Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Int J Biol Macromol ; 256(Pt 1): 128059, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37989428

RESUMEN

This study aimed to functionalize a novel porous PLGA (Poly lactic-co-glycolic acid) composite scaffold in combination with nano­calcium sulphate (nCS) and/or fucoidan (FU) to induce osteogenic differentiation of human bone marrow stromal cells. The composite scaffolds (PLGA-nCS-FU, PLGA-nCS or PLGA-FU) were fabricated and subjected to characterization using Fourier-transform infrared spectroscopy (FTIR), X-ray powder diffraction (XRD), Scanning electron microscopy (SEM) and Energy Dispersive X-Ray (EDX). The biocompatibility and osteogenic induction potential of scaffolds on seeded human bone marrow derived mesenchymal stromal cells (hBMSCs) were studied using cell attachment and alamar blue cell viability and alkaline phosphatase (ALP), osteocalcin and osteogenic gene expression, respectively. The composition of different groups was reflected in FTIR, XRD and EDX. The SEM micrographs revealed a difference in the surface of the scaffold before and after FU addition. The confocal imaging and SEM micrographs confirmed the attachment of cells onto all three composite scaffolds. However, the AB assay indicated a significant increase (p < 0.05) in cell viability/proliferation seeded on PLGA-nCS-FU on day 21 and 28 as compared with other combinations. A 2-fold significant increase (p < 0.05) in ALP and OC secretion of seeded hBMSCs onto PLGA-nCS-FU was observed when compared with other combinations. A significant increase in RUNX2, OPN, COL-I and ALP genes were observed in the cells seeded on PLGA-nCS-FU on day 14 and 28 as compared with day 0. In conclusion, the incorporation of both Fucoidan and Nano­calcium sulphate with PLGA showed a promising improvement in the osteogenic potential of hBMSCs. Therefore, PLGA-nCS-FU could be the ideal candidate for subsequent pre-clinical studies to develop a successful bone substitute to repair critical bone defects.


Asunto(s)
Glicolatos , Células Madre Mesenquimatosas , Polisacáridos , Ingeniería de Tejidos , Humanos , Ingeniería de Tejidos/métodos , Osteogénesis , Andamios del Tejido/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Glicoles , Médula Ósea , Diferenciación Celular , Sulfatos , Células de la Médula Ósea
2.
Int J Mol Sci ; 24(23)2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38069268

RESUMEN

The effective and long-term treatment of cartilage defects is an unmet need among patients worldwide. In the past, several synthetic and natural biomaterials have been designed to support functional articular cartilage formation. However, they have mostly failed to enhance the terminal stage of chondrogenic differentiation, leading to scar tissue formation after the operation. Growth factors substantially regulate cartilage regeneration by acting on receptors to trigger intracellular signaling and cell recruitment for tissue regeneration. In this study, we investigated the effect of recombinant insulin-like growth factor 1 (rIGF-1), loaded in fibrin microbeads (FibIGF1), on cartilage regeneration. rIGF-1-loaded fibrin microbeads were injected into full-thickness cartilage defects in the knees of goats. The stability, integration, and quality of tissue repair were evaluated at 1 and 6 months by gross morphology, histology, and collagen type II staining. The in vivo results showed that compared to plain fibrin samples, particularly at 6 months, FibIGF1 improved the functional cartilage formation, confirmed through gross morphology, histology, and collagen type II immunostaining. FibIGF1 could be a promising candidate for cartilage repair in the clinic.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Humanos , Animales , Colágeno Tipo II/metabolismo , Fibrina/metabolismo , Cabras , Cartílago Articular/metabolismo , Enfermedades de los Cartílagos/metabolismo , Condrocitos
3.
J Diabetes Metab Disord ; 22(2): 1039-1052, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37975135

RESUMEN

Objectives: Diabetes mellitus (DM) is a complex metabolic disease that results from impaired insulin secreting pancreatic ß-cells or insulin resistance. Although available medications help control the disease, patients suffer from its complications. Therefore, finding effective therapeutic approaches to treat DM is a priority. Adipose Derived Stem Cells (ADSCs) based therapy is a promising strategy in various regenerative medicine applications, but its systematic translational use is still somewhat out of reach. This review is aimed at clarifying achievements as well as challenges facing the application of ADSCs for the treatment of DM, with a special focus on the mechanisms involved. Methods: Literature searches were carried out on "Scopus", "PubMed" and "Google Scholar" up to September 2022 to find relevant articles in the English language for the scope of this review. Results: Recent evidence showed a significant role of ADSC therapies in DM by ameliorating insulin resistance and hyperglycemia, regulating hepatic glucose metabolism, promoting ß cell function and regeneration, and functioning as a gene delivery tool. In addition, ADSCs could improve diabetic wound healing by promoting collagen deposition, inhibiting inflammation, and enhancing angiogenesis. Conclusion: Overall, this literature review revealed the great clinical implications of ADSCs for translating into the clinical setting for the treatment of diabetes. However, further large-scale and controlled studies are needed to overcome challenges and confirm the safety and optimal therapeutic scheme before daily clinical application. Supplementary Information: The online version contains supplementary material available at 10.1007/s40200-023-01280-8.

4.
Tissue Cell ; 82: 102075, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37004269

RESUMEN

Tendon injuries account up to 50% of all musculoskeletal problems and remains a challenge to treat owing to the poor intrinsic reparative ability of tendon tissues. The natural course of tendon healing is very slow and often leads to fibrosis and disorganized tissues with inferior biomechanical properties. Mesenchymal stem cells (MSC) therapy is a promising alternative strategy to augment tendon repair due to its proliferative and multilineage differentiation potential. Hypoxic conditioning of MSC have been shown to enhance their tenogenic differentiation capacity. However, the mechanistic pathway by which this is achieved is yet to be fully defined. A key factor involved in this pathway is hypoxia-inducible factor-1-alpha (HIF-1α). This review aims to discuss the principal mechanism underlying the enhancement of MSC tenogenic differentiation by hypoxic conditioning, particularly the central role of HIF-1α in mediating activation of tenogenic pathways in the MSC. We focus on the interaction between HIF-1α with fibroblast growth factor-2 (FGF-2) and transforming growth factor-beta 1 (TGF-ß1) in regulating MSC tenogenic differentiation pathways in hypoxic conditions. Strategies to promote stabilization of HIF-1α either through direct manipulation of oxygen tension or the use of hypoxia mimicking agents are therefore beneficial in increasing the efficacy of MSC therapy for tendon repair.


Asunto(s)
Células Madre Mesenquimatosas , Traumatismos de los Tendones , Humanos , Tendones/metabolismo , Diferenciación Celular , Traumatismos de los Tendones/terapia , Traumatismos de los Tendones/metabolismo , Hipoxia/metabolismo
5.
Stem Cells Int ; 2023: 4907230, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36756494

RESUMEN

The present study was conducted to determine whether adipose derived mesenchymal stromal cells (AD-MSCs) or bone marrow derived-MSCs (BM-MSCs) would provide superior tenogenic expressions when subjected to cyclical tensile loading. The results for this would indicate the best choice of MSCs source to be used for cell-based tendon repair strategies. Both AD-MSCs and BM-MSCs were obtained from ten adult donors (N = 10) and cultured in vitro. At passaged-2, cells from both groups were subjected to cyclical stretching at 1 Hz and 8% of strain. Cellular morphology, orientation, proliferation rate, protein, and gene expression levels were compared at 0, 24, and 48 hours of stretching. In both groups, mechanical stretching results in similar morphological changes, and the redirection of cell alignment is perpendicular to the direction of stretching. Loading at 8% strain did not significantly increase proliferation rates but caused an increase in total collagen expression and tenogenic gene expression levels. In both groups, these levels demonstrated no significant differences suggesting that in a similar loading environment, both cell types possess similar tenogenic potential. In conclusion, AD-MSCs and BM-MSCs both demonstrate similar tenogenic phenotypic and gene expression levels when subjected to cyclic tensile loading at 1 Hz and 8% strain, thus, suggesting that the use of either cell source may be suitable for tendon repair.

6.
J Basic Clin Physiol Pharmacol ; 34(2): 177-185, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34182614

RESUMEN

OBJECTIVES: Primary Osteoarthritis (OA) is a disease of progressive joints degeneration due to idiopathic causes. Recent evidence showed a positive relationship between OA and metabolic syndrome. This pilot study aimed to assess the baseline level of pro and anti-inflammatory cytokines in OA patients with or without Diabetic Mellitus (DM) and assess the effect of hydrogen peroxide (H2O2) in cytokine production. METHODS: Patients with primary hip and knee OA were recruited, and 3 mL of bone marrow was harvested during joint replacement surgery. Bone marrow stromal cells (BMSC) was isolated and cultured in a culture flask for three passages. Later experiment was then sub-cultured in a well plate labeled as the control group and H2O2 (0.1 mM) treated group. ProcartaPlex® Multiplex Immunoassay was performed to measure cytokine levels produced by the BMSC at 0 h, as well as 72 h. RESULTS: Cytokines such as tumor necrosis factor-alpha, interleukin (IL)-6, IL-8, and IL-1ß generally exhibited higher cytokine levels in subjects with DM than in nonDM subjects at 0 and 72 h. For IL-17, its expression was similar in nonDM and DM groups at 0 and 72 h. Cytokine IL-10 showed no significant difference in both the groups while DM and nonDM groups treated with H2O2 showed decreased IL-4 levels compared to control groups at 72 h. Bone marrow cells from DM-OA are more vulnerable to chemical insult and are associated with higher levels of proinflammatory cytokines production and lower IL-4 level production. CONCLUSIONS: This study provides a clue that management of OA with co-morbidity like DM needs future studies.

7.
Tissue Eng Part C Methods ; 28(10): 501-510, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36082992

RESUMEN

Transforming growth factor-beta 1 (TGF-ß1) has been reported to promote chondrogenic differentiation and proliferation in the multipotent stromal cell (MSCs), and the transforming growth factor-beta 3 (TGF-ß3) tends to be exclusively in promoting cell differentiation alone. The objective of this study was to determine the effect of TGF-ß1 and -ß3 on the MSCs chondrogenic differentiation on the poly (vinyl alcohol)-chitosan-poly (ethylene glycol) (PVA-NOCC-PEG) scaffold, compared with that of monolayer and pellet cultures. In this study, P2 rabbit bone marrow-derived MSCs were seeded either on the untreated six-well plate (for monolayer culture) or onto the PVA-NOCC-PEG scaffold or cultured as a pellet culture. The cultures were maintained in a chemically defined serum-free medium supplemented with 10 ng/mL of either TGF-ß1 or TGF-ß3. Cell viability assay, biochemical assay, and real-time polymerase chain reaction were performed to determine the net effect of cell proliferation and chondrogenic differentiation of each of the growth factors. The results showed that the PVA-NOCC-PEG scaffold enhanced MSCs cell proliferation from day 12 to 30 (p < 0.05); however, no significant differences were observed in the cell proliferation between the cultures supplemented with or without TGF-ß1 and TGF-ß3 (p > 0.05). In terms of chondrogenic differentiation, the PVA-NOCC-PEG scaffold augmented the GAGs secretion in MSCs and the mRNA expression levels of Sox9, Col2a1, Acan, and Comp were elevated (p < 0.05). However, there was no significant difference between both the TGF-ß1 and TGF-ß3-treated groups (p > 0.05). In conclusion, TGF-ß1 and TGF-ß3 enhanced the chondrogenic differentiation of MSCs seeded on the PVA-NOCC-PEG scaffold; however, there was no significant difference between the effect of TGF-ß1 and TGF-ß3. Impact statement Transforming growth factor-beta (TGF-ß) superfamily members is a key requirement for the in vitro chondrogenic differentiation of mesenchymal stem cells (MSCs). In this study, the effects of TGF-ß1 and -ß3 on MSC chondrogenic differentiation and proliferation on a novel three-dimensional scaffold, the poly(vinyl alcohol)-chitosan-poly(ethylene glycol) (PVA-NOCC-PEG) scaffold, was evaluated. In this study, the results showed both TGF-ß1 and TGF-ß3 can enhance the chondrogenic differentiation of MSCs seeded on the PVA-NOCC-PEG scaffold.


Asunto(s)
Quitosano , Células Madre Mesenquimatosas , Animales , Conejos , Factor de Crecimiento Transformador beta3/metabolismo , Factor de Crecimiento Transformador beta3/farmacología , Alcohol Polivinílico/farmacología , Alcohol Polivinílico/metabolismo , Quitosano/farmacología , Quitosano/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Polietilenglicoles/farmacología , Condrogénesis , Diferenciación Celular , Factor de Crecimiento Transformador beta/farmacología , Factores de Crecimiento Transformadores/metabolismo , Factores de Crecimiento Transformadores/farmacología , Células Cultivadas
8.
Mar Drugs ; 20(9)2022 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-36135749

RESUMEN

Our previous study evidenced that the 3D CORAGRAF loaded with PLGA microsphere constitutes PDGF-BB can support cell attachment and proliferation and can induce an osteogenic commitment of mesenchymal stromal cells in the in vitro condition. However, how this construct can perform in pathophysiological conditions in terms of repairing critical bone defects is yet to be understood. A study was therefore conducted to investigate the regeneration potential of calvaria critical-size defects using CORAGRAF + PLGA with PDGF-BB + mesenchymal stromal cells (MSCs) in a rat model. A 5 mm critical bone defect was created on calvaria of 40 male Sprague-Dawley rats. CORAGRAF incorporated either with or without PDGF-BB and seeded with rat bone-marrow-derived MSCs was implanted at the defect region. The bone regeneration potential of implanted constructs was assessed using micro-CT imaging and histological staining in weeks 4 and 8. The micro-CT images indicated a significant closure of defects in the cranial bone of the rats treated with 3D CORAGRAF + PLGA with PDGF-BB + MSCs on week 4 and 8 post-implantation. This finding, further supported with the histology outcome where the rat cranial defect treated with CORAGRAF + PLGA with PDGF-BB + MSCs indicated neo-bony ingrowth with organized and mature bone-like morphology as compared with other groups. The previous in vitro results substantiated with our pre-clinical findings demonstrate that the combination of CORAGRAF + PLGA with PDGF-BB + MSCs could be an ideal construct to support bone regeneration in critical bone defects. Hence, this construct can be further investigated for its safety and efficacy in large animal models, or it can be skipped to human trial prior for commercialization.


Asunto(s)
Células Madre Mesenquimatosas , Animales , Becaplermina , Regeneración Ósea , Humanos , Masculino , Microesferas , Osteogénesis , Ratas , Ratas Sprague-Dawley , Cráneo/diagnóstico por imagen , Cráneo/patología , Cráneo/cirugía
9.
Cartilage ; 13(2_suppl): 1322S-1333S, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-31569963

RESUMEN

OBJECTIVE: Advances in research have shown that the subchondral bone plays an important role in the propagation of cartilage loss and progression of osteoarthritis (OA), but whether the subchondral bone changes precede or lead to articular cartilage loss remains debatable. In order to elucidate the subchondral bone and cartilage changes that occur in early OA, an experiment using anterior cruciate ligament transection (ACLT) induced posttraumatic OA model of the rat knee was conducted. DESIGN: Forty-two Sprague Dawley rats were divided into 2 groups: the ACLT group and the nonoperated control group. Surgery was conducted on the ACLT group, and subsequently rats from both groups were sacrificed at 1, 2, and 3 weeks postsurgery. Subchondral bone was evaluated using a high-resolution peripheral quantitative computed tomography scanner, while cartilage was histologically evaluated and scored. RESULTS: A significant reduction in the subchondral trabecular bone thickness and spacing was found as early as 1 week postsurgery in ACLT rats compared with the nonoperated control. This was subsequently followed by a reduction in bone mineral density and bone fractional volume at week 2, and finally a decrease in the trabecular number at week 3. These changes occurred together with cartilage degeneration as reflected by an increasing Mankin score over all 3 weeks. CONCLUSIONS: Significant changes in subchondral bone occur very early in OA concurrent with surface articular cartilage degenerative change suggest that factors affecting bone remodeling and resorption together with cartilage matrix degradation occur very early in the disease.


Asunto(s)
Cartílago Articular , Osteoartritis , Animales , Ligamento Cruzado Anterior/patología , Huesos/patología , Cartílago Articular/diagnóstico por imagen , Cartílago Articular/patología , Osteoartritis/diagnóstico por imagen , Osteoartritis/patología , Ratas , Ratas Sprague-Dawley
10.
PeerJ ; 8: e8740, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32587790

RESUMEN

BACKGROUND: Type 2 diabetes mellitus (T2DM) had been reported to be associated with tendinopathy. However, the underlying mechanisms of diabetic tendinopathy still remain largely to be discovered. The purpose of this study was to develop insulin resistance (IR) model on primary human tenocytes (hTeno) culture with tumour necrosis factor-alpha (TNF-α) treatment to study tenocytes homeostasis as an implication for diabetic tendinopathy. METHODS: hTenowere isolated from human hamstring tendon. Presence of insulin receptor beta (INSR-ß) on normal tendon tissues and the hTeno monolayer culture were analyzed by immunofluorescence staining. The presence of Glucose Transporter Type 1 (GLUT1) and Glucose Transporter Type 4 (GLUT4) on the hTeno monolayer culture were also analyzed by immunofluorescence staining. Primary hTeno were treated with 0.008, 0.08, 0.8 and 8.0 µM of TNF-α, with and without insulin supplement. Outcome measures include 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino]-2-deoxy-d-glucose (2-NBDG) assay to determine the glucose uptake activity; colourimetric total collagen assay to quantify the total collagen expression levels; COL-I ELISA assay to measure the COL-I expression levels and real-time qPCR to analyze the mRNA gene expressions levels of Scleraxis (SCX), Mohawk (MKX), type I collagen (COL1A1), type III collagen (COL3A1), matrix metalloproteinases (MMP)-9 and MMP-13 in hTeno when treated with TNF-α. Apoptosis assay for hTeno induced with TNF-α was conducted using Annexin-V FITC flow cytometry analysis. RESULTS: Immunofluorescence imaging showed the presence of INSR-ß on the hTeno in the human Achilles tendon tissues and in the hTeno in monolayer culture. GLUT1 and GLUT4 were both positively expressed in the hTeno. TNF-α significantly reduced the insulin-mediated 2-NBDG uptake in all the tested concentrations, especially at 0.008 µM. Total collagen expression levels and COL-I expression levels in hTeno were also significantly reduced in hTeno treated with 0.008 µM of TNF-α. The SCX, MKX and COL1A1 mRNA expression levels were significantly downregulated in all TNF-α treated hTeno, whereas the COL3A1, MMP-9 and MMP-13 were significantly upregulated in the TNF-α treated cells. TNF-α progressively increased the apoptotic cells at 48 and 72 h. CONCLUSION: At 0.008 µM of TNF-α, an IR condition was induced in hTeno, supported with the significant reduction in glucose uptake, as well as significantly reduced total collagen, specifically COL-I expression levels, downregulation of candidate tenogenic markers genes (SCX and MKX), and upregulation of ECM catabolic genes (MMP-9 and MMP-13). Development of novel IR model in hTeno provides an insight on how tendon homeostasis could be affected and can be used as a tool for further discovering the effects on downstream molecular pathways, as the implication for diabetic tendinopathy.

11.
Int J Med Sci ; 17(4): 457-470, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32174776

RESUMEN

Oxidative stress has been linked to senescence and tumorigenesis via modulation of the cell cycle. Using a hydrogen peroxide (H2O2)-induced oxidative stress-induced premature senescence (OSIPS) model previously reported by our group, this study aimed to investigate the effects of oxidative stress on microRNA (miRNA) expression in relation to the G1-to-S-phase (G1/S) transition of the cell cycle and cell proliferation. On global miRNA analysis of the OSIPS cells, twelve significantly up- or down-regulated miRNAs were identified, the target genes of which are frequently associated with cancers. Four down-regulated miR-17 family miRNAs are predicted to target key pro- and anti-proliferative proteins of the p21/cyclin D-dependent kinase (CDK)/E2F1 pathway to modulate G1/S transition. Two miR-17 miRNAs, miR-20-5p and miR-106-5p, were confirmed to be rapidly and stably down-regulated under oxidative stress. While H2O2 treatment hampered G1/S transition and suppressed DNA synthesis, miR-20b-5p/miR-106a-5p over-expression rescued cells from growth arrest in promoting G1/S transition and DNA synthesis. Direct miR-20b-5p/miR-106a-5p regulation of p21, CCND1 and E2F1 was demonstrated by an inverse expression relationship in miRNA mimic-transfected cells. However, under oxidative stress, E2F1 expression was down-regulated, consistent with hampered G1/S transition and suppressed DNA synthesis and cell proliferation. To explain the observed E2F1 down-regulation under oxidative stress, a scheme is proposed which includes miR-20b-5p/miR-106a-5p-dependent regulation, miRNA-E2F1 autoregulatory feedback and E2F1 response to repair oxidative stress-induced DNA damages. The oxidative stress-modulated expression of miR-17 miRNAs and E2F1 may be used to develop strategies to retard or reverse MSC senescence in culture, or senescence in general.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , MicroARNs/metabolismo , Estrés Oxidativo/fisiología , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular , Factor de Transcripción E2F1/genética , Humanos , Peróxido de Hidrógeno/farmacología , MicroARNs/genética , Estrés Oxidativo/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Mater Sci Eng C Mater Biol Appl ; 107: 110304, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31761210

RESUMEN

Surgical site infection associated with surgical instruments has always been a factor in delaying post-operative recovery of patients. The evolution in surface modification of surgical instruments can be a potential choice to overcome the nosocomial infection mainly caused by bacterial populations such as Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli. A study was, therefore, conducted characterising the morphology, hydrophobicity, adhesion strength, phase, Nano-hardness, surface chemistry, antimicrobial and biocompatibility of SS 316L steel deposited with a Nano-composite layer of Silver (Ag) and Tantalum oxide (Ta2O5) using physical vapour deposition magnetron sputtering. The adhesion strength of Ag/AgTa2O5 coating on SS 316L and treated at 250-850 °C of thermal treatment was evaluated using micro-scratch. The Ag/Ag-Ta2O5-400 °C was shown a 154% improvement in adhesion strength on SS 316L when compared with as-sputtered layer or Ag/Ag-Ta2O5-250, 550, 700 and 850 °C. The FESEM, XPS, and XRD indicated the segregation of Ag on the surface of SS 316L after the crystallization. Wettability and Nano-indentation tests demonstrated an increase in hydrophobicity (77.3 ±â€¯0.3°) and Nano-hardness (1.12 ±â€¯0.43 GPa) when compared with as-sputtered layer, after the 400 °C of thermal treatment. The antibacterial performance on Ag/Ag-Ta2O5-400 °C indicated a significant zone of inhibition to Staphylococcus aureus (A-axis: 16.33 ±â€¯0.58 mm; B-axis: 25.67 ±â€¯0.58 mm, p < 0.01) and Escherichia coli (A-axis: 16.33 ±â€¯1.15 mm; B-axis: 26.00 ±â€¯0.00 mm, p < 0.01) when compared with SS 316L or Ag/Ag-Ta2O5-700 °C, which showed no inhibition. The biocompatibility tests on Ag/Ag-Ta2O5-400 °C demonstrated an excellent in cell attachment, F-actin protein expression and proliferation/viability of bone marrow derived mesenchymal stromal on day 14 when compared with uncoated or Ag/Ag-Ta2O5-700 °C. This study shows that the Ag segregation process, hydrophobicity, adhesion strength, crystallization, and hardness progressively improved after the annealing up to 400 °C.


Asunto(s)
Antibacterianos/química , Materiales Biocompatibles/química , Nanocompuestos/química , Acero Inoxidable/química , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Calor , Ensayo de Materiales , Óxidos/química , Plata/química , Tantalio/química
13.
Stem Cells Int ; 2019: 5142518, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30956670

RESUMEN

We have determined the protective effects of Thymus serpyllum (TS) extract and nanoparticle-loaded TS on hydrogen peroxide-induced cell death of mesenchymal stromal cells (MSCs) in vitro. Gas chromatography-mass spectroscopy confirmed the spectrum of active components in the extract. Out of the three different extracts, the hexane extract showed significant free radical scavenging activity. Treatment of MSCs with H2O2 (hydrogen peroxide) significantly increased intracellular cell death; however, pretreatment with TS extract and nanoparticle-loaded TS (200 µg/ml) suppressed H2O2-induced elevation of Cyt-c and MMP13 and increased the survival rates of MSCs. H2O2-induced (0.1 mM) changes in cytokines were attenuated in the extract and nanoparticles by pretreatment and cotreatment at two time points (p < 0.05). H2O2 increased cell apoptosis. In contrast, treatment with nanoparticle-loaded TS suppressed the percentage of apoptosis considerably (p < 0.05). Therefore, TS may be considered as a potential candidate for enhancing the effectiveness of MSC transplantation in cell therapy.

14.
Stem Cells Int ; 2019: 9723025, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30918524

RESUMEN

The present study was conducted to establish the amount of mechanical strain (uniaxial cyclic stretching) required to provide optimal tenogenic differentiation expression in human mesenchymal stromal cells (hMSCs) in vitro, in view of its potential application for tendon maintenance and regeneration. Methods. In the present study, hMSCs were subjected to 1 Hz uniaxial cyclic stretching for 6, 24, 48, and 72 hours; and were compared to unstretched cells. Changes in cell morphology were observed under light and atomic force microscopy. The tenogenic, osteogenic, adipogenic, and chondrogenic differentiation potential of hMSCs were evaluated using biochemical assays, extracellular matrix expressions, and selected mesenchyme gene expression markers; and were compared to primary tenocytes. Results. Cells subjected to loading displayed cytoskeletal coarsening, longer actin stress fiber, and higher cell stiffness as early as 6 hours. At 8% and 12% strains, an increase in collagen I, collagen III, fibronectin, and N-cadherin production was observed. Tenogenic gene expressions were highly expressed (p < 0.05) at 8% (highest) and 12%, both comparable to tenocytes. In contrast, the osteoblastic, chondrogenic, and adipogenic marker genes appeared to be downregulated. Conclusion. Our study suggests that mechanical loading at 8% strain and 1 Hz provides exclusive tenogenic differentiation; and produced comparable protein and gene expression to primary tenocytes.

15.
PLoS One ; 14(3): e0214212, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30917166

RESUMEN

It has been demonstrated that nanocrystalline forsterite powder synthesised using urea as a fuel in sol-gel combustion method had produced a pure forsterite (FU) and possessed superior bioactive characteristics such as bone apatite formation and antibacterial properties. In the present study, 3D-scaffold was fabricated using nanocrystalline forsterite powder in polymer sponge method. The FU scaffold was used in investigating the physicochemical, biomechanics, cell attachment, in vitro biocompatibility and osteogenic differentiation properties. For physicochemical characterisation, Fourier-transform infrared spectroscopy (FTIR), Energy dispersive X-ray (EDX), X-ray diffraction (XRD), Raman spectroscopy, X-ray photoemission spectrometer (XPS) and Brunauer-Emmett-Teller (BET) were used. FTIR, EDX, XRD peaks and Raman spectroscopy demonstrated correlating to FU. The XPS confirmed the surface chemistry associating to FU. The BET revealed FU scaffold surface area of 12.67 m2/g and total pore size of 0.03 cm3/g. Compressive strength of the FU scaffold was found to be 27.18 ± 13.4 MPa. The human bone marrow derived mesenchymal stromal cells (hBMSCs) characterisation prior to perform seeding on FU scaffold verified the stromal cell phenotypic and lineage commitments. SEM, confocal images and presto blue viability assay suggested good cell attachment and proliferation of hBMSCs on FU scaffold and comparable to a commercial bone substitutes (cBS). Osteogenic proteins and gene expression from day 7 onward indicated FU scaffold had a significant osteogenic potential (p<0.05), when compared with day 1 as well as between FU and cBS. These findings suggest that FU scaffold has a greater potential for use in orthopaedic and/or orthodontic applications.


Asunto(s)
Células de la Médula Ósea/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Nanopartículas/química , Osteogénesis/efectos de los fármacos , Compuestos de Silicona , Anciano , Apatitas/metabolismo , Células de la Médula Ósea/citología , Fuerza Compresiva , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Compuestos de Silicona/síntesis química , Compuestos de Silicona/química , Compuestos de Silicona/farmacología
16.
J Physiol Biochem ; 75(2): 153-162, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30796627

RESUMEN

The high glucose concentration is able to disturb chondrocyte homeostasis and contribute to OA pathogenesis. This study was designed to investigate the protective effects of atorvastatin (ATO) on high glucose (HG)-mediated oxidative stress and mitochondrial apoptosis in C28I2 human chondrocytes. The protective effect of ATO (0.01 and 0.1 µM) on HG (75 mM)-induced oxidative stress and apoptosis was evaluated in C28I2 cells. The effects of ATO on HG-induced intracellular ROS production and lipid peroxidation were detected and the protein expression levels of Bax, Bcl-2, caspase-3, total and phosphorylated JNK and P38 MAPKs were analyzed by Western blotting. The mRNA expression levels of antioxidant enzymes including heme oxygenase-1, NAD(P)H quinine oxidoreductase, glutathione S-transferase-P1, catalase, superoxide dismutase-1, glutathione peroxidase-1, -3, -4 were evaluated by reverse transcription-polymerase chain reaction. Pretreatment with ATO remarkably increased the gene expression levels of antioxidant enzymes and reduced HG-induced elevation of ROS, lipid peroxidation, Bax/Bcl-2 ratio, caspase-3 activation, and JNK and P38 phosphorylation. Atorvastatin could considerably reduce HG-induced oxidative stress and mitochondrial apoptosis through increasing the expression of antioxidant enzymes. Atorvastatin may be considered as a promising agent to prevent high glucose-induced cartilage degradation in OA patients.


Asunto(s)
Apoptosis/efectos de los fármacos , Atorvastatina/farmacología , Glucosa/toxicidad , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Humanos , Peroxidación de Lípido/efectos de los fármacos , MAP Quinasa Quinasa 4/metabolismo , Oxidorreductasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Platelets ; 30(1): 66-74, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29090639

RESUMEN

Platelet-rich concentrate (PRC), used in conjunction with other chondroinductive growth factors, have been shown to induce chondrogenesis of human mesenchymal stromal cells (hMSC) in pellet culture. However, pellet culture systems promote cell hypertrophy and the presence of other chondroinductive growth factors in the culture media used in previous studies obscures accurate determination of the effect of platelet itself in inducing chondrogenic differentiation. Hence, this study aimed to investigate the effect of PRC alone in enhancing the chondrogenic differentiation potential of human mesenchymal stromal cells (hMSC) encapsulated in three-dimensional alginate constructs. Cells encapsulated in alginate were cultured in serum-free medium supplemented with only 15% PRC. Scanning electron microscopy was used to determine the cell morphology. Chondrogenic molecular signature of hMSCs was determined by quantitative real-time PCR and verified at protein levels via immunohistochemistry and enzyme-linked immunosorbent assay. Results showed that the cells cultured in the presence of PRC for 24 days maintained a chondrocytic phenotype and demonstrated minimal upregulation of cartilaginous extracellular matrix (ECM) marker genes (SOX9, TNC, COL2, ACAN, COMP) and reduced expression of chondrocyte hypertrophy genes (Col X, Runx2) compared to the standard chondrogenic medium (p < 0.05). PRC group had correspondingly higher levels of glycosaminoglycan and increased concentration of chondrogenic specific proteins (COL2, ACAN, COMP) in the ECM. In conclusion, PRC alone appears to be very potent in inducing chondrogenic differentiation of hMSCs and offers additional benefit of suppressing chondrocyte hypertrophy, rendering it a promising approach for providing abundant pool of chondrogenic MSCs for application in cartilage tissue engineering.


Asunto(s)
Alginatos , Cartílago/metabolismo , Condrocitos/metabolismo , Medio de Cultivo Libre de Suero , Matriz Extracelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Plasma Rico en Plaquetas , Alginatos/química , Biomarcadores , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/patología , Condrogénesis , Ensayo de Inmunoadsorción Enzimática , Glicosaminoglicanos/metabolismo , Humanos , Hipertrofia , Inmunohistoquímica , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/ultraestructura , Regeneración , Medicina Regenerativa , Ingeniería de Tejidos
18.
J Biomed Sci ; 25(1): 57, 2018 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-30025541

RESUMEN

BACKGROUND: Induced pluripotency in cancer cells by ectopic expression of pluripotency-regulating factors may be used for disease modeling of cancers. MicroRNAs (miRNAs) are negative regulators of gene expression that play important role in reprogramming somatic cells. However, studies on the miRNA expression profile and the expression patterns of the mesenchymal-epithelial transition (MET)/epithelial-mesenchymal transition (EMT) genes in induced pluripotent cancer (iPC) cells are lacking. METHODS: iPC clones were generated from two colorectal cancer (CRC) cell lines by retroviral transduction of the Yamanaka factors. The iPC clones obtained were characterized by morphology, expression of pluripotency markers and the ability to undergo in vitro tri-lineage differentiation. Genome-wide miRNA profiles of the iPC cells were obtained by microarray analysis and bioinformatics interrogation. Gene expression was done by real-time RT-PCR and immuno-staining; MET/EMT protein levels were determined by western blot analysis. RESULTS: The CRC-iPC cells showed embryonic stem cell-like features and tri-lineage differentiation abilities. The spontaneously-differentiated post-iPC cells obtained were highly similar to the parental CRC cells. However, down-regulated pluripotency gene expression and failure to form teratoma indicated that the CRC-iPC cells had only attained partial pluripotency. The CRC-iPC cells shared similarities in the genome-wide miRNA expression profiles of both cancer and pluripotent embryonic stem cells. One hundred and two differentially-expressed miRNAs were identified in the CRC-iPC cells, which were predicted by bioinformatics analysis be closely involved in regulating cellular pluripotency and the expression of the MET/EMT genes, possibly via the phosphatidylinositol-3 kinases-protein kinase B (PI3K-Akt) and transforming growth factor beta (TGF-ß) signaling pathways. Irregular and inconsistent expression patterns of the EMT vimentin and Snai1 and MET E-cadherin and occludin proteins were observed in the four CRC-iPC clones analyzed, which suggested an epithelial/mesenchymal hybrid phenotype in the partially reprogrammed CRC cells. MET/EMT gene expression was also generally reversed on re-differentiation, also suggesting epigenetic regulation. CONCLUSIONS: Our data support the elite model for cancer cell-reprogramming in which only a selected subset of cancer may be fully reprogrammed; partial cancer cell reprogramming may also elicit an epithelial-mesenchymal mixed phenotype, and highlight opportunities and challenges in cancer cell-reprogramming.


Asunto(s)
Reprogramación Celular/genética , Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal/genética , Proteínas de Neoplasias/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Movimiento Celular/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , MicroARNs/genética , Células Madre Neoplásicas/patología , Fosfatidilinositol 3-Quinasas/genética
19.
Cell Tissue Res ; 373(2): 407-419, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29582166

RESUMEN

Strategies based on mesenchymal stem cell (MSC) therapy for restoring injured articular cartilage are not effective enough in osteoarthritis (OA). Due to the enhanced inflammation and oxidative stress in OA microenvironment, differentiation of MSCs into chondrocytes would be impaired. This study aims to explore the effects of diallyl disulfide (DADS) on IL-1ß-mediated inflammation and oxidative stress in human adipose derived mesenchymal stem cells (hADSCs) during chondrogenesis. MTT assay was employed to examine the effects of various concentrations of DADS on the viability of hADSCs at different time scales to obtain non-cytotoxic concentration range of DADS. The effects of DADS on IL-1ß-induced intracellular ROS generation and lipid peroxidation were evaluated in hADSCs. Western blotting was used to analyze the protein expression levels of IκBα (np), IκBα (p), NF-κB (np) and NF-κB (p). Furthermore, the gene expression levels of antioxidant enzymes in hADSCs and chondrogenic markers at days 7, 14 and 21 of differentiation were measured using qRT-PCR. The results showed that addition of DADS significantly enhanced the mRNA expression levels of antioxidant enzymes as well as reduced ROS elevation, lipid peroxidation, IκBα activation and NF-κB nuclear translocation in hADSCs treated with IL-1ß. In addition, DADS could significantly increase the expression levels of IL-1ß-induced impaired chondrogenic marker genes in differentiated hADSCs. Treatment with DADS may provide an effective approach to prevent the pro-inflammatory cytokines and oxidative stress as catabolic causes of chondrocyte cell death and enhance the protective anabolic effects by promoting chondrogenesis associated gene expressions in hADSCs exposed to OA condition.


Asunto(s)
Tejido Adiposo/citología , Compuestos Alílicos/farmacología , Antioxidantes/metabolismo , Condrogénesis , Disulfuros/farmacología , Interleucina-1beta/metabolismo , Células Madre Mesenquimatosas/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Humanos , Espacio Intracelular/metabolismo , Malondialdehído/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Inhibidor NF-kappaB alfa/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
Injury ; 49(4): 775-783, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29503013

RESUMEN

BACKGROUND: It has been previously suggested that the use of regenerative promoters, which include bone marrow-derived mesenchymal stem cells (MSCs) or natural growth factors supplement such as platelet-rich concentrate (PRC) could promote cartilage regeneration. However, the notion that the concurrent use of both promoters may provide a synergistic effect that improves the repair outcome of focal cartilage injury has not been previously demonstrated. This study was thus conducted to determine whether the concomitant use of PRC could further enhance the reparative potential of MSCs encapsulated in alginate transplanted into focal cartilage injury in rabbits. METHODS: Artifically created full thickness cartilage defects were made on the weight-bearing region of medial femoral condyles in bilateral knees of New Zealand White rabbits (N = 30). After one month, the right knee was treated with either i) PRC (n = 10), ii) MSCs (n = 10), or, iii) a combination of PRC and MSCs (PRC + MSC) (n = 10), all encapsulated in alginate. The left knee remained untreated (control). Rabbits were sacrificed at 3 and 6 months after treatment. Cartilage tissue regeneration was accessed using ICRS morphologic scoring, histologic grading by O'Driscoll scoring, immunohistochemical staining and quantitative analysis of glycosaminoglycans (GAG) per total protein content. RESULTS: At 3 months, transplantation using PRC alone was equally effective as MSCs in inducing the repair of cartilage defects. However, PRC + MSC resulted in significantly higher ICRS and O'Driscoll scores (p < 0.05) as compared to other groups. The regenerated tissues from the PRC + MSC group also had stronger staining for Safranin-O and collagen type II. By 6 months, in addition to superior ICRS and O'Driscoll scores as well as stronger staining, glycosaminoglycan per total protein content was also significantly higher (p < 0.05) in the PRC + MSC group (3.4 ±â€¯0.3 µg/mg) as compared to the MSC (2.6 ±â€¯0.2 µg/mg) or PRC (2.1 ±â€¯0.2 µg/mg) groups. CONCLUSION: PRC enhances the reparative effects of MSC in treating focal articular cartilage injuries.


Asunto(s)
Productos Biológicos/farmacología , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Condrocitos/metabolismo , Traumatismos de la Rodilla/patología , Traumatismos de la Rodilla/terapia , Plasma Rico en Plaquetas/citología , Alginatos/farmacología , Animales , Productos Biológicos/administración & dosificación , Cartílago Articular/lesiones , Células Cultivadas , Modelos Animales de Enfermedad , Fracturas Intraarticulares/patología , Fracturas Intraarticulares/terapia , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Factor de Crecimiento Derivado de Plaquetas/farmacología , Conejos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA