilares/fisiopatología , Masculino , Ratones , Ratones Endogámicos NOD , Mioblastos/citología , Mioblastos/metabolismo , Mioblastos/trasplante , Infarto del Miocardio/terapia , Miocardio/citología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Función Ventricular Izquierda
9.
Br J Sports Med ; 47(9): 556-60, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23197410

RESUMEN

Skeletal muscle injuries are among the most common and frequently disabling injuries sustained by athletes. Repair of injured skeletal muscle is an area that continues to present a challenge for sports medicine clinicians and researchers due, in part, to complete muscle recovery being compromised by development of fibrosis leading to loss of function and susceptibility to re-injury. Injured skeletal muscle goes through a series of coordinated and interrelated phases of healing including degeneration, inflammation, regeneration and fibrosis. Muscle regeneration initiated shortly after injury can be limited by fibrosis which affects the degree of recovery and predisposes the muscle to reinjury. It has been demonstrated in animal studies that antifibrotic agents that inactivate transforming growth factor (TGF)-ß1 have been effective at decreasing scar tissue formation. Several studies have also shown that vascular endothelial growth factor (VEGF) can increase the efficiency of skeletal muscle repair by increasing angiogenesis and, at the same time, reducing the accumulation of fibrosis. We have isolated and thoroughly characterised a population of skeletal muscle-derived stem cells (MDSCs) that enhance repair of damaged skeletal muscle fibres by directly differentiating into myofibres and secreting paracrine factors that promote tissue repair. Indeed, we have found that MDSCs transplanted into skeletal and cardiac muscles have been successful at repair probably because of their ability to secrete VEGF that works in a paracrine fashion. The application of these techniques to the study of sport-related muscle injuries awaits investigation. Other useful strategies to enhance skeletal muscle repair through increased vascularisation may include gene therapy, exercise, neuromuscular electrical stimulation and, potentially, massage therapy. Based on recent studies showing an accelerated recovery of muscle function from intense eccentric exercise through massage-based therapies, we believe that this treatment modality offers a practical and non-invasive form of therapy for skeletal muscle injuries. However, the biological mechanism(s) behind the beneficial effect of massage are still unclear and require further investigation using animal models and potentially randomised, human clinical studies.


Asunto(s)
Masaje , Músculo Esquelético/lesiones , Trasplante de Células Madre/métodos , Células Madre/fisiología , Traumatismos en Atletas/terapia , Terapia por Estimulación Eléctrica/métodos , Terapia por Ejercicio/métodos , Humanos , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Recuperación de la Función , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Cicatrización de Heridas/fisiología
10.
Med Sci Sports Exerc ; 45(5): 813-23, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23247715

RESUMEN

PURPOSE: Surgical repairs of tears in the vascular region of the meniscus usually heal better than repairs performed in the avascular region; thus, we hypothesized that this region might possess a richer supply of vascular-derived stem cells than the avascular region. METHODS: In this study, we analyzed 6 menisci extracted from aborted human fetuses and 12 human lateral menisci extracted from adult human subjects undergoing total knee arthroplasty. Menisci were immunostained for CD34 (a stem cell marker) and CD146 (a pericyte marker) in situ, whereas other menisci were dissected into two regions (peripheral and inner) and used to isolate meniscus-derived cells by flow cytometry. Cell populations expressing CD34 and CD146 were tested for their multilineage differentiation potentials, including chondrogenic, osteogenic, and adipogenic lineages. Fetal peripheral meniscus cells were transplanted by intracapsular injection into the knee joints of an athymic rat meniscal tear model. Rat menisci were extracted and histologically evaluated after 4 wk posttransplantation. RESULTS: Immunohistochemistry and flow cytometric analyses demonstrated that a higher number of CD34- and CD146-positive cells were found in the peripheral region compared with the inner region. The CD34- and CD146-positive cells isolated from the vascular region of both fetal and adult menisci demonstrated multilineage differentiation capacities and were more potent than cells isolated from the inner (avascular) region. Fetal CD34- and CD146-positive cells transplanted into the athymic rat knee joint were recruited into the meniscal tear sites and contributed to meniscus repair. CONCLUSIONS: The vascularized region of the meniscus contains more stem cells than the avascular region. These meniscal-derived stem cells were multipotent and contributed to meniscal regeneration.


Asunto(s)
Meniscos Tibiales/cirugía , Regeneración/fisiología , Cicatrización de Heridas/fisiología , Anciano , Animales , Antígenos CD34/metabolismo , Antígeno CD146/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Masculino , Meniscos Tibiales/citología , Persona de Mediana Edad , Ratas
11.
Muscle Nerve ; 46(1): 70-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22644812

RESUMEN

INTRODUCTION: It has been reported that suramin treatment can improve muscle healing; however, details about optimizing the dosing requirements remain unclear. The purpose of this study was to determine the optimal timing of suramin administration and investigate the effects it had on the expression of myostatin, follistatin, and muscle vascularity after muscle injury. METHODS: Contusion injured muscles of mice were treated with suramin at 1, 2, or 3 weeks post-injury and evaluated histologically and physiologically at 1, 2, and 10 days after injection. RESULTS: Suramin treatment initiated at 2 weeks post-injury was observed to promote muscle regeneration and muscle strength, and to decrease fibrosis. Suramin reduced myostatin expression and increased follistatin expression and vascularity in injured skeletal muscle. CONCLUSIONS: Suramin's positive effect on muscle regeneration is thought to be due to its enhancement of follistatin expression which increases neoangiogenesis and inhibits myostatin's promotion of fibrosis.


Asunto(s)
Antineoplásicos/uso terapéutico , Contusiones/tratamiento farmacológico , Músculo Esquelético/lesiones , Suramina/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos , Animales , Antineoplásicos/administración & dosificación , Contusiones/metabolismo , Esquema de Medicación , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis/metabolismo , Folistatina/metabolismo , Masculino , Ratones , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Suramina/administración & dosificación
12.
Birth Defects Res C Embryo Today ; 96(1): 82-94, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22457179

RESUMEN

Skeletal muscle injury and repair are complex processes, including well-coordinated steps of degeneration, inflammation, regeneration, and fibrosis. We have reviewed the recent literature including studies by our group that describe how to modulate the processes of skeletal muscle repair and regeneration. Antiinflammatory drugs that target cyclooxygenase-2 were found to hamper the skeletal muscle repair process. Muscle regeneration phase can be aided by growth factors, including insulin-like growth factor-1 and nerve growth factor, but these factors are typically short-lived, and thus more effective methods of delivery are needed. Skeletal muscle damage caused by traumatic injury or genetic diseases can benefit from cell therapy; however, the majority of transplanted muscle cells (myoblasts) are unable to survive the immune response and hypoxic conditions. Our group has isolated neonatal skeletal muscle derived stem cells (MDSCs) that appear to repair muscle tissue in a more effective manner than myoblasts, most likely due to their better resistance to oxidative stress. Enhancing antioxidant levels of MDSCs led to improved regenerative potential. It is becoming increasingly clear that stem cells tissue repair by direct differentiation and paracrine effects leading to neovascularization of injured site and chemoattraction of host cells. The factors invoked in paracrine action are still under investigation. Our group has found that angiotensin II receptor blocker (losartan) significantly reduces fibrotic tissue formation and improves repair of murine injured muscle. Based on these data, we have conducted a case study on two hamstring injury patients and found that losartan treatment was well tolerated and possibly improved recovery time. We believe this medication holds great promise to optimize muscle repair in humans.


Asunto(s)
Músculo Esquelético , Enfermedades Musculares/terapia , Medicina Regenerativa/métodos , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Modelos Animales de Enfermedad , Humanos , Losartán/farmacología , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Neovascularización Fisiológica , Estrés Oxidativo , Comunicación Paracrina , Regeneración , Trasplante de Células Madre , Células Madre/citología , Células Madre/fisiología , Traumatismos de los Tendones/terapia , Resultado del Tratamiento , Cicatrización de Heridas
13.
J Shoulder Elbow Surg ; 21(2): 181-90, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22244061

RESUMEN

BACKGROUND: Despite the advances in surgical procedures to repair the rotator cuff, there is a high incidence of failure. Biologic approaches, such as growth factor delivery and stem cell and gene therapy, are potential targets for optimization to improve the outcome of rotator cuff therapies and reduce rates of reinjury. This article outlines the current evidence for growth factor and stem cell therapy in tendon healing and the augmentation of rotator cuff repair. METHODS: Literature on the PubMed-National Center for Biotechnology Information database was searched using the keywords growth factor, factor, gene therapy, stem cell, mesenchymal, or bone marrow in combination with rotator cuff, supraspinatus, or infraspinatus. Articles that studied growth factors or stem cells alone in rotator cuff repair were selected. Only 3 records showed use of stem cells in rotator cuff repair; thus, we expanded our search to include selected studies on stem cells and Achilles or patellar tendon repairs. Bibliographies and proceedings of meetings were searched to include additional applicable studies. We also included hitherto unpublished data by our group on the use of stem cell transplantation for rotator cuff therapy. RESULTS: More than 70 articles are summarized, with focus on recent original research papers and significant reviews that summarized earlier records. CONCLUSIONS: Use of growth factors, stem cell therapy, and other tissue-engineering means serve to augment classical surgical rotator cuff repair procedures. The combination of stem cells and growth factors resulted in enhanced repair that emulated uninjured tissue, but the literature search reflected paucity of research in this field. Preclinical evidence from gene therapy and stem cell studies can be used as a start to move therapy from the experimental phase to clinical translation in patients.


Asunto(s)
Terapia Biológica/métodos , Lesiones del Manguito de los Rotadores , Traumatismos de los Tendones/terapia , Cicatrización de Heridas/fisiología , Animales , Modelos Animales de Enfermedad , Terapia Genética/métodos , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Trasplante de Células Madre Mesenquimatosas/métodos , Conejos , Traumatismos de los Tendones/patología , Ingeniería de Tejidos
14.
PLoS One ; 6(9): e24801, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21966370

RESUMEN

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


Asunto(s)
Nervio Femoral/fisiopatología , Nervio Femoral/cirugía , Vena Femoral/trasplante , Regeneración Nerviosa , Animales , Axones/metabolismo , Axones/ultraestructura , Movimiento Celular , Proliferación Celular , Células Cultivadas , Femenino , Nervio Femoral/lesiones , Vena Femoral/citología , Vena Femoral/efectos de la radiación , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Microscopía Electrónica de Transmisión , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/ultraestructura , Proteínas de Neurofilamentos/metabolismo , Ratas , Ratas Endogámicas F344 , Células de Schwann/metabolismo , Células de Schwann/ultraestructura , Factores Sexuales , Trasplante Homólogo
15.
Stem Cell Res Ther ; 2(4): 31, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21745421

RESUMEN

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


Asunto(s)
Diferenciación Celular , Trasplante de Células Madre , Animales , Enfermedades de los Cartílagos/patología , Enfermedades de los Cartílagos/terapia , Tratamiento Basado en Trasplante de Células y Tejidos , Microambiente Celular , Cardiopatías/patología , Cardiopatías/terapia , Humanos , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/fisiología , Neovascularización Patológica , Comunicación Paracrina , Regeneración , Células Madre/citología , Células Madre/fisiología
16.
Am J Pathol ; 179(2): 915-30, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21689628

RESUMEN

Recovery from skeletal muscle injury is often incomplete because of the formation of fibrosis and inadequate myofiber regeneration; therefore, injured muscle could benefit significantly from therapies that both stimulate muscle regeneration and inhibit fibrosis. To this end, we focused on blocking myostatin, a member of the transforming growth factor-ß superfamily and a negative regulator of muscle regeneration, with the myostatin antagonist follistatin. In vivo, follistatin-overexpressing transgenic mice underwent significantly greater myofiber regeneration and had less fibrosis formation compared with wild-type mice after skeletal muscle injury. Follistatin's mode of action is likely due to its ability to block myostatin and enhance neovacularization. Furthermore, muscle progenitor cells isolated from follistatin-overexpressing mice were significantly superior to muscle progenitors isolated from wild-type mice at regenerating dystrophin-positive myofibers when transplanted into the skeletal muscle of dystrophic mdx/severe combined immunodeficiency mice. In vitro, follistatin stimulated myoblasts to express MyoD, Myf5, and myogenin, which are myogenic transcription factors that promote myogenic differentiation. Moreover, follistatin's ability to enhance muscle differentiation is at least partially due to its ability to block myostatin, activin A, and transforming growth factor-ß1, all of which are negative regulators of muscle cell differentiation. The findings of this study suggest that follistatin is a promising agent for improving skeletal muscle healing after injury and muscle diseases, such as the muscular dystrophies.


Asunto(s)
Fibrosis/patología , Folistatina/química , Músculo Esquelético/metabolismo , Animales , Línea Celular , Trasplante de Células , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente/métodos , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/metabolismo , Miostatina/metabolismo , Neovascularización Patológica , Regeneración , Factor de Crecimiento Transformador beta/metabolismo
17.
Biomaterials ; 32(13): 3413-22, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21316755

RESUMEN

The capability to engineer microenvironmental cues to direct a stem cell population toward multiple fates, simultaneously, in spatially defined regions is important for understanding the maintenance and repair of multi-tissue units. We have previously developed an inkjet-based bioprinter to create patterns of solid-phase growth factors (GFs) immobilized to an extracellular matrix (ECM) substrate, and applied this approach to drive muscle-derived stem cells toward osteoblasts 'on-pattern' and myocytes 'off-pattern' simultaneously. Here this technology is extended to spatially control osteoblast, tenocyte and myocyte differentiation simultaneously. Utilizing immunofluorescence staining to identify tendon-promoting GFs, fibroblast growth factor-2 (FGF-2) was shown to upregulate the tendon marker Scleraxis (Scx) in C3H10T1/2 mesenchymal fibroblasts, C2C12 myoblasts and primary muscle-derived stem cells, while downregulating the myofibroblast marker α-smooth muscle actin (α-SMA). Quantitative PCR studies indicated that FGF-2 may direct stem cells toward a tendon fate via the Ets family members of transcription factors such as pea3 and erm. Neighboring patterns of FGF-2 and bone morphogenetic protein-2 (BMP-2) printed onto a single fibrin-coated coverslip upregulated Scx and the osteoblast marker ALP, respectively, while non-printed regions showed spontaneous myotube differentiation. This work illustrates spatial control of multi-phenotype differentiation and may have potential in the regeneration of multi-tissue units.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Animales , Línea Celular , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/farmacología , Técnica del Anticuerpo Fluorescente , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Mioblastos/citología , Mioblastos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Tendones/citología
18.
Mol Ther ; 19(4): 790-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21304494

RESUMEN

Wnt signaling plays a crucial role in regulating cell proliferation, differentiation and inducing cardiomyogenesis. Skeletal muscle-derived stem cells (MDSCs) have been shown to be multipotent; however, their potential to aid in the healing of the heart after myocardial infarction appears to be due to the paracrine effects they impart on the host environment. The goal of this study was to investigate whether Wnt11 could promote the differentiation of MDSCs into cardiomyocytes and enhance the repair of infarcted myocardium. MDSCs transduced with a lentivirus encoding for Wnt11 increased mRNA and protein expression of the early cardiac markers NK2 transcription factor related 5 (NKx2.5) and Connexin43 (Cx43) and also led to an increased expression of late-stage cardiac markers including: α, ß-myosin heavy chain (MHC) and brain natriuretic protein (BNP) at the mRNA level, and MHC and Troponin I (TnI) at the protein level. We also observed that Wnt11 expression significantly enhanced c-jun N-terminal kinase activity in transduced MDSCs, and that some of the cells beat spontaneously but are not fully differentiated cardiomyocytes. Finally, lentivirus-Wnt11-transduced MDSCs showed greater survival and cardiac differentiation after being transplanted into acutely infarct-injured myocardium. These findings could one day lead to strategies that could be utilized in cardiomyoplasty treatments of myocardial infarction.


Asunto(s)
Diferenciación Celular/fisiología , Vectores Genéticos/genética , Lentivirus/genética , Fibras Musculares Esqueléticas/citología , Células Madre/citología , Células Madre/metabolismo , Proteínas Wnt/metabolismo , Animales , Western Blotting , Diferenciación Celular/genética , Inmunohistoquímica , Masculino , Ratones , Ratones SCID , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Wnt/genética
19.
Int J Clin Exp Pathol ; 3(7): 681-90, 2010 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-20830239

RESUMEN

The liver is unique for its ability to regenerate after injury, however, critical injuries or disease cause it to lose this quality. Stem cells have been explored as a possibility to restore the function of seriously damaged livers, based on their self-renewability and multiple differentiation capacity. These experiments examine the ability of muscle derived stem cells (MDSCs) to differentiate into hepatocyte-like cells in vitro and acquire functional liver attributes for repairing damaged livers. In vitro experiments were performed using MDSCs from postnatal mice and mouse hepatocyte cell lines. Our data revealed that MDSCs differentiated into hepatocyte-like cells and expressed liver cell markers, albumin, hepatocyte nuclear factor 4 α, and alpha feto-protein, both at the RNA and protein level. Additionally, in vivo studies showed successful engraftment of MDSCs into hepatectomized mouse livers of mice. These results provide evidence suggesting that MDSCs have the capacity to differentiate into liver cell-like cells and may serve as potential candidates to aid in liver regeneration.


Asunto(s)
Hepatocitos/citología , Regeneración Hepática/fisiología , Músculo Esquelético/citología , Células Madre/citología , Animales , Diferenciación Celular/fisiología , Técnicas de Cocultivo , Inmunohistoquímica , Hibridación Fluorescente in Situ , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Biomaterials ; 31(32): 8235-44, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20684982

RESUMEN

The success of small-diameter tissue engineered vascular grafts (TEVGs) greatly relies on an appropriate cell source and an efficient cellular delivery and carrier system. Pericytes have recently been shown to express mesenchymal stem cell features. Their relative availability and multipotentiality make them a promising candidate for TEVG applications. The objective of this study was to incorporate pericytes into a biodegradable scaffold rapidly, densely and efficiently, and to assess the efficacy of the pericyte-seeded scaffold in vivo. Bi-layered elastomeric poly(ester-urethane)urea scaffolds (length = 10 mm; inner diameter = 1.3 mm) were bulk seeded with 3 x 10(6) pericytes using a customized rotational vacuum seeding device in less than 2 min (seeding efficiency > 90%). The seeded scaffolds were cultured in spinner flasks for 2 days and then implanted into Lewis rats as aortic interposition grafts for 8 weeks. Results showed pericytes populated the porous layer of the scaffolds evenly and maintained their original phenotype after the dynamic culture. After implantation, pericyte-seeded TEVGs showed a significant higher patency rate than the unseeded control: 100% versus 38% (p < 0.05). Patent pericyte-seeded TEVGs revealed extensive tissue remodeling with collagen and elastin present. The remodeled tissue consisted of multiple layers of alpha-smooth muscle actin- and calponin-positive cells, and a von Willebrand factor-positive monolayer in the lumen. These results demonstrate the feasibility of a pericyte-based TEVG and suggest that the pericytes play a role in maintaining patency of the TEVG as an arterial conduit.


Asunto(s)
Prótesis Vascular , Pericitos/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Aorta/ultraestructura , Implantación de Prótesis Vascular , Células Cultivadas , Femenino , Humanos , Ratas , Ratas Endogámicas Lew , Grado de Desobstrucción Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA