Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Ultrasonics ; 104: 106110, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32146383

RESUMEN

Surface mechanical attrition treatment (SMAT) of metallic biomaterials has gained significant importance due to its ability to develop nano structure in the surface region. In the present study, the microstructural changes and corrosion behavior of the commercially pure titanium (cp-Ti), following different durations of ultrasonic shot peening (USSP) has been investigated. cp-Ti was shot peened for different durations from 0 to 120 s and the treated samples were examined for microstructural changes in the surface region, cell viability and corrosion behavior. Cell viability was considerably increased after USSP for 60-120 s, exhibiting maximum for the 90 s of USSP. The passivation tendency was also improved with peening duration up to 90 s, however, it declined for longer duration of USSP. The beneficial effects of USSP may be attributed to nano structuring in the surface region and development of higher positive potentials at the USSP treated surface. Transmission Electron Microscope (TEM) examination of the USSPed surface revealed dislocation entanglement and substructure. Also, higher surface volta potential was observed over the USSPed sample exhibiting better cell proliferation. The present work is corollary to previous work of the group and mainly discusses the role of USSP duration, as a process parameter, on the cell viability and corrosion resistance of cp-Ti.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Nanoestructuras/química , Titanio/química , Ultrasonido/métodos , Proliferación Celular , Supervivencia Celular , Corrosión , Técnicas Electroquímicas , Humanos , Ensayo de Materiales , Microscopía de Fuerza Atómica , Microscopía Electrónica , Propiedades de Superficie , Difracción de Rayos X
2.
J Immunol ; 200(2): 595-606, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29203513

RESUMEN

Bone remodeling comprises balanced activities between osteoclasts and osteoblasts, which is regulated by various factors, including hormones and cytokines. We previously reported that IL-3 inhibits osteoclast differentiation and pathological bone loss. IL-3 also enhances osteoblast differentiation and bone formation from mesenchymal stem cells. However, the role of IL-3 in regulation of osteoblast-osteoclast interactions and underlying mechanisms is not yet delineated. In this study, we investigated the role of IL-3 on the regulation of osteoblast-specific molecules, receptor activator of NF-κB ligand (RANKL), and osteoprotegerin (OPG) that modulate bone homeostasis. We found that IL-3 increases RANKL expression at both the transcriptional and translational levels, and it showed no effect on OPG expression in calvarial osteoblasts. The increased RANKL expression by IL-3 induces mononuclear osteoclasts; however, it does not induce multinuclear osteoclasts. Interestingly, IL-3 decreases soluble RANKL by reducing ectodomain shedding of membrane RANKL through downregulation of metalloproteases mainly a disintegrin and metalloproteinase (ADAM)10, ADAM17, ADAM19, and MMP3. Moreover, IL-3 increases membrane RANKL by activating the JAK2/STAT5 pathway. Furthermore, IL-3 enhances RANKL expression in mesenchymal stem cells of wild-type mice but not in STAT5a knockout mice. Interestingly, IL-3 restores RANKL expression in adult mice by enhancing bone-specific RANKL and decreasing serum RANKL. Furthermore, IL-3 increases the serum OPG level in adult mice. Thus, our results reveal, to our knowledge for the first time, that IL-3 differentially regulates two functional forms of RANKL through metalloproteases and the JAK2/STAT5 pathway, and it helps in restoring the decreased RANKL/OPG ratio in adult mice. Notably, our studies indicate the novel role of IL-3 in regulating bone homeostasis in important skeletal disorders.


Asunto(s)
Interleucina-3/metabolismo , Janus Quinasa 2/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Osteoblastos/metabolismo , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Células Cultivadas , Técnicas de Cocultivo , Expresión Génica , Interleucina-3/farmacología , Ratones , Ratones Transgénicos , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligando RANK/sangre , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/genética , Transducción de Señal/efectos de los fármacos
3.
Int J Nanomedicine ; 12: 6437-6459, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28919746

RESUMEN

Bacterial cellulose (BC) is a naturally occurring nanofibrous biomaterial which exhibits unique physical properties and is amenable to chemical modifications. To explore whether this versatile material can be used in the treatment of osteochondral defects (OCD), we developed and characterized novel BC-based nanocomposite scaffolds, for example, BC-hydroxyapatite (BC-HA) and BC-glycosaminoglycans (BC-GAG) that mimic bone and cartilage, respectively. In vitro biocompatibility of BC-HA and BC-GAG scaffolds was established using osteosarcoma cells, human articular chondrocytes, and human adipose-derived mesenchymal stem cells. On subcutaneous implantation, the scaffolds allowed tissue ingrowth and induced no adverse immunological reactions suggesting excellent in vivo biocompatibility. Implantation of acellular bilayered scaffolds in OCD created in rat knees induced progressive regeneration of cartilage tissue, deposition of extracellular matrix, and regeneration of subchondral bone by the host cells. The results of micro-CT revealed that bone mineral density and ratio of bone volume to tissue volume were significantly higher in animals receiving bilayered scaffold as compared to the control animals. To the best of our knowledge, this study proves for the first time, the functional performance of acellular BC-based bilayered scaffolds. Thus, this strategy has great potential for clinical translation and can be used in repair of OCD.


Asunto(s)
Materiales Biocompatibles/química , Condrocitos/citología , Nanocompuestos/química , Andamios del Tejido , Acetobacteraceae/química , Animales , Regeneración Ósea/fisiología , Cartílago/citología , Celulosa/química , Condrocitos/química , Modelos Animales de Enfermedad , Durapatita/química , Glicosaminoglicanos/química , Humanos , Ensayo de Materiales , Células Madre Mesenquimatosas/citología , Ratas , Ratas Wistar , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
4.
Stem Cell Res Ther ; 8(1): 168, 2017 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-28705238

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) represent an important source for cell therapy in regenerative medicine. MSCs have shown promising results for repair of damaged tissues in various degenerative diseases in animal models and also in human clinical trials. However, little is known about the factors that could enhance the migration and tissue-specific engraftment of exogenously infused MSCs for successful regenerative cell therapy. Previously, we have reported that interleukin-3 (IL-3) prevents bone and cartilage damage in animal models of rheumatoid arthritis and osteoarthritis. Also, IL-3 promotes the differentiation of human MSCs into functional osteoblasts and increases their in-vivo bone regenerative potential in immunocompromised mice. However, the role of IL-3 in migration of MSCs is not yet known. In the present study, we investigated the role of IL-3 in migration of human MSCs under both in-vitro and in-vivo conditions. METHODS: MSCs isolated from human bone marrow, adipose and gingival tissues were used for in-vitro cell migration, motility and wound healing assays in the presence or absence of IL-3. The effect of IL-3 preconditioning on expression of chemokine receptors and integrins was examined by flow cytometry and real-time PCR. The in-vivo migration of IL-3-preconditioned MSCs was investigated using a subcutaneous matrigel-releasing stromal cell-derived factor-1 alpha (SDF-1α) model in immunocompromised mice. RESULTS: We observed that human MSCs isolated from all three sources express IL-3 receptor-α (IL-3Rα) both at gene and protein levels. IL-3 significantly enhances in-vitro migration, motility and wound healing abilities of MSCs. Moreover, IL-3 preconditioning upregulates expression of chemokine (C-X-C motif) receptor 4 (CXCR4) on MSCs, which leads to increased migration of cells towards SDF-1α. Furthermore, CXCR4 antagonist AMD3100 decreases the migration of IL-3-treated MSCs towards SDF-1α. Importantly, IL-3 also induces in-vivo migration of MSCs towards subcutaneously implanted matrigel-releasing-SDF-1α in immunocompromised mice. CONCLUSIONS: The present study demonstrates for the first time that IL-3 has an important role in enhancing the migration of human MSCs through regulation of the CXCR4/SDF-1α axis. These findings suggest a potential role of IL-3 in improving the efficacy of MSCs in regenerative cell therapy.


Asunto(s)
Movimiento Celular , Regulación de la Expresión Génica , Interleucina-3/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptores CXCR4/biosíntesis , Animales , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...