Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Intervalo de año de publicación
1.
PLoS One ; 16(9): e0251166, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34529657

RESUMEN

Platelet-rich plasma (PRP) is a widely used autologous treatment for tendon injuries in clinics. Platelets (PLTs) are a major source of high mobility group box1 (HMGB1) that is gaining attention as a chemoattractant that can recruit stem cells to the wound area to enhance healing of injured tissues; however, the contribution of PLT HMGB1 in wounded tendon healing remains unexplored. This study investigated the effect of PLT HMGB1 within PRP on tendon healing using PLT HMGB1 knockout (KO) and GFP mice. A window defect was created in the patellar tendons of both groups of mice, and wounds were treated with either saline, PRP isolated from PLT HMGB1-KO mice, or PRP isolated from GFP mice. Seven days post-treatment, animals were sacrificed and analyzed by gross inspection, histology, and immunostaining for characteristic signs of tendon healing and repair. Our results showed that in comparison to mice treated with PRP from PLT HMGB1-KO mice, wounds treated with PRP from GFP mice healed faster and exhibited a better organization in tendon structure. Mice treated with PRP from PLT HMGB1-KO mice produced tendon tissue with large premature wound areas and low cell densities. However, wounds of PLT HMGB1-KO mice showed better healing with PRP from HMGB1-KO mice compared to saline treatment. Moreover, wounds treated with PRP from GFP mice had increased extracellular HMGB1, decreased CD68, increased stem cell markers CD146 and CD73, and increased collagen III protein expression levels compared to those treated with PRP from PLT HMGB1-KO mice. Thus, PLT HMGB1 within PRP plays an important role in tendon wound healing by decreasing inflammation, increasing local HMGB1 levels, and recruiting stem cells to the wound area in the tendon. Our findings also suggest that the efficacy of PRP treatment for tendon injuries in clinics may depend on PLT HMGB1 within PRP preparations.


Asunto(s)
Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Plasma Rico en Plaquetas/fisiología , Traumatismos de los Tendones/terapia , Cicatrización de Heridas , 5'-Nucleotidasa/metabolismo , Animales , Antígeno CD146/metabolismo , Colágeno Tipo III/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Plasma Rico en Plaquetas/metabolismo , Traumatismos de los Tendones/genética , Traumatismos de los Tendones/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Regulación hacia Arriba
2.
Hepatology ; 73(6): 2494-2509, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32924145

RESUMEN

BACKGROUND AND AIMS: Liver ischemia/reperfusion injury (IRI) induces local and systemic inflammation in which neutrophil extracellular traps (NETs) are major drivers. IRI markedly augments metastatic growth, which is consistent with the notion that the liver IRI can serve as a premetastatic niche. Exercise training (ExT) confers a sustainable protection, reducing IRI in some animal models, and has been associated with improved survival in patients with cancer; however, the impact of ExT on liver IRI or development of hepatic metastases is unknown. APPROACH AND RESULTS: Mice were randomized into exercise (ExT) and sedentary groups before liver IRI and tumor injection. Computerized dynamic network analysis of 20 inflammatory mediators was used to dissect the sequence of mediator interactions after ischemia/reperfusion (I/R) that induce injury. ExT mice showed a significant decrease in hepatic IRI and tissue necrosis. This coincided with disassembly of complex networks among inflammatory mediators seen in sedentary mice. Neutrophil infiltration and NET formation were decreased in the ExT group, which suppressed the expression of liver endothelial cell adhesion molecules. Concurrently, ExT mice revealed a distinct population of infiltrating macrophages expressing M2 phenotypic genes. In a metastatic model, fewer metastases were present 3 weeks after I/R in the ExT mice, a finding that correlated with a marked increase in tumor-suppressing T cells within the tumor microenvironment. CONCLUSIONS: ExT preconditioning mitigates the inflammatory response to liver IRI, protecting the liver from injury and metastases. In light of these findings, potential may exist for the reduction of liver premetastatic niches induced by liver IRI through the use of ExT as a nonpharmacologic therapy before curative surgical approaches.


Asunto(s)
Trampas Extracelulares/inmunología , Inflamación , Hepatopatías , Metástasis de la Neoplasia , Infiltración Neutrófila/inmunología , Condicionamiento Físico Animal/métodos , Daño por Reperfusión , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Inmunidad , Inflamación/etiología , Inflamación/inmunología , Inflamación/terapia , Hepatopatías/inmunología , Hepatopatías/patología , Hepatopatías/terapia , Ratones , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/terapia , Factores Protectores , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Daño por Reperfusión/terapia , Resultado del Tratamiento
3.
PLoS One ; 15(12): e0242640, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33373386

RESUMEN

To examine the differential mechanobiological responses of specific resident tendon cells, we developed an in vivo model of whole-body irradiation followed by injection of either tendon stem/progenitor cells (TSCs) expressing green fluorescent protein (GFP-TSCs) or mature tenocytes expressing GFP (GFP-TNCs) into the patellar tendons of wild type C57 mice. Injected mice were subjected to short term (3 weeks) treadmill running, specifically moderate treadmill running (MTR) and intensive treadmill running (ITR). In MTR mice, both GFP-TSC and GFP-TNC injected tendons maintained normal cell morphology with elevated expression of tendon related markers collagen I and tenomodulin. In ITR mice injected with GFP-TNCs, cells also maintained an elongated shape similar to the shape found in normal/untreated control mice, as well as elevated expression of tendon related markers. However, ITR mice injected with GFP-TSCs showed abnormal changes, such as cell morphology transitioning to a round shape, elevated chondrogenic differentiation, and increased gene expression of non-tenocyte related genes LPL, Runx-2, and SOX-9. Increased gene expression data was supported by immunostaining showing elevated expression of SOX-9, Runx-2, and PPARγ. This study provides evidence that while MTR maintains tendon homeostasis by promoting the differentiation of TSCs into TNCs, ITR causes the onset of tendinopathy development by inducing non-tenocyte differentiation of TSCs, which may eventually lead to the formation of non-tendinous tissues in tendon tissue after long term mechanical overloading conditions on the tendon.


Asunto(s)
Condrocitos/citología , Células Madre/citología , Tendinopatía/patología , Tendones/patología , Tenocitos/citología , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Forma de la Célula , Rastreo Celular , Condrocitos/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Prueba de Esfuerzo , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , PPAR gamma/genética , PPAR gamma/metabolismo , Condicionamiento Físico Animal/efectos adversos , Carrera , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Células Madre/metabolismo , Tendinopatía/etiología , Tendinopatía/genética , Tendinopatía/metabolismo , Tendones/metabolismo , Tenocitos/metabolismo
4.
Arthritis Rheumatol ; 70(2): 230-241, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29024567

RESUMEN

OBJECTIVE: To establish an unbiased, 3-dimensional (3-D) approach that quantifies subchondral bone plate (SBP) changes in mouse joints, and to investigate the mechanism that mediates SBP sclerosis at a late stage of osteoarthritis (OA). METHODS: A new micro-computed tomography (micro-CT) protocol was developed to characterize the entire thickness of the SBP in the distal femur of a normal mouse knee. Four mouse models of severe joint OA were generated: cartilage-specific Egfr-knockout (Egfr-CKO) mice at 2 months after surgical destabilization of the medial meniscus (DMM), Egfr-CKO mice with aging-related spontaneous OA, wild-type (WT) mice at 10 months after DMM, and WT mice at 14 weeks after DMM plus hemisectomy of the meniscus (DMMH) surgery. As an additional model, mice with knockout of the sclerostin gene (Sost-KO) were subjected to DMMH surgery. Knee joints were examined by micro-CT, histology, and immunohistochemical analyses. RESULTS: Examination of the mouse distal femur by 3-D micro-CT revealed a positive correlation between SBP thickness and the loading status in normal knees. In all 4 mouse models of late-stage OA, SBP sclerosis was restricted to the areas under severely eroded articular cartilage. This was accompanied by elevated bone formation at the bone marrow side of the SBP and a drastic reduction in the levels of sclerostin in osteocytes within the SBP. Unlike in WT mice, no further increase in the thickness of the SBP was observed in response to DMMH in Sost-KO mice. CONCLUSION: Since focal stress on the SBP underlying sites of cartilage damage increases during late stages of OA, these findings establish mechanical loading-induced attenuation of sclerostin expression and elevation of bone formation along the SBP surface as the major mechanisms characterizing subchondral bone phenotypes associated with severe late-stage OA in mice.


Asunto(s)
Huesos/patología , Glicoproteínas/metabolismo , Articulación de la Rodilla/patología , Osteoartritis de la Rodilla/patología , Osteosclerosis/etiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Huesos/metabolismo , Modelos Animales de Enfermedad , Fémur/patología , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular , Articulación de la Rodilla/metabolismo , Masculino , Ratones , Ratones Noqueados , Osteoartritis de la Rodilla/complicaciones , Osteoartritis de la Rodilla/metabolismo , Osteosclerosis/metabolismo , Estrés Mecánico , Microtomografía por Rayos X
5.
Biochemistry ; 55(43): 6046-6055, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27733037

RESUMEN

Cell traction force (CTF) plays a critical role in controlling cell shape, permitting cell motility, and maintaining cellular homeostasis in many biological processes such as angiogenesis, development, wound healing, and cancer metastasis. Calponin is an actin filament-associated cytoskeletal protein in smooth muscles and multiple types of non-muscle cells. An established biochemical function of calponin is the inhibition of myosin ATPase in smooth muscle cells. Vertebrates have three calponin isoforms. Among them, calponin 2 is expressed in epithelial cells, endothelial cells, macrophages, myoblasts, and fibroblasts and plays a role in regulating cytoskeleton activities such as cell adhesion, migration, and cytokinesis. Knockout (KO) of the gene encoding calponin 2 (Cnn2) in mice increased cell motility, suggesting a function of calponin 2 in modulating CTF. In this study, we examined fibroblasts isolated from Cnn2 KO and wild-type (WT) mice using CTF microscopy. Primary mouse fibroblasts were cultured on polyacrylamide gel substrates embedded with fluorescent beads to measure root-mean-square traction, total strain energy, and net contractile movement. The results showed that calponin 2-null fibroblasts exhibit traction force greater than that of WT cells. Adherent calponin 2-null fibroblasts de-adhered faster than the WT control during mild trypsin treatment, consistent with an increased CTF. Blebbistatin, an inhibitor of myosin II ATPase, is more effective upon an alteration in cell morphology when calponin 2 is present in WT fibroblasts than that on Cnn2 KO cells, indicating their additive effects in inhibiting myosin motor activity. The novel finding that calponin 2 regulates myosin-dependent CTF in non-muscle cells demonstrates a mechanism for controlling cell motility-based functions.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Microfilamentos/metabolismo , Miosina Tipo II/metabolismo , Animales , Células Cultivadas , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Ratones Noqueados , Calponinas
6.
Biomed Res Int ; 2016: 9103792, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27610386

RESUMEN

Platelet-Rich Plasma (PRP) has been widely used in orthopaedic surgery and sport medicine to treat tendon injuries. However, the efficacy of PRP treatment for tendinopathy is controversial. This paper focuses on reviewing the basic science studies on PRP performed under well-controlled conditions. Both in vitro and in vivo studies describe PRP's anabolic and anti-inflammatory effects on tendons. While some clinical trials support these findings, others refute them. In this review, we discuss the effectiveness of PRP to treat tendon injuries with evidence presented in basic science studies and the potential reasons for the controversial results in clinical trials. Finally, we comment on the approaches that may be required to improve the efficacy of PRP treatment for tendinopathy.


Asunto(s)
Plasma Rico en Plaquetas , Tendinopatía/inmunología , Tendinopatía/metabolismo , Tendinopatía/terapia , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Inflamación/terapia , Tendinopatía/patología
7.
Adv Exp Med Biol ; 920: 53-62, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27535248

RESUMEN

Millions of people suffer from tendon injuries in both occupational and athletic settings. However, the restoration of normal structure and function to injured tendons still remains as one of the greatest challenges in orthopaedics and sports medicine. In recent years, a remarkable advancement in tendon research field has been the discovery of tendon stem/progenitor cells (TSCs). Unlike tenocytes, the predominant resident cell in tendons, TSCs have the ability to self-renew and multi-differentiate. Because of these distinct properties, TSCs may play a critical role in tendon physiology as well as pathology such as tendinopathy, which is a prevalent chronic tendon injury. Additionally, because TSCs are tendon-specific stem cells, they could potentially be used in tendon tissue engineering in vitro, and serve as a promising cell source for cell-based therapy to effectively repair or even regenerate injured tendons in clinical settings.


Asunto(s)
Biofisica , Trasplante de Células Madre , Células Madre/fisiología , Tendinopatía/patología , Tendones/fisiología , Animales , Fenómenos Biomecánicos , Humanos , Células Madre/citología , Tendinopatía/terapia , Tendones/citología , Resistencia a la Tracción , Ingeniería de Tejidos
8.
Acta Biomater ; 42: 136-146, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27329787

RESUMEN

UNLABELLED: Tissue-engineering approaches have a great potential to improve the treatment of tendon injuries that affect millions of people. The present study tested the hypothesis that introduction of a tendon derived stem/progenitor cell (TSC) sheet accelerates tendon healing and tendon regeneration in a rat model. TSC sheets were produced on temperature-responsive culture dishes. Then, they were grafted on unwounded Achilles tendons and at sites of a 3mm of Achilles tendon defect. At 2 and 4weeks after implantation tendons were examined by histology, immunohistochemistry, transmission electron microscopy (TEM) and mechanical testing. The results showed that the implanted TSC sheet remained stably attached on the tendon surface at 4 weeks after implantation. Moreover, in the tendon defect model, tendon defect area where TSC sheet was implanted was well regenerated and had better organized collagen fibers with elongated spindle shaped cells, compared to relatively disorganized collagen fibers and round shaped cells in the control group. TEM observations revealed longitudinally aligned collagen fibers and thick collagen fibrils in the TSC sheet implanted group. Finally, at 4weeks mechanical property of the TSC sheet implanted tendon had better ultimate load than the control. In conclusion, this study demonstrates the feasibility of implanting TSC sheets on tendons in vivo. Introduction of the cell sheets into a tendon defect significantly improved histological properties and collagen content at both 2 and 4 weeks after implantation, indicating that TSC sheets may effectively promote tendon remodeling in the early stages of tendon healing. STATEMENT OF SIGNIFICANCE: Tendon injury is a highly prevalent clinical problem that debilitates millions of people worldwide in both occupational and athletic settings. It also costs billions of healthcare dollars in treatment every year. In this study, we showed the feasibility of using tendon derived stem cell sheet to deliver biologically active tenogenic-constructs and promote tendon regeneration. This work has the potential to impact the orthopaedic surgery and sports medicine fields in the treatment of tendon injury.


Asunto(s)
Tendón Calcáneo/patología , Trasplante de Células Madre , Células Madre/citología , Traumatismos de los Tendones/patología , Traumatismos de los Tendones/terapia , Tendón Calcáneo/ultraestructura , Animales , Fenómenos Biomecánicos , Separación Celular , Modelos Animales de Enfermedad , Inmunohistoquímica , Implantes Experimentales , Masculino , Ratas , Reproducibilidad de los Resultados
9.
Oncotarget ; 7(8): 8498-512, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26885754

RESUMEN

The effect of exercise on wound healing in aging tendon was tested using a rat moderate treadmill running (MTR) model. The rats were divided into an MTR group that ran on a treadmill for 4 weeks and a control group that remained in cages. After MTR, a window defect was created in the patellar tendons of all rats and wound healing was analyzed. We found that MTR accelerated wound healing by promoting quicker closure of wounds, improving the organization of collagen fibers, and decreasing senescent cells in the wounded tendons when compared to the cage control. MTR also lowered vascularization, increased the numbers of tendon stem/progenitor cells (TSCs) and TSC proliferation than the control. Besides, MTR significantly increased the expression of stem cell markers, OCT-4 and Nanog, and tenocyte genes, Collagen I, Collagen III and tenomodulin, and down-regulated PPAR-γ, Collagen II and Runx-2 (non-tenocyte genes). These findings indicated that moderate exercise enhances healing of injuries in aging tendons through TSC based mechanisms, through which exercise regulates beneficial effects in tendons. This study reveals that appropriate exercise may be used in clinics to enhance tendon healing in aging patients.


Asunto(s)
Envejecimiento/fisiología , Biomarcadores/metabolismo , Ligamento Rotuliano/citología , Carrera , Células Madre/citología , Traumatismos de los Tendones/prevención & control , Cicatrización de Heridas , Animales , Diferenciación Celular , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Técnicas para Inmunoenzimas , Masculino , Ligamento Rotuliano/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Estrés Mecánico
10.
Stem Cell Res Ther ; 6: 173, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26373929

RESUMEN

INTRODUCTION: Platelet-rich plasma (PRP) is widely used to treat tendon injuries in clinics. These PRP preparations often contain white blood cells or leukocytes, and the precise cellular effects of leukocyte-rich PRP (L-PRP) on tendons are not well defined. Therefore, in this study, we determined the effects of L-PRP on tendon stem/progenitor cells (TSCs), which play a key role in tendon homeostasis and repair. METHODS: TSCs isolated from the patellar tendons of rabbits were treated with L-PRP or P-PRP (pure PRP without leukocytes) in vitro, followed by measuring cell proliferation, stem cell marker expression, inflammatory gene expression, and anabolic and catabolic protein expression by using immunostaining, quantitative real-time polymerase chain reaction, Western blot, and enzyme-linked immunosorbent assay, respectively. RESULTS: Cell proliferation was induced by both L-PRP and P-PRP in a dose-dependent manner with maximum proliferation at a 10 % PRP dose. Both PRP treatments also induced differentiation of TSCs into active tenocytes. Nevertheless, the two types of PRP largely differed in several effects exerted on TSCs. L-PRP induced predominantly catabolic and inflammatory changes in differentiated tenocytes; its treatment increased the expression of catabolic marker genes, matrix metalloproteinase-1 (MMP-1), MMP-13, interleukin-1beta (IL-1ß), IL-6 and tumor necrosis factor-alpha (TNF-α), and their respective protein expression and prostaglandin E2 (PGE 2) production. In contrast, P-PRP mainly induced anabolic changes; that is, P-PRP increased the gene expression of anabolic genes, alpha-smooth muscle actin (α-SMA), collagen types I and III. CONCLUSIONS: These findings indicate that, while both L-PRP and P-PRP appear to be "safe" in inducing TSC differentiation into active tenocytes, L-PRP may be detrimental to the healing of injured tendons because it induces catabolic and inflammatory effects on tendon cells and may prolong the effects in healing tendons. On the other hand, when P-PRP is used to treat acutely injured tendons, it may result in the formation of excessive scar tissue due to the strong potential of P-PRP to induce inordinate cellular anabolic effects.


Asunto(s)
Proliferación Celular , Leucocitos/metabolismo , Plasma Rico en Plaquetas , Trasplante de Células Madre/métodos , Células Madre/citología , Tendones/citología , Actinas/genética , Actinas/metabolismo , Animales , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Dinoprostona/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Conejos , Células Madre/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
11.
PLoS One ; 9(4): e93968, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24699667

RESUMEN

Wound healing requires the vasculature to re-establish itself from the severed ends; endothelial cells within capillaries must detach from neighboring cells before they can migrate into the nascent wound bed to initiate angiogenesis. The dissociation of these endothelial capillaries is driven partially by platelets' release of growth factors and cytokines, particularly the chemokine CXCL4/platelet factor-4 (PF4) that increases cell-cell de-adherence. As this retraction is partly mediated by increased transcellular contractility, the protein kinase c-δ/myosin light chain-2 (PKCδ/MLC-2) signaling axis becomes a candidate mechanism to drive endothelial dissociation. We hypothesize that PKCδ activation induces contractility through MLC-2 to promote dissociation of endothelial cords after exposure to platelet-released CXCL4 and VEGF. To investigate this mechanism of contractility, endothelial cells were allowed to form cords following CXCL4 addition to perpetuate cord dissociation. In this study, CXCL4-induced dissociation was reduced by a VEGFR inhibitor (sunitinib malate) and/or PKCδ inhibition. During combined CXCL4+VEGF treatment, increased contractility mediated by MLC-2 that is dependent on PKCδ regulation. As cellular force is transmitted to focal adhesions, zyxin, a focal adhesion protein that is mechano-responsive, was upregulated after PKCδ inhibition. This study suggests that growth factor regulation of PKCδ may be involved in CXCL4-mediated dissociation of endothelial cords.


Asunto(s)
Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Neovascularización Fisiológica/fisiología , Factor Plaquetario 4/farmacología , Proteína Quinasa C-delta/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Cicatrización de Heridas/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Humanos , Indoles/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Pirroles/farmacología , Sunitinib , Cicatrización de Heridas/efectos de los fármacos
12.
PLoS One ; 9(2): e87706, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24504456

RESUMEN

Prostaglandin E2 (PGE2) has been reported to exert different effects on tissues at low and high levels. In the present study, cell culture experiments were performed to determine the potential biphasic effects of PGE2 on human tendon stem/progenitor cells (hTSCs). After treatment with PGE2, hTSC proliferation, stemness, and differentiation were analyzed. We found that high concentrations of PGE2 (>1 ng/ml) decreased cell proliferation and induced non-tenocyte differentiation. However, at lower concentrations (<1 ng/ml), PGE2 markedly enhanced hTSC proliferation. The expression levels of stem cell marker genes, specifically SSEA-4 and Stro-1, were more extensive in hTSCs treated with low concentrations of PGE2 than in cells treated with high levels of PGE2. Moreover, high levels of PGE2 induced hTSCs to differentiate aberrantly into non-tenocytes, which was evident by the high levels of PPARγ, collagen type II, and osteocalcin expression in hTSCs treated with PGE2 at concentrations >1 ng/ml. The findings of this study reveal that PGE2 can exhibit biphasic effects on hTSCs, indicating that while high PGE2 concentrations may be detrimental to tendons, low levels of PGE2 may play a vital role in the maintenance of tendon homeostasis in vivo.


Asunto(s)
Dinoprostona/farmacología , Células Madre/citología , Células Madre/efectos de los fármacos , Tendones/citología , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Inmunofenotipificación , Ratas , Trasplante de Células Madre , Células Madre/metabolismo
13.
Ann Biomed Eng ; 41(10): 2109-19, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23549900

RESUMEN

Tendon stem cells (TSCs) may be used to effectively repair or regenerate injured tendons. However, the fates of TSCs once implanted in vivo remain unclear. This study was aimed to determine the feasibility of labeling TSCs with super-paramagnetic iron oxide (SPIO) nano-particles to track TSCs in vivo using MRI. Rabbit TSCs were labeled by incubation with 50 µg/mL SPIO. Labeling efficiency, cell viability, and proliferation were then measured, and the stemness of TSCs was tested by quantitative real time RT-PCR (qRT-PCR) and immunocytochemistry. We found that the labeling efficiency of TSCs reached as high as 98%, and that labeling at 50 µg/mL SPIO concentrations did not alter cell viability and cell proliferation compared to non-labeled control cells. Moreover, the expression levels of stem cell markers (Nucleostemin, Nanog, and Oct-4) did not change in SPIO-labeled TSCs compared to non-labeled cells. Both labeled and non-labeled cells also exhibited similar differentiation potential. Finally, labeled TSCs could be detected by MRI both in vitro and in vivo. Taken together, the findings of this study show that labeling TSCs with SPIO particles is a feasible approach to track TSCs in vivo by MRI, which offers a non-invasive method to monitor repair of injured tendons.


Asunto(s)
Compuestos Férricos/farmacología , Imagen por Resonancia Magnética , Nanopartículas de Magnetita , Trasplante de Células Madre , Células Madre , Tendones , Aloinjertos , Animales , Supervivencia Celular , Compuestos Férricos/química , Masculino , Conejos , Radiografía , Células Madre/diagnóstico por imagen , Células Madre/metabolismo , Traumatismos de los Tendones/diagnóstico por imagen , Traumatismos de los Tendones/metabolismo , Traumatismos de los Tendones/terapia , Tendones/diagnóstico por imagen , Tendones/metabolismo
14.
Cell Stress Chaperones ; 18(4): 397-404, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23292503

RESUMEN

Dupuytren's contracture (DC) is a fibroproliferative disorder of unknown etiology characterized by a scar-like contracture that develops in the palm and/or digits. We have previously reported that the eta subunit of the chaperonin containing T-complex polypeptide (CCT-eta) is increased in fibrotic wound healing, and is essential for the accumulation of α-smooth muscle actin (α-SMA) in fibroblasts. The purpose of this study was to determine if CCT-eta is similarly implicated in the aberrant fibrosis seen in DC and to investigate the role of CCT-eta in the behavior of myo/fibroblasts in DC. Fibroblasts were obtained from DC-affected palmar fascia, from adjacent phenotypically normal palmar fascia in the same DC patients (PF), and from non-DC palmar fascial tissues in patients undergoing carpal tunnel (CT) release. Inherent contractility in these three populations was examined using fibroblast-populated collagen lattices (FPCLs) and by cell traction force microscopy. Expression of CCT-eta and α-SMA protein was determined by Western blot. The effect of CCT-eta inhibition on the contractility of DC cells was determined by deploying an siRNA versus CCT-eta. DC cells were significantly more contractile than both matching palmar fascial (PF) cells and CT cells in both assays, with PF cells demonstrating an intermediate contractility in the FPCL assay. Whereas α-SMA protein was significantly increased only in DC cells compared to PF and CT cells, CCT-eta protein was significantly increased in both PF and DC cells compared to CT cells. siRNA-mediated depletion of CCT-eta inhibited the accumulation of both CCT-eta and α-SMA protein in DC cells, and also significantly decreased the contractility of treated DC cells. These observations suggest that increased expression of CCT-eta appears to be a marker for latent and active disease in these patients and to be essential for the increased contractility exhibited by these fibroblasts.


Asunto(s)
Chaperonina con TCP-1/metabolismo , Fibroblastos/fisiología , Actinas/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Chaperonina con TCP-1/antagonistas & inhibidores , Chaperonina con TCP-1/genética , Contractura de Dupuytren/metabolismo , Contractura de Dupuytren/patología , Fascia/citología , Fibroblastos/citología , Humanos , Contracción Muscular/fisiología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
15.
J Orthop Res ; 31(1): 105-10, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22886634

RESUMEN

Injection of Dexamethasone (Dex) is commonly used in clinics to treat tendon injury such as tendinopathy because of its anti-inflammatory capabilities. However, serious adverse effects have been reported as a result of Dex treatment, such as impaired tendon healing and tendon rupture. Using both in vitro and in vivo approaches, this study was to determine the effects of Dex treatment on the proliferation and differentiation of human tendon stem cells (hTSCs), which can directly impact tendon healing. We found that Dex treatment stimulated cell proliferation at lower concentrations (<1,000 nM), whereas a high concentration (1,000 nM) decreased cell proliferation. Moreover, at all concentrations used (5, 10, 100, and 1,000 nM), Dex treatment induced non-tenocyte differentiation of hTSCs, as evidenced by a change in cell shape, a nearly complete suppression of collagen type I expression, and an upregulation of non-tenocyte related genes (PPARγ and Sox-9), which was especially evident when higher concentrations (>10 nM) of Dex were used. Implantation of Dex-treated hTSCs for a short time (3 weeks) resulted in the extensive formation of fatty tissues, cartilage-like tissues, and bony tissues. These findings suggest that Dex treatment in clinics may cause a paradoxical effect on the injured tendons it is supposed to treat: by inducing non-tenocyte differentiation of hTSCs, Dex treatment depletes the stem cell pool and leads to the formation of non-tendinous tissues (e.g., fatty and cartilage-like tissues), which make tendon susceptible to rupture.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Dexametasona/farmacología , Ligamento Rotuliano/citología , Traumatismos de los Tendones/tratamiento farmacológico , Traumatismos de los Tendones/patología , Adulto , Células Madre Adultas/citología , Células Madre Adultas/trasplante , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Glucocorticoides/farmacología , Humanos , Masculino , Ligamento Rotuliano/efectos de los fármacos , Ratas , Ratas Desnudas , Trasplante Heterólogo , Cicatrización de Heridas/efectos de los fármacos , Adulto Joven
16.
J Orthop Res ; 30(1): 47-52, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21688312

RESUMEN

Tendon stem cells (TSCs) have been proposed to play a major role in the development of tendinopathy, which refers to pathological changes, such as calcification, in affected tendons. Using a human TSC (hTSC) culture model, this study investigated the effects of PGE(2) , an inflammatory mediator present in injured tendons, on hTSC proliferation and differentiation as well as the molecular mediator for such PGE(2) -induced effects. We found that PGE(2) treatment of hTSCs decreased cell proliferation and caused osteogenic differentiation of hTSCs in a dose-dependent manner. Also, PGE(2) treatment of hTSCs induced dose-dependent BMP-2 production in culture, and moreover, addition of BMP-2 to hTSC culture decreased cell proliferation and induced hTSC differentiation into osteoblasts. Finally, addition of BMP-2 antibodies to hTSC culture treated with PGE(2) nearly abolished PGE(2) effects on both cell proliferation and osteogenic differentiation. Taken together, the findings of this study showed that BMP-2 mediates PGE(2) -induced reduction of proliferation and osteogenic differentiation of hTSCs. We suggest that such a mechanism may be partially responsible for the formation of calcified tissues in tendinopathic tendons seen in clinical settings.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Dinoprostona/metabolismo , Osteogénesis/fisiología , Ligamento Rotuliano/citología , Ligamento Rotuliano/metabolismo , Células Madre/citología , Adulto , Calcinosis/metabolismo , Calcinosis/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Células Cultivadas , Dinoprostona/farmacología , Humanos , Persona de Mediana Edad , Osteogénesis/efectos de los fármacos , Células Madre/metabolismo , Tendinopatía/metabolismo , Tendinopatía/patología
17.
PLoS One ; 5(4): e10063, 2010 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-20442790

RESUMEN

Integumentary wounds in mammalian fetuses heal without scar; this scarless wound healing is intrinsic to fetal tissues and is notable for absence of the contraction seen in postnatal (adult) wounds. The precise molecular signals determining the scarless phenotype remain unclear. We have previously reported that the eta subunit of the chaperonin containing T-complex polypeptide (CCT-eta) is specifically reduced in healing fetal wounds in a rabbit model. In this study, we examine the role of CCT-eta in fibroblast motility and contractility, properties essential to wound healing and scar formation. We demonstrate that CCT-eta (but not CCT-beta) is underexpressed in fetal fibroblasts compared to adult fibroblasts. An in vitro wound healing assay demonstrated that adult fibroblasts showed increased cell migration in response to epidermal growth factor (EGF) and platelet derived growth factor (PDGF) stimulation, whereas fetal fibroblasts were unresponsive. Downregulation of CCT-eta in adult fibroblasts with short inhibitory RNA (siRNA) reduced cellular motility, both basal and growth factor-induced; in contrast, siRNA against CCT-beta had no such effect. Adult fibroblasts were more inherently contractile than fetal fibroblasts by cellular traction force microscopy; this contractility was increased by treatment with EGF and PDGF. CCT-eta siRNA inhibited the PDGF-induction of adult fibroblast contractility, whereas CCT-beta siRNA had no such effect. In each of these instances, the effect of downregulating CCT-eta was to modulate the behavior of adult fibroblasts so as to more closely approximate the characteristics of fetal fibroblasts. We next examined the effect of CCT-eta modulation on alpha-smooth muscle actin (alpha-SMA) expression, a gene product well known to play a critical role in adult wound healing. Fetal fibroblasts were found to constitutively express less alpha-SMA than adult cells. Reduction of CCT-eta with siRNA had minimal effect on cellular beta-actin but markedly decreased alpha-SMA; in contrast, reduction of CCT-beta had minimal effect on either actin isoform. Direct inhibition of alpha-SMA with siRNA reduced both basal and growth factor-induced fibroblast motility. These results indicate that CCT-eta is a specific regulator of fibroblast motility and contractility and may be a key determinant of the scarless wound healing phenotype by means of its specific regulation of alpha-SMA expression.


Asunto(s)
Movimiento Celular , Chaperonina con TCP-1/fisiología , Fibroblastos/citología , Cicatrización de Heridas , Factores de Edad , Animales , Tamaño de la Célula , Chaperonina con TCP-1/análisis , Chaperonina con TCP-1/biosíntesis , Cicatriz , Feto , Fibroblastos/química , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/farmacología , Subunidades de Proteína , ARN Interferente Pequeño/farmacología , Conejos
18.
BMC Musculoskelet Disord ; 11: 10, 2010 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-20082706

RESUMEN

BACKGROUND: Tendons are traditionally thought to consist of tenocytes only, the resident cells of tendons; however, a recent study has demonstrated that human and mouse tendons also contain stem cells, referred to as tendon stem/progenitor cells (TSCs). However, the differential properties of TSCs and tenocytes remain largely undefined. This study aims to characterize the properties of these tendon cells derived from rabbits. METHODS: TSCs and tenocytes were isolated from patellar and Achilles tendons of rabbits. The differentiation potential and cell marker expression of the two types of cells were examined using histochemical, immunohistochemical, and qRT-PCR analysis as well as in vivo implantation. In addition, morphology, colony formation, and proliferation of TSCs and tenocytes were also compared. RESULTS: It was found that TSCs were able to differentiate into adipocytes, chondrocytes, and osteocytes in vitro, and form tendon-like, cartilage-like, and bone-like tissues in vivo. In contrast, tenocytes had little such differentiation potential. Moreover, TSCs expressed the stem cell markers Oct-4, SSEA-4, and nucleostemin, whereas tenocytes expressed none of these markers. Morphologically, TSCs possessed smaller cell bodies and larger nuclei than ordinary tenocytes and had cobblestone-like morphology in confluent culture whereas tenocytes were highly elongated. TSCs also proliferated more quickly than tenocytes in culture. Additionally, TSCs from patellar tendons formed more numerous and larger colonies and proliferated more rapidly than TSCs from Achilles tendons. CONCLUSIONS: TSCs exhibit distinct properties compared to tenocytes, including differences in cell marker expression, proliferative and differentiation potential, and cell morphology in culture. Future research should investigate the mechanobiology of TSCs and explore the possibility of using TSCs to more effectively repair or regenerate injured tendons.


Asunto(s)
Diferenciación Celular/fisiología , Células del Tejido Conectivo/fisiología , Regeneración/fisiología , Trasplante de Células Madre/métodos , Células Madre/fisiología , Tendones/fisiología , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Proteínas Portadoras/genética , Linaje de la Célula/fisiología , Forma de la Célula/fisiología , Células Cultivadas , Células del Tejido Conectivo/citología , Femenino , Proteínas de Unión al GTP , Proteínas Nucleares/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Mensajero/metabolismo , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Antígenos Embrionarios Específico de Estadio/genética , Células Madre/citología , Traumatismos de los Tendones/patología , Traumatismos de los Tendones/fisiopatología , Traumatismos de los Tendones/terapia , Tendones/citología
19.
J Biol Chem ; 285(7): 4995-5002, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20007974

RESUMEN

Toll-like receptor-4 (TLR4) is the receptor for bacterial lipopolysaccharide, yet it may also respond to a variety of endogenous molecules. Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in newborn infants and is characterized by intestinal mucosal destruction and impaired enterocyte migration due to increased TLR4 signaling on enterocytes. The endogenous ligands for TLR4 that lead to impaired enterocyte migration remain unknown. High mobility group box-1 (HMGB1) is a DNA-binding protein that is released from injured cells during inflammation. We thus hypothesize that extracellular HMGB1 inhibits enterocyte migration via activation of TLR4 and sought to define the pathways involved. We now demonstrate that murine and human NEC are associated with increased intestinal HMGB1 expression, that serum HMGB1 is increased in murine NEC, and that HMGB1 inhibits enterocyte migration in vitro and in vivo in a TLR4-dependent manner. This finding was unique to enterocytes as HMGB1 enhanced migration of inflammatory cells in vitro and in vivo. In seeking to understand the mechanisms involved, TLR4-dependent HMGB1 signaling increased RhoA activation in enterocytes, increased phosphorylation of focal adhesion kinase, and increased phosphorylation of cofilin, resulting in increased stress fibers and focal adhesions. Using single cell force traction microscopy, the net effect of HMGB1 signaling was a TLR4-dependent increase in cell force adhesion, accounting for the impaired enterocyte migration. These findings demonstrate a novel pathway by which TLR4 activation by HMGB1 delays mucosal repair and suggest a novel potential therapeutic target in the amelioration of intestinal inflammatory diseases like NEC.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Enterocitos/citología , Proteína HMGB1/metabolismo , Proteína HMGB1/farmacología , Mucosa Intestinal/metabolismo , Receptor Toll-Like 4/metabolismo , Actinas/metabolismo , Animales , Línea Celular , Movimiento Celular/genética , Quimiotaxis/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Enterocolitis Necrotizante/metabolismo , Enterocitos/efectos de los fármacos , Citometría de Flujo , Humanos , Técnicas In Vitro , Recién Nacido , Mucosa Intestinal/citología , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Receptor Toll-Like 4/genética , Proteína de Unión al GTP rhoA/metabolismo
20.
Int Rev Cell Mol Biol ; 271: 301-46, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19081546

RESUMEN

Mechanical forces, including gravity, tension, compression, hydrostatic pressure, and fluid shear stress, play a vital role in human physiology and pathology. They particularly influence extracellular matrix (ECM) gene expression, ECM protein synthesis, and production of inflammatory mediators of many load-sensitive adult cells such as fibroblasts, chondrocytes, smooth muscle cells, and endothelial cells. Furthermore, the mechanical forces generated by cells themselves, known as cell traction forces (CTFs), also influence many biological processes such as wound healing, angiogenesis, and metastasis. Thus, the quantitative characterization of CTFs by qualities such as magnitude and distribution is useful for understanding physiological and pathological events at the tissue and organ levels. Recently, the effects of mechanical loads on embryonic and adult stem cells in terms of self-renewal, differentiation, and matrix protein expression have been investigated. While it seems certain that mechanical loads applied to stem cells regulate their self-renewal and induce controlled cell lineage differentiation, the detailed molecular signaling mechanisms responsible for these mechano-effects remain to be elucidated. Challenges in the fields of both adult- and stem-cell mechanobiology include devising novel experimental and theoretical methodologies to examine mechano-responses more closely to various forms of mechanical forces and mechanotransduction mechanisms of these cells in a more physiologically accurate setting. Such novel methodologies will lead to better understanding of various pathological diseases, their management, and translational applications in the ever expanding field of tissue engineering.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , Mecanotransducción Celular/fisiología , Animales , Humanos , Estrés Mecánico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA