Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Aging (Albany NY) ; 13(3): 3605-3617, 2021 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-33461165

RESUMEN

Bony injuries lead to compromised skeletal functional ability which further increase in aging population due to decreased bone mineral density. Therefore, we aimed to investigate the therapeutic potential of platelet-derived biomaterials (PDB) against bone injury. Specifically, we assessed the impact of PDB on osteo-inductive characteristics and migration of mouse embryonic fibroblasts (MEFs). Osteogenic lineage, matrix mineralization and cell migration were determined by gene markers (RUNX2, OPN and OCN), alizarin Red S staining, and migration markers (FAK, pFAK and Src) and EMT markers, respectively. The therapeutic impact of TGF-ß1, a key component of PDB, was confirmed by employing inhibitor of TGF-ß receptor I (Ti). Molecular imaging-based in vivo cellular migration in mice was determined by establishing bone injury at right femurs. Results showed that PDB markedly increased expression of osteogenic markers, matrix mineralization, migration and EMT markers, revealing higher osteogenic and migratory potential of PDB-treated MEFs. In vivo cell migration was manifested by expression of migratory factors, SDF-1 and CXCR4. Compared to control, PDB-treated mice exhibited higher bone density and volume. Ti treatment inhibited both migration and osteogenic potential of MEFs, affirming impact of TGF-ß1. Collectively, our study clearly indicated PDB-rescued bone injury through enhancing migratory potential of MEFs and osteogenesis.


Asunto(s)
Materiales Biocompatibles , Plaquetas/metabolismo , Regeneración Ósea , Movimiento Celular , Fémur/lesiones , Fibroblastos/metabolismo , Osteogénesis , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Densidad Ósea , Calcificación Fisiológica , Linaje de la Célula , Quimiocina CXCL12 , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Transición Epitelial-Mesenquimal , Fémur/metabolismo , Fémur/patología , Fibroblastos/citología , Quinasa 1 de Adhesión Focal , Técnicas In Vitro , Ratones , Células 3T3 NIH , Osteocalcina/genética , Osteopontina/genética , Receptores CXCR4 , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Familia-src Quinasas
3.
Stem Cells Dev ; 25(23): 1833-1842, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27596042

RESUMEN

Adipose-derived stem cells (ADSCs) are multipotent cells that have attracted much recent attention and emerged as therapeutic approaches in several medical fields. Although current knowledge of the biological impacts of ADSCs in cancer research is greatly improved, the underlying effects of ADSCs in tumor development remain controversial and cause the safety concerns in clinical utilization. Hence, we isolated primary ADSCs from the abdominal fat of mice and conducted interaction of ADSCs with Lewis lung carcinoma cells in culture and in mice to investigate the impacts of ADSCs on tumor development. Cytokine array and neutralizing antibody were further utilized to identify the key regulator and downstream signaling pathway. In this study, we demonstrated that ADSCs enhance the malignant characteristics of LLC1 cells, including cell growth ability and especially cancer stem cell property. ADSCs were then identified to promote tumor formation and growth in mice. We further determined that ADSC interaction with LLC1 cells stimulates increased secretion of interleukin-6 mainly from ADSCs, which then act in a paracrine manner on LLC1 cells to enhance their malignant characteristics. Interleukin-6 was also identified to regulate genes related to cell proliferation and cancer stem cell, as well as to activate JAK2/STAT3, a predominant interleukin-6-activated pathway, in LLC1 cells. Collectively, we demonstrated that ADSCs play a pro-malignant role in tumor development of Lewis lung carcinoma cells by particularly promoting cancer stem cell property through interleukin-6 paracrine circuit, which is important for safety considerations regarding the clinical application of ADSCs.


Asunto(s)
Tejido Adiposo/citología , Carcinogénesis/patología , Carcinoma Pulmonar de Lewis/patología , Interleucina-6/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Neoplásicas/patología , Comunicación Paracrina , Animales , Carcinogénesis/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Proliferación Celular , Femenino , Janus Quinasa 2/metabolismo , Ratones Endogámicos C57BL , Células Madre Neoplásicas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Células del Estroma/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-27143981

RESUMEN

Antrodia camphorata has previously demonstrated the efficacy in treating cancer and anti-inflammation. In this study, we are the first to evaluate Antrodia camphorata alcohol extract (ACAE) for osteoporosis recovery in vitro with preosteoblast cells (MC3T3-E1) and in vivo with an osteoporosis mouse model established in our previous studies, ovariectomized senescence accelerated mice (OVX-SAMP8). Our results demonstrated that ACAE treatment was slightly cytotoxic to preosteoblast at 25 µg/mL, by which the osteogenic gene expression (RUNX2, OPN, and OCN) was significantly upregulated with an increased ratio of OPG to RANKL, indicating maintenance of the bone matrix through inhibition of osteoclastic pathway. Additionally, evaluation by Alizarin Red S staining showed increased mineralization in ACAE-treated preosteoblasts. For in vivo study, our results indicated that ACAE inhibits bone loss and significantly increases percentage bone volume, trabecular bone number, and bone mineral density in OVX-SAMP8 mice treated with ACAE. Collectively, in vitro and in vivo results showed that ACAE could promote osteogenesis and prevent bone loss and should be considered an evidence-based complementary and alternative medicine for osteoporosis therapy through the maintenance of bone health.

5.
Oncotarget ; 6(10): 7713-26, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25797257

RESUMEN

Adipose-derived stem cells (ADSCs) are multipotent cells that have attracted much recent attention. Here, we show that ADSCs enhance sphere formation and in vivo tumor initiation of breast and colon cancer cells. In co-culture, ADSCs induced several stem cell markers in cancer cells. ADSCs also accelerated tumor growth. Interaction of ADSCs and cancer cells stimulated secretion of interlukin-6 in ADSCs, which in turn acted in a paracrine manner on cancer cells to enhance their malignant properties. Interleukin-6 regulated stem cell-related genes and activated JAK2/STAT3 in cancer cells. We suggest that ADSCs may enhance tumor initiation and promotion.


Asunto(s)
Interleucina-6/biosíntesis , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Adipocitos/metabolismo , Adipocitos/patología , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C
6.
Biomaterials ; 35(37): 9767-9776, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25239042

RESUMEN

Aging is related to loss of functional stem cell accompanying loss of tissue and organ regeneration potentials. Previously, we demonstrated that the life span of ovariectomy-senescence accelerated mice (OVX-SAMP8) was significantly prolonged and similar to that of the congenic senescence-resistant strain of mice after platelet rich plasma (PRP)/embryonic fibroblast transplantation. The aim of this study is to investigate the potential of PRP for recovering cellular potential from senescence and then delaying animal aging. We first examined whether stem cells would be senescent in aged mice compared to young mice. Primary adipose derived stem cells (ADSCs) and bone marrow derived stem cells (BMSCs) were harvested from young and aged mice, and found that cell senescence was strongly correlated to animal aging. Subsequently, we demonstrated that PRP could recover cell potential from senescence, such as promote cell growth (cell proliferation and colony formation), increase osteogenesis, decrease adipogenesis, restore cell senescence related markers and resist the oxidative stress in stem cells from aged mice. The results also showed that PRP treatment in aged mice could delay mice aging as indicated by survival, body weight and aging phenotypes (behavior and gross morphology) in term of recovering the cellular potential of their stem cells compared to the results on aged control mice. In conclusion these findings showed that PRP has potential to delay aging through the recovery of stem cell senescence and could be used as an alternative medicine for tissue regeneration and future rejuvenation.


Asunto(s)
Envejecimiento , Senescencia Celular , Plasma Rico en Plaquetas/metabolismo , Células Madre/citología , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Células Cultivadas , Femenino , Humanos , Ratones , Osteogénesis , Células Madre/metabolismo
7.
Oncotarget ; 5(19): 9514-29, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25237908

RESUMEN

Several reports suggest that malignant cells generate phenotypic diversity through fusion with various types of stromal cells within the tumor microenvironment. Mesenchymal stem cell (MSC) is one of the critical components in the tumor microenvironment and a promising fusogenic candidate, but the underlying functions of MSC fusion with malignant cell have not been fully examined. Here, we demonstrate that MSCs fuse spontaneously with lung cancer cells, and the latter is reprogrammed to slow growth and stem-like state. Transcriptome profiles reveal that lung cancer cells are reprogrammed to a more benign state upon MSC fusion. We further identified FOXF1 as a reprogramming mediator that contributes not only to the reprogramming toward stemness but also to the p21-regulated growth suppression in fusion progeny. Collectively, MSC fusion does not enhance the intrinsic malignancy of lung cancer cells. The anti-malignant effects of MSC fusion-induced reprogramming on lung cancer cells were accomplished by complementation of tumorigenic defects, including restoration of p21 function and normal terminal differentiation pathways as well as up-regulation of FOXF1, a putative tumor suppressor. Such fusion process raises the therapeutic potential that MSC fusion can be utilized to reverse cellular phenotypes in cancer.


Asunto(s)
Transdiferenciación Celular/genética , Reprogramación Celular , Factores de Transcripción Forkhead/genética , Neoplasias Pulmonares/patología , Células Madre Mesenquimatosas/citología , Animales , Fusión Celular , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Factores de Transcripción Forkhead/biosíntesis , Humanos , Neoplasias Pulmonares/genética , Ratones , Ratones SCID , Interferencia de ARN , ARN Interferente Pequeño , Esferoides Celulares , Células Tumorales Cultivadas , Microambiente Tumoral
8.
Biomaterials ; 35(36): 9599-607, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25176059

RESUMEN

Osteoarthritis (OA) is a common disease associated with tissue inflammation, physical disability and imbalanced homeostasis in cartilage. For advanced treatments, biological approaches are currently focused on tissue regeneration and anti-inflammation. This study was undertaken to evaluate the therapeutic efficacies of hyaluronic acid (HA) and platelet-rich plasma (PRP) (HA+PRP) on OA. Articular chondrocytes were obtained from five OA patients. The optimal HA and PRP concentrations were evaluated by MTT assay. The expressions of chondrogenic and inflammatory genes were analyzed by RT-PCR. Signaling pathway was examined by immunoblotting and the expressions of OA pathology-related chemokines and cytokines was demonstrated by real-time PCR-based SuperArray. The therapeutic efficacies of HA+PRP were then demonstrated in 3D arthritic neo-cartilage and ACLT-OA model. Here we showed that HA+PRP could greatly retrieve pro-inflammatory cytokines-reduced articular chondrocytes proliferation and chondrogenic phenotypes, the mechanism of which involve the sequential activation of specific receptors CD44 and TGF-ßRII, downstream mediators Smad2/3 and Erk1/2, and the chondrogenic transcription factor SOX9. The real-time PCR-based SuperArray results also indicated that OA pathology-related chemokines and cytokines could be efficiently suppressed by HA+PRP. Moreover, the cartilaginous ECM could be retrieved from inflammation-induced degradation by HA+PRP in both 2D monolayer and 3D neo-cartilage model. Finally, the intra-articular injection of HA+PRP could strongly rescue the meniscus tear and cartilage breakdown and then decrease OA-related immune cells. The combination of HA+PRP can synergistically promote cartilage regeneration and inhibit OA inflammation. This study might offer an advanced and alternative OA treatment based on detailed regenerative mechanisms.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Condrocitos/citología , Ácido Hialurónico/uso terapéutico , Inflamación/terapia , Osteoartritis/terapia , Plasma Rico en Plaquetas , Adyuvantes Inmunológicos/administración & dosificación , Animales , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/inmunología , Condrocitos/patología , Humanos , Ácido Hialurónico/administración & dosificación , Inflamación/inmunología , Inflamación/patología , Inyecciones Intraarticulares , Ratones , Osteoartritis/inmunología , Osteoartritis/patología , Plasma Rico en Plaquetas/citología
9.
Artículo en Inglés | MEDLINE | ID: mdl-24069046

RESUMEN

The aim of this study is to examine the therapeutic potential of deep sea water (DSW) on osteoporosis. Previously, we have established the ovariectomized senescence-accelerated mice (OVX-SAMP8) and demonstrated strong recovery of osteoporosis by stem cell and platelet-rich plasma (PRP). Deep sea water at hardness (HD) 1000 showed significant increase in proliferation of osteoblastic cell (MC3T3) by MTT assay. For in vivo animal study, bone mineral density (BMD) was strongly enhanced followed by the significantly increased trabecular numbers through micro-CT examination after a 4-month deep sea water treatment, and biochemistry analysis showed that serum alkaline phosphatase (ALP) activity was decreased. For stage-specific osteogenesis, bone marrow-derived stromal cells (BMSCs) were harvested and examined. Deep sea water-treated BMSCs showed stronger osteogenic differentiation such as BMP2, RUNX2, OPN, and OCN, and enhanced colony forming abilities, compared to the control group. Interestingly, most untreated OVX-SAMP8 mice died around 10 months; however, approximately 57% of DSW-treated groups lived up to 16.6 months, a life expectancy similar to the previously reported life expectancy for SAMR1 24 months. The results demonstrated the regenerative potentials of deep sea water on osteogenesis, showing that deep sea water could potentially be applied in osteoporosis therapy as a complementary and alternative medicine (CAM).

10.
Biomaterials ; 33(26): 6105-12, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22698723

RESUMEN

Adipose-derived stem cells (ADSCs) have been shown to be pluoripotent and explored for their usage in tissue engineering. Previously, we have established a cell-based approach comprised of platelet-enriched plasma and osteo-progenitor cells for treating osteoporosis in an ovariectomized-senescence-accelerated mice (OVX-SAMP8) model. In the present study, we intend to explore the feasibility of using ADSCs as a cell-based therapeutic approach for treating osteoporosis, and to examine the effects of aging on the pluoripotency of ADSCs and the efficiency of bone formation both in vitro and in vivo. Flow cytometry was used to characterize ADSCs isolated from young and aged female SAMP8 mice and showed that the highly positive expression of surface markers such as CD44 and CD105 and negative for CD34 and CD45. Therefore, to compare the aging effects on the growth kinetics and differentiation potential of young and aged ADSCs, we found that there was a significant decline in both the proliferation rate (approximately 13.3%) and osteo-differentiation potential in aged ADSC. Subsequently, young and aged ADSCs were transplanted into the bone marrow of osteoporotic mice (OVX-SAMP8) to evaluate their bone formation ability. ADSC transplants were shown effective in restoring bone mineral density in the right/left knees, femurs and spine, 4 months post-transplantation; mice which received young ADSC transplants showed significantly higher bone regeneration (an average of 24.3% of improved BMD) over those received aged ADSCs. In conclusion, these findings showed that aging impedes osteoporosis-ameliorating potential of ADSC by diminishing osteogenic signal, and that ADSC could be used as a potential cell-based therapy for osteoporosis.


Asunto(s)
Tejido Adiposo/citología , Envejecimiento/fisiología , Osteoporosis/metabolismo , Células Madre/citología , Células Madre/metabolismo , Envejecimiento/genética , Animales , Antígenos CD34/metabolismo , Regeneración Ósea/genética , Regeneración Ósea/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Endoglina , Femenino , Receptores de Hialuranos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Ratones , Osteoporosis/genética , Ovariectomía
11.
Biomaterials ; 32(28): 6773-80, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21700330

RESUMEN

The aim of this study was to develop a new diagnostic and therapeutic approach for the treatment of osteoporosis. Previously, we demonstrated that intraosseous transplantation of platelet-rich plasma (PRP) treated-osteoblast-like cells into ovariectomized senescence-accelerated mice (OVX-SAMP8) prevented the development of osteoporosis. In continuation, we aimed to explore the complex etiology of osteoporosis using this platform. An inverse relationship between bone marrow adipogenesis and osteogenesis has been suggested in the development of osteoporosis but the underlying mechanisms remain poorly described. To address these issues, we used PRP to inhibit adipocyte differentiation by promoting osteoblastic differentiation in adipocytes. In addition, a positive correlation between an increase in bone marrow adipocytes and bone loss was established. We assessed this relationship using an osteoporotic animal disease model which consisted of young (for prevention) and old (for treatment) OVX-SAMP8 mice. This animal model demonstrated that PRP treatment mainly exerted its action via promoting bone regeneration but also appeared to suppress adipogenesis within the marrow. The findings and methodology of this study could potentially be applied in the prevention and treatment of osteoporosis.


Asunto(s)
Adipogénesis/fisiología , Transfusión Sanguínea , Regeneración Ósea/fisiología , Osteogénesis/fisiología , Osteoporosis/terapia , Plasma Rico en Plaquetas , Adipocitos/citología , Adipocitos/fisiología , Animales , Densidad Ósea , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Línea Celular , Transdiferenciación Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Osteoblastos/citología , Osteoblastos/fisiología , Osteoporosis/diagnóstico
12.
Biomaterials ; 30(29): 5523-33, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19646749

RESUMEN

An ex vivo degenerative intervertebral disc (IVD) organ culture system was established for the screening of disc regeneration agents. Its application was demonstrated by a stem cell and growth factor-based therapeutic approach for the amelioration of IVD. An ex vivo culture system using chymopapain to partially digest nucleus proposus tissue was established to mimic human IVD degeneration. This system was then used for the evaluation of different therapeutic regimens including: mesenchymal stem cell derived from eGFP-transgenic porcine (MSC-GFP), platelet-rich plasma (PRP) and MSC-GFP/PRP combined treatment, and confirmed in in vivo animal model. Chondrogenic-specific gene products including Col II and aggrecan were found upregulated and chondrogenic matrix deposition increased, as evident by sustained fluorescent signals over 4 weeks, in the MSC-GFP implanted group. Previously, we demonstrated in vitro stage-specific chondrogenesis of MSC by chondrocytic commitment. These same molecules upregulated for chondrogenesis were also observed in MSC-GFP group. PRP that has been shown to promote nucleus pulposus (NP) regeneration also resulted in significant increased levels of mRNA involved in chondrogenesis and matrices accumulation. The ex vivo IVD regeneration results were repeated and supported by in vivo porcine degenerative system. Moreover, the disc height index (DHI) was significantly increased in both in vivo MSC-GFP and PRP regeneration groups. Unexpectedly, the MSC-GFP/PRP combined therapy demonstrated an inclination towards osteogenesis in ex vivo system. The ex vivo degenerative IVD culture system described in this study could serve as an alternative and more accessible model over large animal model. This system also provides a high-throughput platform for screening therapeutic agents for IVD regeneration.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Condrogénesis/fisiología , Desplazamiento del Disco Intervertebral/patología , Desplazamiento del Disco Intervertebral/cirugía , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Técnicas de Cultivo de Órganos/métodos , Regeneración/fisiología , Animales , Células Cultivadas , Células Madre Mesenquimatosas/fisiología , Porcinos
13.
J Nucl Med ; 50(5): 765-73, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19372470

RESUMEN

UNLABELLED: The aim of this study was to develop a cell-based bone-regeneration approach evaluated by molecular imaging and immunohistochemistry. METHODS: Genetically modified NIH3T3 embryonic fibroblasts carrying enhanced green fluorescent protein (NIH3T3-G) were predifferentiated into osteoblastlike cells using platelet-rich plasma (PRP) medium, followed by intraosseous transplantation into ovariectomized senescence-accelerated mouse prone substrain 8 (OVX-SAMP8 mice). RESULTS: PRP-conditioned NIH3T3-G (PRP/NIH3T3-G) engraftment prevented the development of osteoporosis. Molecular imaging and immunohistochemistry demonstrated the migration of NIH3T3-G cells from the implantation site throughout the skeleton. In situ analyses revealed coexpression of osteopontin and green fluorescent protein in the newly formed bone tissue, demonstrating that the transplant restored the bone trabecular architecture and mineral density in treated OVX-SAMP8 mice. Interestingly, the life span of OVX-SAMP8 mice receiving PRP/NIH3T3-G transplantation was significantly prolonged and similar to that of the congenic senescence-resistant strain of mice. CONCLUSION: This unique and yet simple approach could potentially be applied to the treatment of senile postmenopausal osteoporosis and perhaps inborn genetic syndromes associated with accelerated aging, such as Hutchinson-Gilford progeria syndrome, and for the prolongation of life expectancy in general.


Asunto(s)
Células Madre Embrionarias/trasplante , Fibroblastos/trasplante , Osteogénesis/fisiología , Osteoporosis/patología , Osteoporosis/cirugía , Plasma Rico en Plaquetas/fisiología , Animales , Línea Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias/patología , Fibroblastos/patología , Humanos , Ratones , Células 3T3 NIH , Ovariectomía
14.
J Cell Physiol ; 209(3): 744-54, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16955489

RESUMEN

Human intervertebral disc (IVD) degeneration often initiated from the human nucleus pulposus (hNP) with aging leading to IVD destruction and extracellular matrix (ECM) depletion. Previously, we have successfully employed transforming growth factor-beta1 (TGF-beta1) to promote chondrogenesis of mesenchymal progenitor cells (MPCs) and immortalized human mesenchymal stem cells. In this study, we examine the role of TGF-beta1 in platelet-rich plasma (PRP) on disc regeneration, including proliferation, redifferentiation, and the reconstitution of tissue-engineered NP. hNP cells were isolated from volunteers with different ages and cultured in the presence of PRP. We found that the most effective concentration for hNP proliferation was 1 ng/ml TGF-beta1 in PRP, which was further applied in the following experiments. hNP cell proliferation in all age groups were increased time-dependently by PRP and cell morphologies showed aggregation. The mRNA of Sox9, type II collagen, and aggrecan were all significantly upregulated by PRP through RT-PCR. Glycosaminoglycan (GAG) accumulation reached the highest value at day 7 and continued to day 9 culture. PRP promoted NP regeneration via the Smad pathway was also determined and highly activated p-Smad2/3 at 30 min and continuously sustained to 120 min. Immunostaining of type II collagen indicates that PRP participates in chondrogenesis of tissue-engineered NP with collagen scaffolds. We concluded that growth factors in PRP can effectively react as a growth factor cocktail to induce hNP proliferation and differentiation, and also promote tissue-engineered NP formation. These findings are the first to demonstrate that PRP might be a therapeutic candidate for prevention of disc degeneration.


Asunto(s)
Plaquetas/metabolismo , Condrogénesis/fisiología , Disco Intervertebral , Plasma/metabolismo , Ingeniería de Tejidos , Factor de Crecimiento Transformador beta1/metabolismo , Adulto , Anciano , Animales , Apoptosis , Proliferación Celular , Medios de Cultivo Condicionados/química , Matriz Extracelular/metabolismo , Humanos , Disco Intervertebral/citología , Disco Intervertebral/fisiología , Persona de Mediana Edad , Plasma/citología , Proteoglicanos/metabolismo , ARN Mensajero/metabolismo , Regeneración , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA