Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Arthroscopy ; 36(8): 2215-2228.e2, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32302651

RESUMO

PURPOSE: To compare the efficacy of mesenchymal stem cell (MSC) exosomes with hyaluronic acid (HA) against HA alone for functional cartilage regeneration in a rabbit osteochondral defect model. METHODS: Critical-size osteochondral defects (4.5-mm diameter and 1.5-mm depth) were created on the trochlear grooves in the knees of 18 rabbits and were randomly allocated to 2 treatment groups: (1) exosomes and HA combination and (2) HA alone. Three 1-mL injections of either exosomes and HA or HA alone were administered intra-articularly immediately after surgery and thereafter at 7 and 14 days after surgery. At 6 and 12 weeks, gross evaluation, histologic and immunohistochemical analysis, and scoring were performed. The functional biomechanical competence of the repaired cartilage also was evaluated. RESULTS: Compared with defects treated with HA, defects treated with exosomes and HA showed significant improvements in macroscopic scores (P = .032; P = .001) and histologic scores (P = .005; P < .001) at 6 and 12 weeks, respectively. Defects treated with exosomes and HA also demonstrated improvements in mechanical properties compared with HA-treated defects, with significantly greater Young's moduli (P < .05) and stiffness (P < .05) at 6 and 12 weeks. By 12 weeks, the newly-repaired tissues in defects treated with exosomes and HA composed mainly of hyaline cartilage that are mechanically and structurally superior to that of HA-treated defects and demonstrated mechanical properties that approximated that of adjacent native cartilage (P > .05). In contrast, HA-treated defects showed some repair at 6 weeks, but this was not sustained, as evidenced by significant deterioration of histologic scores (P = .002) and a plateau in mechanical properties from 6 to 12 weeks. CONCLUSIONS: This study shows that the combination of MSC exosomes and HA administered at a clinically acceptable frequency of 3 intra-articular injections can promote sustained and functional cartilage repair in a rabbit post-traumatic cartilage defect model, when compared with HA alone. CLINICAL RELEVANCE: Human MSC exosomes and HA administered in combination promote functional cartilage repair and may represent a promising cell-free therapy for cartilage repair in patients.


Assuntos
Doenças das Cartilagens/terapia , Cartilagem Articular/cirurgia , Exossomos , Ácido Hialurônico/uso terapêutico , Transplante de Células-Tronco Mesenquimais , Animais , Doenças das Cartilagens/patologia , Módulo de Elasticidade , Feminino , Humanos , Injeções Intra-Articulares , Células-Tronco Mesenquimais/citologia , Coelhos
2.
Stem Cells ; 34(2): 277-87, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26676652

RESUMO

In recent years, the highly conserved promyelocytic leukemia zinc finger (PLZF, also known as ZBTB16, ZNF145) has attracted attention as a multifunctional transcription factor involved in major biological processes during development. As a transcription factor, PLZF shows tight regulation in its cell-type-specific and stage-specific expression patterns. Emerging evidence shows that PLZF regulates the balance of self-renewal and differentiation in stem cells. However, the gene regulatory network of PLZF is only beginning to be understood. In this review, we discuss the diverse functions of PLZF, in particular its role in self-renewal versus differentiation of stem cells. We also discuss the current state of knowledge on the gene regulatory network of PLZF, in conjunction with its upstream factors, post-translational modifications and binding cofactors for multiprotein complexes. This review aims to provide the reader with an in-depth understanding of the molecular mechanisms underlying PLZF and the potential applications in tissue regeneration.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Regulação da Expressão Gênica/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Células-Tronco/metabolismo , Animais , Humanos , Proteína com Dedos de Zinco da Leucemia Promielocítica , Células-Tronco/citologia
3.
J Pediatr Orthop ; 36(1): 63-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26642076

RESUMO

BACKGROUND: Spine surgery for neuromuscular scoliosis in patients with Duchenne's Muscular Dystrophy (DMD) and Spinal Muscular Atrophy (SMA) remained controversial. This study aimed to review the long-term results of spine surgery and its effect on pulmonary function in these patients. METHODS: A retrospective review was conducted for the above patients who had undergone surgery from 1990 to 2006 in a tertiary hospital. Their yearly lung function tests, clinical records, and x-ray films before and after surgery were reviewed. All patients had at least 2 lung function tests performed before surgery and at least 3 lung function tests performed after surgery. Records of perioperative pulmonary infections that resulted in hospital admissions were also retrieved from the hospital computer system. RESULTS: Forty patients were reviewed: 29 with DMD, 11 with SMA. The mean follow-up period was 11.6 years. For patients with DMD, the mean correction of Cobb's angle from surgery was 34.1 degrees. The rate of decline of the predicted forced vital capacity preoperatively was 7.80% per year, and was reduced to 4.26% per year postoperatively (P<0.001). For patients with SMA, the mean correction of Cobb's angle from surgery was 44.1 degrees. The rate of decline of the predicted forced vital capacity preoperatively was 5.31% per year, and was reduced to 1.77% per year postoperatively (P<0.001). For both DMD and SMA patients, the difference between the rate of preoperative and postoperative pulmonary infections that resulted in hospital admission were, however, not significant (P=0.433 and 0.452, respectively). CONCLUSIONS: Scoliosis surgery in patients with DMD and SMA results in a long-term decreased rate of decline in pulmonary function over a follow-up period of more than 10 years. The level of the apical vertebrae of the scoliosis did not demonstrate a significant trend on the pulmonary function. The frequency of chest infections did not improve by scoliosis surgery. LEVEL OF SIGNIFICANCE: Level III­Retrospective study.


Assuntos
Volume Expiratório Forçado/fisiologia , Previsões , Pulmão/fisiopatologia , Atrofia Muscular Espinal/complicações , Distrofia Muscular de Duchenne/complicações , Escoliose/cirurgia , Fusão Vertebral , Adolescente , Feminino , Seguimentos , Humanos , Masculino , Atrofia Muscular Espinal/fisiopatologia , Atrofia Muscular Espinal/cirurgia , Distrofia Muscular de Duchenne/fisiopatologia , Distrofia Muscular de Duchenne/cirurgia , Testes de Função Respiratória , Estudos Retrospectivos , Escoliose/etiologia , Escoliose/fisiopatologia , Resultado do Tratamento
4.
Nanomedicine ; 10(7): 1507-16, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24768908

RESUMO

To reproduce a complex and functional tissue, it is crucial to provide a biomimetic cellular microenvironment that not only incorporates biochemical cues, but also physical features including the nano-topographical patterning, for cell/matrix interaction. We developed spatially-controlled nano-topography in the form of nano-pillar, nano-hole and nano-grill on polycaprolactone surface via thermal nanoimprinting. The effects of chondroitin sulfate-coated nano-topographies on cell characteristics and chondrogenic differentiation of human mesenchymal stem cell (MSC) were investigated. Our results show that various nano-topographical patterns triggered changes in MSC morphology and cytoskeletal structure, affecting cell aggregation and differentiation. Compared to non-patterned surface, nano-pillar and nano-hole topography enhanced MSC chondrogenesis and facilitated hyaline cartilage formation. MSCs experienced delayed chondrogenesis on nano-grill topography and were induced to fibro/superficial zone cartilage formation. This study demonstrates the sensitivity of MSC differentiation to surface nano-topography and highlights the importance of incorporating topographical design in scaffolds for cartilage tissue engineering. From the clinical editor: These authors have developed spatially-controlled nano-topography in the form of nano-pillar, nano-hole and nano-grill on polycaprolactone surface via thermal nanoimprinting, and the effects of chondroitin sulfate-coated nano-topographies on cell characteristics and chondrogenic differentiation of human mesenchymal stem cells (MSC) were investigated. It has been concluded that MSC differentiation is sensitive to surface nano-topography, and certain nano-imprinted surfaces are more useful than others for cartilage tissue engineering.


Assuntos
Cartilagem/citologia , Condrogênese , Células-Tronco Mesenquimais/citologia , Linhagem da Célula , Proliferação de Células , Humanos , Microscopia de Força Atômica , Microscopia Eletrônica de Varredura , Alicerces Teciduais
5.
Arthroscopy ; 30(4): 497-505, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24680310

RESUMO

PURPOSE: Our purpose was to examine the Level I and II evidence for the use of osteochondral cylinder transfer technique (OCT) for cartilage repair. METHODS: A literature search was carried out for Level I and II evidence studies on cartilage repair using the PubMed database. All the studies that involved OCT were identified. Only Level I and II studies that compared OCT to other modalities of treatment such as microfracture (MF) and autologous chondrocyte implantation (ACI) were selected. RESULTS: A total of 8 studies matched the selection criteria with 2 Level I and 6 Level II studies. Four studies compared OCT with MF, 3 compared OCT with ACI, and one compared all 3 techniques. Of 3 studies, 4 came from a single center. Mean age of patients ranged from 24 to 33 years, and mean follow-up ranged from 9 to 124 months. The studies from the single center showed superior results from OCT over MF, especially in younger patients, with one study having long-term follow-up of 10 years. They also showed an earlier return to sports. The size of the lesions were small (average < 3 cm(2)). The 4 other independent studies did not show any difference between OCT and ACI, with one study showing inferior outcome in the OCT group. Magnetic resonance imaging (MRI) showed good osseous integration of the osteochondral plugs to the subchondral bone. Histologic examination showed that there was hyaline cartilage in the transplanted osteochondral plugs but no hyaline cartilage between the plugs. CONCLUSIONS: From the studies of a single center, OCT had an advantage over MF in younger patients with small chondral lesions. Comparison of outcomes between OCT and ACI showed no significant difference in 2 studies and contrasting results in another 2 studies. There was insufficient evidence for long-term results for OCT. LEVEL OF EVIDENCE: Level II, systematic review of Level I and II studies.


Assuntos
Doenças das Cartilagens/cirurgia , Cartilagem Articular/cirurgia , Adolescente , Adulto , Artroplastia Subcondral , Transplante Ósseo , Cartilagem Articular/lesões , Condrócitos/transplante , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Transplante Autólogo , Adulto Jovem
6.
Am J Sports Med ; 52(2): 503-515, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38186352

RESUMO

BACKGROUND: The functional heterogeneity of culture-expanded mesenchymal stem cells (MSCs) has hindered the clinical application of MSCs. Previous studies have shown that MSC subpopulations with superior chondrogenic capacity can be isolated using a spiral microfluidic device based on the principle of inertial cell focusing. HYPOTHESIS: The delivery of microfluidic-enriched chondrogenic MSCs that are consistent in size and function will overcome the challenge of the functional heterogeneity of expanded MSCs and will significantly improve MSC-based cartilage repair. STUDY DESIGN: Controlled laboratory study. METHODS: A next-generation, fully automated multidimensional double spiral microfluidic device was designed to provide more refined and efficient isolation of MSC subpopulations based on size. Analysis of in vitro chondrogenic potential and RNA sequencing was performed on size-sorted MSC subpopulations. In vivo cartilage repair efficacy was demonstrated in an osteochondral injury model in 12-week-old rats. Defects were implanted with MSC subpopulations (n = 6 per group) and compared with those implanted with unsegregated MSCs (n = 6). Osteochondral repair was assessed at 6 and 12 weeks after surgery by histological, micro-computed tomography, and mechanical analysis. RESULTS: A chondrogenic MSC subpopulation was efficiently isolated using the multidimensional double spiral device. RNA sequencing revealed distinct transcriptomic profiles and identified differential gene expression between subpopulations. The delivery of a chondrogenic MSC subpopulation resulted in improved cartilage repair, as indicated by histological scoring, the compression modulus, and micro-computed tomography of the subchondral bone. CONCLUSION: We have established a rapid, label-free, and reliable microfluidic protocol for more efficient size-based enrichment of a chondrogenic MSC subpopulation. Our proof-of-concept in vivo study demonstrates the enhanced cartilage repair efficacy of these enriched chondrogenic MSCs. CLINICAL RELEVANCE: The delivery of microfluidic-enriched chondrogenic MSCs that are consistent in size and function can overcome the challenge of the functional heterogeneity of expanded MSCs, resulting in significant improvement in MSC-based cartilage repair. The availability of such rapid, label-free enriched chondrogenic MSCs can enable better cell therapy products for cartilage repair with improved treatment outcomes.


Assuntos
Cartilagem Articular , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Ratos , Cartilagem Articular/cirurgia , Microfluídica , Microtomografia por Raio-X , Diferenciação Celular , Transplante de Células-Tronco Mesenquimais/métodos , Condrogênese
7.
PLoS One ; 19(3): e0297769, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38547243

RESUMO

Mesenchymal stromal cells (MSCs) are promising therapeutic agents for cartilage regeneration, including the potential of cells to promote chondrogenesis in vivo. However, process development and regulatory approval of MSCs as cell therapy products benefit from facile in vitro approaches that can predict potency for a given production run. Current standard in vitro approaches include a 21 day 3D differentiation assay followed by quantification of cartilage matrix proteins. We propose a novel biophysical marker that is cell population-based and can be measured from in vitro monolayer culture of MSCs. We hypothesized that the self-assembly pattern that emerges from collective-cell behavior would predict chondrogenesis motivated by our observation that certain features in this pattern, namely, topological defects, corresponded to mesenchymal condensations. Indeed, we observed a strong predictive correlation between the degree-of-order of the pattern at day 9 of the monolayer culture and chondrogenic potential later estimated from in vitro 3D chondrogenic differentiation at day 21. These findings provide the rationale and the proof-of-concept for using self-assembly patterns to monitor chondrogenic commitment of cell populations. Such correlations across multiple MSC donors and production batches suggest that self-assembly patterns can be used as a candidate biophysical attribute to predict quality and efficacy for MSCs employed therapeutically for cartilage regeneration.


Assuntos
Condrogênese , Células-Tronco Mesenquimais , Humanos , Cartilagem/metabolismo , Diferenciação Celular , Doadores de Tecidos , Células Cultivadas
8.
Arthroscopy ; 29(9): 1579-88, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23992991

RESUMO

PURPOSE: Although many newer cartilage repair techniques have evolved over the past 2 decades, microfracture is still being advocated as the first line of treatment. Therefore it is timely to conduct a comprehensive review of the literature to assess and report on the current status of Level I and II evidence studies related to microfracture techniques. METHODS: A literature search was carried out for Level I and II evidence studies on cartilage repair using the PubMed database. All the studies that dealt with microfracture techniques were selected. RESULTS: Fifteen studies that involved microfracture techniques met the inclusion criteria of this review article, with 6 long-term and 9 short-term studies. These studies compared the clinical outcomes of microfracture with those of other treatments such as autologous chondrocyte implantation and osteochondral cylinder transfers. The majority of the studies reported poor clinical outcomes, whereas 2 studies reported the absence of any significant difference in the results. Small-sized lesions and younger patients showed good results in the short-term. However, osteoarthritis and treatment failures were observed at later postoperative periods of 5 to 10 years. CONCLUSIONS: The use of microfracture for the treatment of small lesions in patients with low postoperative demands was observed to result in good clinical outcomes at short-term follow-up. Beyond 5 years postoperatively, treatment failure after microfracture could be expected regardless of lesion size. Younger patients showed better clinical outcomes. LEVEL OF EVIDENCE: Level II, systematic review of Level I and II studies.


Assuntos
Cartilagem Articular/cirurgia , Osteotomia/métodos , Fatores Etários , Cartilagem Articular/fisiologia , Condrócitos/transplante , Humanos , Regeneração/fisiologia , Transplante Autólogo , Falha de Tratamento
9.
Arthroscopy ; 29(11): 1872-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24075851

RESUMO

PURPOSE: The purpose of this study was to examine the Level I and II evidence for newer generations of autologous chondrocyte implantation (ACI) versus first-generation ACI and to establish whether the newer generations have overcome the limitations associated with first-generation ACI. METHODS: A literature search was carried out for Level I and II evidence studies on cartilage repair using the PubMed database. All the studies that dealt with ACI were identified. Only Level I and II studies that compared newer generations against earlier generations were selected, whereas studies that compared ACI against other methods of cartilage repair were excluded. RESULTS: A total of 7 studies matched the selection criteria. Two studies compared periosteum-based autologous chondrocyte implantation (P-ACI) against collagen membrane-based autologous chondrocyte implantation (C-ACI), whereas one study each compared membrane-associated autologous chondrocyte implantation (MACI) against P-ACI and C-ACI. One study on C-ACI compared results related to age, whereas 2 studies evaluated postoperative rehabilitation after MACI. There was weak evidence showing that C-ACI is better than P-ACI and that MACI is comparable with both P-ACI and C-ACI. The weak evidence is because of studies with short durations of follow-up, small numbers of patients, medium-sized defects, and younger age groups. There is good evidence favoring an accelerated weight-bearing regimen after MACI. There is currently no evidence that supports scaffold-based ACI or arthroscopic implantation over first-generation ACI. CONCLUSIONS: The hypothesis is thus partly proved in favor of C-ACI/MACI against P-ACI with weak evidence, in favor of accelerated weight bearing after MACI with strong evidence, and not in favor of arthroscopic and scaffold-based implantations because of unavailable evidence. LEVEL OF EVIDENCE: Level II, systematic review of Level I and II studies.


Assuntos
Cartilagem Articular/cirurgia , Condrócitos/transplante , Medicina Baseada em Evidências/métodos , Humanos , Procedimentos Ortopédicos/métodos , Transplante Autólogo/métodos
10.
Arthroscopy ; 29(12): 2020-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24286801

RESUMO

PURPOSE: To analyze the results of the use of intra-articular cultured autologous bone marrow-derived mesenchymal stem cell (MSC) injections in conjunction with microfracture and medial opening-wedge high tibial osteotomy (HTO). METHODS: Fifty-six knees in 56 patients with unicompartmental osteoarthritic knees and genu varum were randomly allocated to the cell-recipient group (n = 28) or control group (n = 28). Patients who had a joint line congruity angle of more than 2°, malalignment of the knee from femoral causes, a fixed flexion deformity, or age older than 55 years were excluded. All patients underwent HTO and microfracture. The cell-recipient group received intra-articular injection of cultured MSCs with hyaluronic acid 3 weeks after surgery, whereas the control group only received hyaluronic acid. The primary outcome measure was the International Knee Documentation Committee (IKDC) score at intervals of 6 months, 1 year, and 2 years postoperatively. Secondary outcome measures were Tegner and Lysholm clinical scores and 1-year postoperative Magnetic Resonance Observation of Cartilage Repair Tissue (MOCART) scores. RESULTS: The median age of the patients was 51 years, with a mean body mass index of 23.85. Both treatment arms achieved improvements in Tegner, Lysholm, and IKDC scores. After adjustment for age, baseline scores, and time of evaluation, the cell-recipient group showed significantly better scores. The effect of treatment showed an added improvement of 7.65 (95% confidence interval [CI], 3.04 to 12.26; P = .001) for IKDC scores, 7.61 (95% CI, 1.44 to 13.79; P = .016) for Lysholm scores, and 0.64 (95% CI, 0.10 to 1.19; P = .021) for Tegner scores. Magnetic resonance imaging scans performed 1 year after surgical intervention showed significantly better MOCART scores for the cell-recipient group. The age-adjusted mean difference in MOCART score was 19.6 (95% CI, 10.5 to 28.6; P < .001). CONCLUSIONS: Intra-articular injection of cultured MSCs is effective in improving both short-term clinical and MOCART outcomes in patients undergoing HTO and microfracture for varus knees with cartilage defects. LEVEL OF EVIDENCE: Level II, randomized controlled trial.


Assuntos
Cartilagem Articular/anormalidades , Cartilagem Articular/cirurgia , Genu Varum/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Osteoartrite do Joelho/complicações , Adulto , Artroplastia Subcondral , Células da Medula Óssea/citologia , Células Cultivadas , Feminino , Seguimentos , Genu Varum/complicações , Humanos , Ácido Hialurônico , Injeções Intra-Articulares , Masculino , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade , Osteotomia/reabilitação , Estudos Prospectivos , Resultado do Tratamento , Adulto Jovem
11.
Tissue Eng Part B Rev ; 29(3): 310-330, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36416231

RESUMO

Articular cartilage is composed of superficial, medial, and deep zones, which endow the tissue with biphasic mechanical properties to withstand shearing force and compressional loading. The tissue has very limited self-healing capacity once it is damaged due to its avascular nature. To prevent the early onset of osteoarthritis, surgical intervention is often needed to repair the injured cartilage. Current noncell-based and cell-based treatments focus on the regeneration of homogeneous cartilage to achieve bulk compressional properties without recapitulating the zonal matrix and mechanical properties, and often oversight in aiding cartilage integration between host and repair cartilage. It is hypothesized that achieving zonal architecture in articular cartilage tissue repair could improve the structural and mechanical integrity and thus the life span of the regenerated tissue. Engineering stratified cartilage constructs using zonal chondrocytes have been hypothesized to improve the functionality and life span of the regenerated tissues. However, stratified articular cartilage repair has yet to be realized to date due to the lack of an efficient zonal chondrocyte isolation method and an expansion platform that would allow both cell propagation and phenotype maintenance. Various attempts and challenges in achieving stratified articular cartilage repair in a clinical setting are evaluated. In this review, different perspectives on achieving stratified articular cartilage repair using zonal chondrocytes are described. The effectiveness of different zonal chondrocyte isolation and zonal chondrocyte phenotype maintenance methodologies during expansion are compared, with the focus on recent advancements in zonal chondrocyte isolation and expansion that could present a possible strategy to overcome the limitation of applying zonal chondrocytes to facilitate zonal architecture development in articular cartilage regeneration. Impact Statement The zonal properties of articular cartilage contribute to the biphasic mechanical properties of the tissues. Recapitulation of the zonal architecture in regenerated articular cartilage has been hypothesized to improve the mechanical integrity and life span of the regenerated tissue. This review provides a comprehensive discussion on the current state of research relevant to achieving stratified articular cartilage repair using zonal chondrocytes from different perspectives. This review further elaborates on a zonal chondrocyte production pipeline that can potentially overcome the current clinical challenges and future work needed to realize stratified zonal chondrocyte implantation in a clinical setting.


Assuntos
Cartilagem Articular , Osteoartrite , Humanos , Condrócitos , Engenharia Tecidual/métodos , Regeneração
12.
Bioengineering (Basel) ; 10(3)2023 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-36978745

RESUMO

Articular cartilage defects commonly result from trauma and are associated with significant morbidity. Since cartilage is an avascular, aneural, and alymphatic tissue with a poor intrinsic healing ability, the regeneration of functional hyaline cartilage remains a difficult clinical problem. Mesenchymal stem cells (MSCs) are multipotent cells with multilineage differentiation potential, including the ability to differentiate into chondrocytes. Due to their availability and ease of ex vivo expansion, clinicians are increasingly applying MSCs in the treatment of cartilage lesions. However, despite encouraging pre-clinical and clinical data, inconsistencies in MSC proliferative and chondrogenic potential depending on donor, tissue source, cell subset, culture conditions, and handling techniques remain a key barrier to widespread clinical application of MSC therapy in cartilage regeneration. In this review, we highlight the strategies to manage the heterogeneity of MSCs ex vivo for more effective cartilage repair, including reducing the MSC culture expansion period, and selecting MSCs with higher chondrogenic potential through specific genetic markers, surface markers, and biophysical attributes. The accomplishment of a less heterogeneous population of culture-expanded MSCs may improve the scalability, reproducibility, and standardisation of MSC therapy for clinical application in cartilage regeneration.

13.
Bioact Mater ; 27: 98-112, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37006826

RESUMO

Secretome derived from mesenchymal stem cells (MSCs) have profound effects on tissue regeneration, which could become the basis of future MSCs therapies. Hypoxia, as the physiologic environment of MSCs, has great potential to enhance MSCs paracrine therapeutic effect. In our study, the paracrine effects of secretome derived from MSCs preconditioned in normoxia and hypoxia was compared through both in vitro functional assays and an in vivo rat osteochondral defect model. Specifically, the paracrine effect of total EVs were compared to that of soluble factors to characterize the predominant active components in the hypoxic secretome. We demonstrated that hypoxia conditioned medium, as well as the corresponding EVs, at a relatively low dosage, were efficient in promoting the repair of critical-sized osteochondral defects and mitigated the joint inflammation in a rat osteochondral defect model, relative to their normoxia counterpart. In vitro functional test shows enhancement through chondrocyte proliferation, migration, and matrix deposition, while inhibit IL-1ß-induced chondrocytes senescence, inflammation, matrix degradation, and pro-inflammatory macrophage activity. Multiple functional proteins, as well as a change in EVs' size profile, with enrichment of specific EV-miRNAs were detected with hypoxia preconditioning, implicating complex molecular pathways involved in hypoxia pre-conditioned MSCs secretome generated cartilage regeneration.

14.
Arthritis Rheum ; 63(9): 2711-20, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21547890

RESUMO

OBJECTIVE: Human mesenchymal stem cells (hMSCs) represent one of the most promising stem cell therapies for traumatic injury and age-related degenerative diseases involving cartilage. However, few genetic factors regulating chondrogenesis of MSCs have been identified. One study showed that zinc-finger protein 145 (ZNF145), a transcription factor, was up-regulated during 3-lineage differentiation of hMSCs. The present study was undertaken to validate whether this novel transcription factor is useful for the repair and regeneration of cartilage. METHODS: Human MSCs were transfected with lentiviral short hairpin RNA (for small interfering RNA knockdown of ZNF145) and a lentiviral vector for overexpression of ZNF145, and the effects of ZNF145 on chondrogenesis were studied using quantitative polymerase chain reaction and immunostaining. Microarray and transient expression analyses were used to determine whether ZNF145 is a factor operating upstream of SOX9. Allogeneic transplantation of hMSCs into osteochondral defects in rats was performed to determine the effects of ZNF145 on repair of cartilage in vivo. RESULTS: Small interfering RNA-mediated gene silencing of ZNF145 slowed down chondrogenesis, whereas overexpression of ZNF145 enhanced chondrogenesis. Global gene expression profiling showed up-regulated gene expression in ZNF145-overexpressing MSCs, and transient overexpression of ZNF145 enhanced the expression of SOX9, suggesting that ZNF145 acts as a factor upstream of SOX9, the master regulator of chondrogenesis. Moreover, allogeneic transplantation of hMSCs into osteochondral defects of rat knees showed that ZNF145-overexpressing MSCs repaired cartilage defects better and earlier than empty control MSCs. CONCLUSION: These findings suggest that ZNF145 gene therapy may be a very useful strategy for improving the quality of cartilage regeneration and repair.


Assuntos
Cartilagem/metabolismo , Condrogênese/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fatores de Transcrição SOX9/metabolismo , Animais , Linhagem da Célula , Condrócitos/metabolismo , Humanos , Masculino , Proteína com Dedos de Zinco da Leucemia Promielocítica , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Regeneração/fisiologia , Transdução de Sinais/fisiologia
15.
Tissue Eng Part B Rev ; 28(5): 966-977, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34569290

RESUMO

Mesenchymal stem cells (MSCs) have been demonstrated as promising cell sources for tissue regeneration due to their capability of self-regeneration, differentiation, and immunomodulation. MSCs also exert extensive paracrine effects through release of trophic factors and extracellular vesicles (EVs). However, despite extended exploration of MSCs in preclinical studies, the results are far from satisfactory due to the poor engraftment and low level of survival after implantation. Hypoxia preconditioning has been proposed as an engineering approach to improve the therapeutic potential of MSCs. During in vitro culture, hypoxic conditions can promote MSC proliferation, survival, and migration through various cellular responses to the reduction of oxygen tension. The multilineage differentiation potential of MSCs is altered under hypoxia, with consistent reports of enhanced chondrogenesis. Hypoxia also stimulates the paracrine activities of MSCs and increases the production of secretome both in terms of soluble factors as well as EVs. The secretome from hypoxia-preconditioned MSCs play important roles in promoting cell proliferation and migration, enhancing angiogenesis while inhibiting apoptosis and inflammation. In this review, we summarize current knowledge of hypoxia-induced changes in MSCs and discuss the application of hypoxia-preconditioned MSCs as well as hypoxic secretome in different kinds of disease models. Impact statement Mesenchymal stem cells (MSCs) have been applied in numerous cell-based and secretome-based therapies for tissue regeneration. Hypoxic conditions enhance the function of MSCs by increasing proliferation, survival, homing, differentiation, and paracrine activities. A timely up-to-date comprehensive overview of the effect of low oxygen tension to MSC, with emphasis on the influence and molecular mechanism of hypoxia preconditioning toward MSC's functionality is provided, including the therapeutic use of hypoxia-preconditioned MSC as well as hypoxic secretome in various prove-of-concept disease models. This knowledge would contribute to future engineering of MSC culture conditions for improved translational application.


Assuntos
Células-Tronco Mesenquimais , Humanos , Diferenciação Celular , Hipóxia , Cicatrização , Oxigênio/farmacologia
16.
Cartilage ; 13(2): 19476035221093063, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35446156

RESUMO

OBJECTIVE: The zonal properties of articular cartilage critically contribute to the mechanical support and lubrication of the tissue. Current treatments for articular cartilage have yet to regenerate this zonal architecture, thus compromising the functional efficacy of the repaired tissue and leading to tissue degeneration in the long term. In this study, the efficacy of zonal cartilage regeneration through bilayered implantation of expanded autologous zonal chondrocytes was investigated in a porcine chondral defect model. DESIGN: Autologous chondrocytes extracted from articular cartilage in the non-weight bearing trochlea region of the knee were subjected to an expansion-sorting strategy, integrating dynamic microcarrier (dMC) culture, and spiral microchannel size-based zonal chondrocyte separation. Zonal chondrocytes were then implanted as bilayered fibrin hydrogel construct in a porcine knee chondral defect model. Repair efficacy was compared with implantation with cell-free fibrin hydrogel and full thickness (FT) cartilage-derived heterogenous chondrocytes. Cartilage repair was evaluated 6 months after implantation. RESULTS: Sufficient numbers of zonal chondrocytes for implantation were generated from the non-weight bearing cartilage. Six-month repair outcomes showed that bilayered implantation of dMC-expanded zonal chondrocytes resulted in substantial recapitulation of zonal architecture, including chondrocyte arrangement, specific Proteoglycan 4 distribution, and collagen alignment, that was accompanied by healthier underlying subchondral bone. CONCLUSION: These results demonstrate that with appropriate expansion and isolation of zonal chondrocytes, the strategy of stratified zonal chondrocyte implantation represents a significant advancement to Autologous Chondrocyte Implantation-based cartilage regeneration, with the potential to improve the long-term integrity of the regenerated tissues.


Assuntos
Doenças das Cartilagens , Cartilagem Articular , Animais , Cartilagem Articular/cirurgia , Condrócitos , Fibrina , Hidrogéis , Suínos
17.
Mol Pharm ; 8(4): 994-1001, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21500855

RESUMO

Articular cartilage injuries are one of the most challenging problems in musculoskeletal medicine due to the poor intrinsic regenerative capacity of this tissue. The lack of efficient treatment modalities motivates research into tissue engineering: combining cells, biomaterials mimicking extracellular matrix (scaffolds) and microenvironmental signaling cues. The aim of this review is to focus on the use of biomaterials as delivery systems for microenvironmental cues in relation to their applications for treatment of cartilage defects. The latest advances in cartilage tissue engineering and regeneration are critically reviewed to demonstrate an outline of challenges toward biomaterial-based approaches of cartilage regeneration.


Assuntos
Materiais Biocompatíveis/química , Cartilagem Articular/citologia , Engenharia Tecidual/métodos , Cartilagem Articular/lesões , Humanos , Medicina Regenerativa/métodos
18.
Stem Cell Res Ther ; 12(1): 100, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33536060

RESUMO

BACKGROUND: Secretome profiles of mesenchymal stem cells (MSCs) are reflective of their local microenvironments. These biologically active factors exert an impact on the surrounding cells, eliciting regenerative responses that create an opportunity for exploiting MSCs towards a cell-free therapy for cartilage regeneration. The conventional method of culturing MSCs on a tissue culture plate (TCP) does not provide the physiological microenvironment for optimum secretome production. In this study, we explored the potential of electrospun fiber sheets with specific orientation in influencing the MSC secretome production and its therapeutic value in repairing cartilage. METHODS: Conditioned media (CM) were generated from MSCs cultured either on TCP or electrospun fiber sheets of distinct aligned or random fiber orientation. The paracrine potential of CM in affecting chondrogenic differentiation, migration, proliferation, inflammatory modulation, and survival of MSCs and chondrocytes was assessed. The involvement of FAK and ERK mechanotransduction pathways in modulating MSC secretome were also investigated. RESULTS: We showed that conditioned media of MSCs cultured on electrospun fiber sheets compared to that generated from TCP have improved secretome yield and profile, which enhanced the migration and proliferation of MSCs and chondrocytes, promoted MSC chondrogenesis, mitigated inflammation in both MSCs and chondrocytes, as well as protected chondrocytes from apoptosis. Amongst the fiber sheet-generated CM, aligned fiber-generated CM (ACM) was better at promoting cell proliferation and augmenting MSC chondrogenesis, while randomly oriented fiber-generated CM (RCM) was more efficient in mitigating the inflammation assault. FAK and ERK signalings were shown to participate in the modulation of MSC morphology and its secretome production. CONCLUSIONS: This study demonstrates topographical-dependent MSC paracrine activities and the potential of employing electrospun fiber sheets to improve the MSC secretome for cartilage regeneration.


Assuntos
Células-Tronco Mesenquimais , Cartilagem , Diferenciação Celular , Condrogênese , Mecanotransdução Celular , Comunicação Parácrina
19.
Acta Biomater ; 119: 169-183, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33130304

RESUMO

Mesenchymal stem cell (MSC) chondrogenesis is modulated by diverse biophysical cues. We have previously shown that brief, low-amplitude pulsed electromagnetic fields (PEMFs) differentially enhance MSC chondrogenesis in scaffold-free pellet cultures versus conventional tissue culture plastic (TCP), indicating an interplay between magnetism and micromechanical environment. Here, we examined the influence of PEMF directionality over the chondrogenic differentiation of MSCs laden on electrospun fibrous scaffolds of either random (RND) or aligned (ALN) orientations. Correlating MSCs' chondrogenic outcome to pFAK activation and YAP localisation, MSCs on the RND scaffolds experienced the least amount of resting mechanical stress and underwent greatest chondrogenic differentiation in response to brief PEMF exposure (10 min at 1 mT) perpendicular to the dominant plane of the scaffolds (Z-directed). By contrast, in MSC-impregnated RND scaffolds, greatest mitochondrial respiration resulted from X-directed PEMF exposure (parallel to the scaffold plane), and was associated with curtailed chondrogenesis. MSCs on TCP or the ALN scaffolds exhibited greater resting mechanical stress and accordingly, were unresponsive, or negatively responsive, to PEMF exposure from all directions. The efficacy of PEMF-induced MSC chondrogenesis is hence regulated in a multifaceted manner involving focal adhesion dynamics, as well as mitochondrial responses, culminating in a final cellular response. The combined contributions of micromechanical environment and magnetic field orientation hence will need to be considered when designing magnetic exposure paradigms.


Assuntos
Condrogênese , Células-Tronco Mesenquimais , Adesão Celular , Diferenciação Celular , Campos Eletromagnéticos , Alicerces Teciduais
20.
Stem Cells ; 27(8): 1792-801, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19544435

RESUMO

The monoclonal antibody mAb 84, which binds to podocalyxin-like protein-1 (PODXL) on human embryonic stem cells (hESCs), was previously reported to bind and kill undifferentiated cells in in vitro and in vivo assays. In this study, we investigate the mechanism responsible for mAb 84-induced hESCs cytotoxicity. Apoptosis was likely not the cause of mAb 84-mediated cell death because no elevation of caspase activities or increased DNA fragmentation was observed in hESCs following incubation with mAb 84. Instead, it was preceded by cell aggregation and damage to cell membranes, resulting in the uptake of propidium iodide, and the leakage of intracellular sodium ions. Furthermore, examination of the cell surface by scanning electron microscopy revealed the presence of pores on the cell surface of mAb 84-treated cells, which was absent from the isotype control. This mechanism of cell death resembles that described for oncosis, a form of cell death resulting from membrane damage. Additional data suggest that the binding of mAb 84 to hESCs initiates a sequence of events prior to membrane damage, consistent with oncosis. Degradation of actin-associated proteins, namely, alpha-actinin, paxillin, and talin, was observed. The perturbation of these actin-associated proteins consequently permits the aggregation of PODXL, thus leading to the formation of pores. To our knowledge, this is the first report of oncotic cell death with hESCs as a model.


Assuntos
Anticorpos Monoclonais/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Actinina/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Agregação Celular/efeitos dos fármacos , Agregação Celular/imunologia , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Diferenciação Celular/imunologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/imunologia , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Humanos , Camundongos , Camundongos SCID , Microscopia Eletrônica de Varredura , Paxilina/metabolismo , Sialoglicoproteínas/metabolismo , Talina/metabolismo , Teratoma/imunologia , Teratoma/prevenção & controle , Vinculina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA