Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Cell Physiol ; 236(11): 7421-7439, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34008188

RESUMO

Chondrogenic progenitor cells (CPCs) may be used as an alternative source of cells with potentially superior chondrogenic potential compared to mesenchymal stem cells (MSCs), and could be exploited for future regenerative therapies targeting articular cartilage in degenerative diseases such as osteoarthritis (OA). In this study, we hypothesised that CPCs derived from OA cartilage may be characterised by a distinct channelome. First, a global transcriptomic analysis using Affymetrix microarrays was performed. We studied the profiles of those ion channels and transporter families that may be relevant to chondroprogenitor cell physiology. Following validation of the microarray data with quantitative reverse transcription-polymerase chain reaction, we examined the role of calcium-dependent potassium channels in CPCs and observed functional large-conductance calcium-activated potassium (BK) channels involved in the maintenance of the chondroprogenitor phenotype. In line with our very recent results, we found that the KCNMA1 gene was upregulated in CPCs and observed currents that could be attributed to the BK channel. The BK channel inhibitor paxilline significantly inhibited proliferation, increased the expression of the osteogenic transcription factor RUNX2, enhanced the migration parameters, and completely abolished spontaneous Ca2+ events in CPCs. Through characterisation of their channelome we demonstrate that CPCs are a distinct cell population but are highly similar to MSCs in many respects. This study adds key mechanistic data to the in-depth characterisation of CPCs and their phenotype in the context of cartilage regeneration.


Assuntos
Cartilagem Articular/metabolismo , Movimento Celular , Condrócitos/metabolismo , Canais Iônicos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Osteoartrite do Joelho/metabolismo , Células-Tronco/metabolismo , Transcriptoma , Sinalização do Cálcio , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Perfilação da Expressão Gênica , Humanos , Canais Iônicos/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Potenciais da Membrana , Proteínas de Membrana Transportadoras/genética , Osteoartrite do Joelho/genética , Osteoartrite do Joelho/patologia , Bloqueadores dos Canais de Potássio/farmacologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Fatores de Tempo
2.
Cell Tissue Res ; 386(2): 309-320, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34241697

RESUMO

Cartilage has little intrinsic capacity for repair, so transplantation of exogenous cartilage cells is considered a realistic option for cartilage regeneration. We explored whether human-induced pluripotent stem cells (hiPSCs) could represent such unlimited cell sources for neo-cartilage comparable to human primary articular chondrocytes (hPACs) or human bone marrow-derived mesenchymal stromal cells (hBMSCs). For this, chondroprogenitor cells (hiCPCs) and hiPSC-derived mesenchymal stromal cells (hiMSCs) were generated from two independent hiPSC lines and characterized by morphology, flow cytometry, and differentiation potential. Chondrogenesis was compared to hBMSCs and hPACs by histology, immunohistochemistry, and RT-qPCR, while similarities were estimated based on Pearson correlations using a panel of 20 relevant genes. Our data show successful differentiations of hiPSC into hiMSCs and hiCPCs. Characteristic hBMSC markers were shared between hBMSCs and hiMSCs, with the exception of CD146 and CD45. However, neo-cartilage generated from hiMSCs showed low resemblances when compared to hBMSCs (53%) and hPACs (39%) characterized by lower collagen type 2 and higher collagen type 1 expression. Contrarily, hiCPC neo-cartilage generated neo-cartilage more similar to hPACs (65%), with stronger expression of matrix deposition markers. Our study shows that taking a stepwise approach to generate neo-cartilage from hiPSCs via chondroprogenitor cells results in strong similarities to neo-cartilage of hPACs within 3 weeks following chondrogenesis, making them a potential candidate for regenerative therapies. Contrarily, neo-cartilage deposited by hiMSCs seems more prone to hypertrophic characteristics compared to hPACs. We therefore compared chondrocytes derived from hiMSCs and hiCPCs with hPACs and hBMSCs to outline similarities and differences between their neo-cartilage and establish their potential suitability for regenerative medicine and disease modelling.


Assuntos
Cartilagem/citologia , Condrócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/citologia , Cartilagem/metabolismo , Diferenciação Celular , Linhagem Celular , Condrócitos/metabolismo , Condrogênese , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Transcriptoma
3.
Mol Cell Biochem ; 476(8): 3191-3205, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33864569

RESUMO

Intervertebral disc (IVD) degeneration is an asymptomatic pathophysiological condition and a strong causative factor of low back pain. There is no cure available except spinal fusion and pain management. Stem cell-based regenerative medicine is being considered as an alternative approach to treat disc diseases. The current study aimed to differentiate human umbilical cord-mesenchymal stem cells (hUC-MSCs) into chondrocyte-like cells and to elucidate their feasibility and efficacy in the degenerated IVD rat model. Chondrogenic induction medium was used to differentiate hUC-MSCs into chondroprogenitors. Rat tail IVD model was established with three consecutive coccygeal discs. qPCR was performed to quantify the molecular markers of pain and inflammation. Histological staining was performed to evaluate the degree of regeneration. Induced chondroprogenitors showed the expression of chondrogenic genes, SOX9, TGF-ß1, ACAN, BMP2, and GDF5. Immunocytochemical staining showed positive expression of chondrogenic proteins SOX9, TGF-ß1, TGF-ß2, and Collagen 2. In in vivo study, transplanted chondroprogenitors showed better survival, homing, and distribution in IVD as compared to normal MSCs. Expression of pain and inflammatory genes at day 5 of cell transplantation modulated immune response significantly. The transplanted labeled MSCs and induced chondroprogenitors differentiated into functional nucleus pulposus (NP) cells as evident from co-localization of red (DiI) and green fluorescence for SOX9, TGF-ß1, and TGF-ß2. Alcian blue and H & E staining showed standard histological features, indicating better preservation of the NP structure and cellularity than degenerated discs. hUC-MSCs-derived chondroprogenitors showed better regeneration potential as compared to normal MSCs. The pain and inflammation genes were downregulated in the treated group as compared to the degenerated IVD.


Assuntos
Condrogênese , Inflamação/prevenção & controle , Degeneração do Disco Intervertebral/terapia , Disco Intervertebral/citologia , Células-Tronco Mesenquimais/citologia , Dor/prevenção & controle , Regeneração , Animais , Diferenciação Celular , Humanos , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Degeneração do Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral/patologia , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Dor/etiologia , Dor/metabolismo , Dor/patologia , Ratos , Ratos Wistar , Transdução de Sinais , Cordão Umbilical/citologia
4.
Stem Cells ; 37(1): 102-114, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30358021

RESUMO

Meniscus injuries are among the most common orthopedic injuries. Tears in the inner one-third of the meniscus heal poorly and present a significant clinical challenge. In this study, we hypothesized that progenitor cells from healthy human articular cartilage (chondroprogenitor cells [C-PCs]) may be more suitable than bone-marrow mesenchymal stem cells (BM-MSCs) to mediate bridging and reintegration of fibrocartilage tissue tears in meniscus. C-PCs were isolated from healthy human articular cartilage based on their expression of mesenchymal stem/progenitor marker activated leukocyte cell adhesion molecule (ALCAM) (CD166). Our findings revealed that healthy human C-PCs are CD166+, CD90+, CD54+, CD106- cells with multilineage differentiation potential, and elevated basal expression of chondrogenesis marker SOX-9. We show that, similar to BM-MSCs, C-PCs are responsive to the chemokine stromal cell-derived factor-1 (SDF-1) and they can successfully migrate to the area of meniscal tissue damage promoting collagen bridging across inner meniscal tears. In contrast to BM-MSCs, C-PCs maintained reduced expression of cellular hypertrophy marker collagen X in monolayer culture and in an explant organ culture model of meniscus repair. Treatment of C-PCs with SDF-1/CXCR4 pathway inhibitor AMD3100 disrupted cell localization to area of injury and prevented meniscus tissue bridging thereby indicating that the SDF-1/CXCR4 axis is an important mediator of this repair process. This study suggests that C-PCs from healthy human cartilage may potentially be a useful tool for fibrocartilage tissue repair/regeneration because they resist cellular hypertrophy and mobilize in response to chemokine signaling. Stem Cells 2019;37:102-114.


Assuntos
Cartilagem Articular/efeitos dos fármacos , Condrogênese/genética , Menisco/fisiopatologia , Receptores CXCR4/genética , Animais , Diferenciação Celular , Humanos , Ratos
5.
Connect Tissue Res ; 61(2): 137-151, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-30526130

RESUMO

Bioprinting is a promising tool to fabricate organized cartilage. This study aimed to investigate the printability of gelatin-methacryloyl/gellan gum (gelMA/gellan) hydrogels with and without methacrylated hyaluronic acid (HAMA), and to explore (zone-specific) chondrogenesis of chondrocytes, articular cartilage progenitor cells (ACPCs), and multipotent mesenchymal stromal cells (MSCs) embedded in these bio-inks.The incorporating of HAMA in gelMA/gellan bio-ink increased filament stability, as measured using a filament collapse assay, but did not influence (zone-specific) chondrogenesis of any of the cell types. Highest chondrogenic potential was observed for MSCs, followed by ACPCs, which displayed relatively high proteoglycan IV mRNA levels. Therefore, two-zone constructs were printed with gelMA/gellan/HAMA containing ACPCs in the superficial region and MSCs in the middle/deep region. Chondrogenic differentiation was confirmed, however, printing influence cellular differentiation.ACPC- and MSC-laden gelMA/gellan/HAMA hydrogels are of interest for the fabrication of cartilage constructs. Nevertheless, this study underscores the need for careful evaluation of the effects of printing on cellular differentiation.


Assuntos
Bioimpressão , Cartilagem/metabolismo , Condrócitos/metabolismo , Tinta , Impressão Tridimensional , Células-Tronco/metabolismo , Engenharia Tecidual , Animais , Cartilagem/citologia , Condrócitos/citologia , Cavalos , Células-Tronco/citologia
6.
Osteoarthritis Cartilage ; 24(5): 868-72, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26687821

RESUMO

OBJECTIVE: Total bone marrow-derived mesenchymal stem cell (BMSC) populations differ in their potential to undergo chondrogenesis, with individual BMSCs differing in their chondrogenic capacity. The aim of this study was to explore the use of CD105 as a marker to isolate a chondrogenic subpopulation of BMSCs from the total, heterogeneous population. DESIGN: BMSCs were isolated from patients undergoing total hip replacement and following expansion (Passage 1-Passage 5), CD105 expression was investigated by FACS analysis. FACS was also used to sort BMSCs based on the presence of CD105 (CD105(+)/CD105(-)) or their amount of CD105 expression (CD105(Bright)/CD105(Dim)). After 3 or 5 weeks of differentiation, chondrogenic potential was determined by thionine staining for glycosaminoglycan (GAG) content and by detection of collagen type II using immunohistochemistry. RESULTS: Expanded total BMSC populations were composed almost exclusively of CD105(+) cells, the percentage of which did not correlate to subsequent chondrogenic potential; chondrogenic potential was observed to diminish with culture although CD105 expression remained stable. Similarly, differences in chondrogenic potential were observed between donors despite similar levels of CD105(+) BMSCs. Comparison of CD105(Bright) and CD105(Dim) BMSCs did not reveal a subpopulation with superior chondrogenic potential. CONCLUSIONS: Chondrogenic potential of BMSCs is often linked to CD105 expression. This study demonstrates that CD105 expression on culture expanded BMSC populations does not associate with a chondroprogenitor phenotype and CD105 should not be pursued as a marker to obtain a chondroprogenitor population from BMSCs.


Assuntos
Condrogênese/fisiologia , Endoglina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Condrócitos/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade
7.
Connect Tissue Res ; 56(4): 265-71, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26075411

RESUMO

Chondroprogenitor cells are a subpopulation of multipotent progenitors that are primed for chondrogenesis. They are believed to have the biological repertoire to be ideal for cell-based cartilage therapy. In addition to summarizing recent advances in chondroprogenitor cell characterization, this review discusses the projected pros and cons of utilizing chondroprogenitors in regenerative medicine and compares them with that of pre-existing methods, including autologous chondrocyte implantation (ACI) and the utilization of bone marrow derived mesenchymal stem cells (MSCs) for the purpose of cartilage tissue repair.


Assuntos
Condrócitos/transplante , Condrogênese , Células-Tronco Multipotentes/transplante , Medicina Regenerativa/métodos , Animais , Autoenxertos , Condrócitos/metabolismo , Humanos , Células-Tronco Multipotentes/metabolismo
8.
Front Bioeng Biotechnol ; 12: 1421111, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39391600

RESUMO

Introduction: The ability to bioprint facial cartilages could revolutionise reconstructive surgery, but identifying the optimum cell source remains one of the great challenges of tissue engineering. Tissue specific stem cells: chondroprogenitors, have been extracted previously using preferential adhesion to fibronectin based on the expression of CD49e: a perceived chondroprogenitor stem cell marker present on <1% of cartilage cells. This study sought to determine whether these fibronectin-adherent chondroprogenitor cells could be exploited for cartilage tissue engineering applications in isolation, or combined with differentiated chondrocytes. Methods: Nasoseptal cartilage samples from 20 patients (10 male, 10 female) were digested to liberate cartilage-derived cells (CDCs) from extracellular matrix. Total cell number was counted using the Trypan Blue exclusion assay and added to fibronectin coated plates for 20 min, to determine the proportion of fibronectin-adherent (FAC) and non-adherent cells (NFACs). All populations underwent flow cytometry to detect mesenchymal stem/progenitor cell markers and were cultured in osteogenic, chondrogenic and adipogenic media to determine trilineage differentiation potential. Cell adherence and growth kinetics of the different populations were compared using iCELLigence growth assays. Chondrogenic gene expression was assessed using RT-qPCR for Type 2 collagen, aggrecan and SOX9 genes. Varying proportions of NFAC and FACs were cultured in alginate beads to assess tissue engineering potential. Results: 52.6% of cells were fibronectin adherent in males and 57.7% in females, yet on flow cytometrical analysis, only 0.19% of cells expressed CD49e. Moreover, all cells (CDC, FAC and NFACs) demonstrated an affinity for trilineage differentiation by first passage and the expression of stem/progenitor cell markers increased significantly from digest to first passage (CD29, 44, 49e, 73 and 90, p < 0.0001). No significant differences were seen in adhesion or growth rates. Collagen and aggrecan gene expression was higher in FACs than CDCs (2-fold higher, p = 0.008 and 0.012 respectively), but no differences in chondrogenic potential were seen in any cell mixtures in 3D culture models. Conclusion: The fibronectin adhesion assay does not appear to reliably isolate a chondroprogenitor cell population from nasoseptal cartilage, and these cells confer no advantageous properties for cartilage tissue engineering. Refinement of cell isolation methods and chondroprogenitor markers is warranted for future nasoseptal cartilage tissue engineering efforts.

9.
Cell Stem Cell ; 30(9): 1179-1198.e7, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37683603

RESUMO

Osteoarthritis is a degenerative joint disease that causes pain, degradation, and dysfunction. Excessive canonical Wnt signaling in osteoarthritis contributes to chondrocyte phenotypic instability and loss of cartilage homeostasis; however, the regulatory niche is unknown. Using the temporomandibular joint as a model in multiple species, we identify Lgr5-expressing secretory cells as forming a Wnt inhibitory niche that instruct Wnt-inactive chondroprogenitors to form the nascent synovial joint and regulate chondrocyte lineage and identity. Lgr5 ablation or suppression during joint development, aging, or osteoarthritis results in depletion of Wnt-inactive chondroprogenitors and a surge of Wnt-activated, phenotypically unstable chondrocytes with osteoblast-like properties. We recapitulate the cartilage niche and create StemJEL, an injectable hydrogel therapy combining hyaluronic acid and sclerostin. Local delivery of StemJEL to post-traumatic osteoarthritic jaw and knee joints in rabbit, rat, and mini-pig models restores cartilage homeostasis, chondrocyte identity, and joint function. We provide proof of principal that StemJEL preserves the chondrocyte niche and alleviates osteoarthritis.


Assuntos
Condrócitos , Osteoartrite , Suínos , Animais , Coelhos , Ratos , Porco Miniatura , Cartilagem , Envelhecimento , Receptores Acoplados a Proteínas G
10.
Biomed Mater ; 18(6)2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37827163

RESUMO

With the advancement of tissue engineering technologies, implantable materials have been developed for use in facial plastic surgery, including auriculoplasty and rhinoplasty. Tissue-engineered cartilage comprising only cells and cell-produced extracellular matrix is considered valuable as there is no need to consider problems associated with scaffold absorption or immune responses commonly related to conventional artificial materials. However, it is exceedingly difficult to produce large-sized complex shapes of cartilage without the use of scaffolds. In this study, we describe the production of shape-designable cartilage using a novel cell self-aggregation technique (CAT) and chondroprogenitor cells derived from human induced pluripotent stem cells as the source. The method described does not require special equipment such as bio-3D printers, and the produced tissue can be induced into well-matured cartilage with abundant cartilage matrixin vitro. Using CAT, we were able to generate cartilage in the form of rings or tubes with adjustable inner diameter and curvature, over a range of several centimeters, without the use of scaffolds. Thein vitrofabrication of shape-designable cartilage using CAT is a promising development in facial plastic surgery.


Assuntos
Células-Tronco Pluripotentes Induzidas , Alicerces Teciduais , Humanos , Cartilagem/fisiologia , Engenharia Tecidual/métodos , Matriz Extracelular , Condrogênese
11.
J Tissue Eng ; 13: 20417314221143484, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36582939

RESUMO

Morphologically stable scaffold-free elastic cartilage tissue is crucial for treating external ear abnormalities. However, establishing adequate mechanical strength is challenging, owing to the difficulty of achieving chondrogenic differentiation in vitro; thus, cartilage reconstruction is a complex task. Auricular perichondrial chondroprogenitor cells exhibit high proliferation potential and can be obtained with minimal invasion. Therefore, these cells are an ideal resource for elastic cartilage reconstruction. In this study, we aimed to develop a novel in vitro scaffold-free method for elastic cartilage reconstruction, using human auricular perichondrial chondroprogenitor cells. Inducing chondrogenesis by using microscopic spheroids similar to auricular hillocks significantly increased the chondrogenic potential. The size and elasticity of the tissue were maintained after craniofacial transplantation in immunodeficient mice, suggesting that the reconstructed tissue was morphologically stable. Our novel tissue reconstruction method may facilitate the development of future treatments for external ear abnormalities.

12.
Regen Med ; 17(9): 659-675, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35703020

RESUMO

Cartilage defects trouble millions of patients worldwide and their repair via conventional treatment is difficult. Excitingly, tissue engineering technology provides a promising strategy for efficient cartilage regeneration with structural regeneration and functional reconstruction. Seed cells, as biological prerequisites for cartilage regeneration, determine the quality of regenerated cartilage. The proliferation, differentiation and chondrogenesis of seed cells are greatly affected by their type, origin and generation. Thus, a systematic description of the characteristics of seed cells is necessary. This article reviews in detail the cellular characteristics, research progress, clinical translation challenges and future research directions of seed cells while providing guidelines for selecting appropriate seed cells for cartilage regeneration.


Cartilage defects affect millions of patients worldwide and their repair via conventional treatment is quite difficult. Excitingly, tissue engineering technology provides a promising strategy for efficient cartilage regeneration. The seed cell, as a biological prerequisite for cartilage regeneration, determines the quality of regenerated cartilage. This article reviews in detail the cellular characteristics, research progress, clinical translation challenges and future research directions of various chondrocytes, chondroprogenitor cells and stem cells. Chondrocytes, especially elastic chondrocytes, could complete subcutaneous cartilage regeneration, whereas stem cells are superior for composite defects, allografts and cartilage defects caused by inflammation. In brief, this article provides a guide for selecting appropriate seed cells for cartilage regeneration.


Assuntos
Cartilagem Articular , Células-Tronco Mesenquimais , Cartilagem , Diferenciação Celular , Condrócitos , Condrogênese , Humanos , Regeneração , Engenharia Tecidual , Alicerces Teciduais
13.
Bone ; 137: 115368, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32380258

RESUMO

Wnt signaling together with other signaling pathways governs cartilage development and the growth plate function during long bone formation and growth. ß-catenin-dependent Wnt signaling is a specific lineage determinant of skeletal mesenchymal cells toward chondrogenic or osteogenic direction. Once cartilage forms and the growth plate organize, Wnt signaling continues to regulate proliferation and differentiation of the growth plate chondrocytes. Although chondrocytes in the growth plate have a high capacity to proliferate, new cells must be supplied to the growth plate from chondroprogenitor population. Advances in in vivo cell tracking techniques have demonstrated the importance of Wnt signaling in driving tissue renewal. The Wnt-responsive cells, genetically marked by the Wnt-reporter system, are found as stem cells in various tissues. Similarly, Wnt-responsive cells are found in the periphery of the growth plate and expanded to constitute entire column structure, indicating that Wnt signaling participates in the regulation of chondroprogenitors in the growth plate. This review will discuss advancements in research of progenitors in the growth plate, specifically focusing on Wnt/ß-catenin signaling.


Assuntos
Desenvolvimento Ósseo , Condrogênese , Via de Sinalização Wnt , Animais , Diferenciação Celular , Condrócitos/metabolismo , Lâmina de Crescimento/metabolismo , Humanos , beta Catenina/metabolismo
14.
Knee ; 27(3): 755-759, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32563433

RESUMO

BACKGROUND: Enhanced chondrogenesis and reduction in hypertrophy are essential pre-requisites for cell-based therapy in regenerative research for cartilage loss. Chondroprogenitors, isolated by fibronectin adhesion assay (FAA), have shown promising results in various preclinical studies due to their inherent characteristics. However, the need for monolayer culture and the effect of expansion on cell phenotype render differentiation between chondroprogenitors and chondrocytes (native cartilage cells) difficult. This is further complicated due to reported de-differentiation of chondrocytes in culture. Thus, the aim of our study was to harvest cells from articular cartilage and compare their gene expression to cells demonstrating adherence and non-adherence to fibronectin. METHOD: Fresh-cells (FC) were isolated from human osteoarthritic knee joints(n = 3) and subjected to FAA. Cells unbound to fibronectin (20 min after plating) were termed as FAA-ve. Attached cells were further cultured for five population doublings and designated FAA+ve. RNA from all three cell groups was assessed for SOX-9, ACAN, COL2A1, COL1A1, RUNX2 and COL10A1. RESULTS: All three groups exhibited moderate to high expression of markers of chondrogenesis and marker of chondrocyte hypertrophy. FAA+ve group exhibited significantly lower levels of hypertrophy markers: RUNX2 (vs FC and FAA-ve, P = 0.018) and COL10A1(vs FAA-ve, P = 0.005). CONCLUSIONS: Our results demonstrated that fibronectin effectively isolated cells distinct from mature chondrocytes in terms of reduced hypertrophic tendency. This is noteworthy as cells isolated by FAA, retaining their inherent progenitor phenotype, with upregulation of chondrogenic markers may be used successfully for cartilage repair in future translational work.


Assuntos
Cartilagem Articular/metabolismo , Condrócitos/citologia , Condrogênese/genética , DNA/genética , Fibronectinas/genética , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Condrócitos/metabolismo , Feminino , Fibronectinas/biossíntese , Humanos , Masculino , Pessoa de Meia-Idade
15.
Stem Cell Res Ther ; 11(1): 66, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32070421

RESUMO

BACKGROUND: Articular cartilage shows little or no capacity for intrinsic repair, generating a critical need of regenerative therapies for joint injuries and diseases such as osteoarthritis. Human-induced pluripotent stem cells (hiPSCs) offer a promising cell source for cartilage tissue engineering and in vitro human disease modeling; however, off-target differentiation remains a challenge during hiPSC chondrogenesis. Therefore, the objective of this study was to identify cell surface markers that define the true chondroprogenitor population and use these markers to purify iPSCs as a means of improving the homogeneity and efficiency of hiPSC chondrogenic differentiation. METHODS: We used a CRISPR-Cas9-edited COL2A1-GFP knock-in reporter hiPSC line, coupled with a surface marker screen, to identify a novel chondroprogenitor population. Single-cell RNA sequencing was then used to analyze the distinct clusters within the population. An unpaired t test with Welch's correction or an unpaired Kolmogorov-Smirnov test was performed with significance reported at a 95% confidence interval. RESULTS: Chondroprogenitors expressing CD146, CD166, and PDGFRß, but not CD45, made up an average of 16.8% of the total population. Under chondrogenic culture conditions, these triple-positive chondroprogenitor cells demonstrated decreased heterogeneity as measured by single-cell RNA sequencing with fewer clusters (9 clusters in unsorted vs. 6 in sorted populations) closer together. Additionally, there was more robust and homogenous matrix production (unsorted: 1.5 ng/ng vs. sorted: 19.9 ng/ng sGAG/DNA; p < 0.001) with significantly higher chondrogenic gene expression (i.e., SOX9, COL2A1, ACAN; p < 0.05). CONCLUSIONS: Overall, this study has identified a unique hiPSC-derived subpopulation of chondroprogenitors that are CD146+/CD166+/PDGFRß+/CD45- and exhibit high chondrogenic potential, providing a purified cell source for cartilage tissue engineering or disease modeling studies.


Assuntos
Sistemas CRISPR-Cas/genética , Condrócitos/metabolismo , Condrogênese/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Estudos Prospectivos
16.
Stem Cell Res Ther ; 11(1): 111, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32160923

RESUMO

Cartilage-derived pluripotent cells reside in hyaline cartilage and fibrocartilage. These cells have the potential for multidirectional differentiation; can undergo adipogenesis, osteogenesis, and chondrogenesis; and have been classified as mesenchymal stem cells (MSCs) conforming to the minimal criteria of the International Society for Cellular Therapy. Cartilage tissue is prone to injury and is difficult to repair. As cartilage-derived pluripotent cells are the closest cell source to cartilage tissue, they are expected to have the strongest ability to differentiate into cartilage compared to other MSCs. This review focuses on the organizational distribution, expression, and function of cartilage-derived pluripotent cells in joint development and repair to help explore the therapeutic potential of in situ cartilage-derived pluripotent cells for joint cartilage repair.


Assuntos
Cartilagem Articular , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Diferenciação Celular , Condrogênese , Osteogênese
17.
Stem Cell Res Ther ; 11(1): 177, 2020 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-32408888

RESUMO

BACKGROUND: Since cartilage-derived stem/progenitor cells (CSPCs) were first identified in articular cartilage using differential adhesion to fibronectin, their self-renewal capacity and niche-specific lineage preference for chondrogenesis have propelled their application for cartilage tissue engineering. In many adult tissues, stem/progenitor cells are recognised to be involved in tissue homeostasis. However, the role of nasoseptal CSPCs has not yet been elucidated. Our aim was to isolate and characterise nasoseptal CSPCs alongside nasoseptal chondrocyte populations and determine chondrogenic capacity. METHODS: Here, we isolated nasoseptal CSPCs using differential adhesion to fibronectin and assessed their colony forming efficiency, proliferation kinetics, karyotype and trilineage potential. CSPCs were characterised alongside non-fibronectin-adherent nasoseptal chondrocytes (DNCs) and cartilage-derived cells (CDCs, a heterogenous combination of DNCs and CSPCs) by assessing differences in gene expression profiles using PCR Stem Cell Array, immunophenotype using flow cytometry and chondrogencity using RT-PCR and histology. RESULTS: CSPCs were clonogenic with increased gene expression of the neuroectodermal markers NCAM1 and N-Cadherin, as well as Cyclins D1 and D2, compared to DNCs. All three cell populations expressed recognised mesenchymal stem cell surface markers (CD29, CD44, CD73, CD90), yet only CSPCs and CDCs showed multilineage differentiation potential. CDC populations expressed significantly higher levels of type 2 collagen and bone morphogenetic protein 2 genes, with greater cartilage extracellular matrix secretion. When DNCs were cultured in isolation, there was reduced chondrogenicity and higher expression of type 1 collagen, stromal cell-derived factor 1 (SDF-1), CD73 and CD90, recognised markers of a fibroblast-like phenotype. CONCLUSIONS: Fibronectin-adherent CSPCs demonstrate a unique gene expression profile compared to non-fibronectin-adherent DNCs. DNCs cultured in isolation, without CSPCs, express fibroblastic phenotype with reduced chondrogenicity. Mixed populations of stem/progenitor cells and chondrocytes were required for optimal chondrogenesis, suggesting that CSPCs may be required to retain phenotypic stability and chondrogenic potential of DNCs. Crosstalk between DNCs and CSPCs is proposed based on SDF-1 signalling.


Assuntos
Cartilagem Articular , Células-Tronco Mesenquimais , Diferenciação Celular , Células Cultivadas , Condrócitos , Condrogênese/genética , Células-Tronco
18.
ACS Appl Mater Interfaces ; 11(45): 41906-41924, 2019 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-31639302

RESUMO

Graphene foam holds promise for tissue engineering applications. In this study, graphene foam was used as a three-dimension scaffold to evaluate cell attachment, cell morphology, and molecular markers of early differentiation. The aim of this study was to determine if cell attachment and elaboration of an extracellular matrix would be modulated by functionalization of graphene foam with fibronectin, an extracellular matrix protein that cells adhere well to, prior to the establishment of three-dimensional cell culture. The molecular dynamic simulation demonstrated that the fibronectin-graphene interaction was stabilized predominantly through interaction between the graphene and arginine side chains of the protein. Quasi-static and dynamic mechanical testing indicated that fibronectin functionalization of graphene altered the mechanical properties of graphene foam. The elastic strength of the scaffold increased due to fibronectin, but the viscoelastic mechanical behavior remained unchanged. An additive effect was observed in the mechanical stiffness when the graphene foam was both coated with fibronectin and cultured with cells for 28 days. Cytoskeletal organization assessed by fluorescence microscopy demonstrated a fibronectin-dependent reorganization of the actin cytoskeleton and an increase in actin stress fibers. Gene expression assessed by quantitative real-time polymerase chain reaction of 9 genes encoding cell attachment proteins (Cd44, Ctnna1, Ctnnb1, Itga3, Itga5, Itgav, Itgb1, Ncam1, Sgce), 16 genes encoding extracellular matrix proteins (Col1a1, Col2a1, Col3a1, Col5a1, Col6a1, Ecm1, Emilin1, Fn1, Hapln1, Lamb3, Postn, Sparc, Spp1, Thbs1, Thbs2, Tnc), and 9 genes encoding modulators of remodeling (Adamts1, Adamts2, Ctgf, Mmp14, Mmp2, Tgfbi, Timp1, Timp2, Timp3) indicated that graphene foam provided a microenvironment conducive to expression of genes that are important in early chondrogenesis. Functionalization of graphene foam with fibronectin modified the cellular response to graphene foam, demonstrated by decreases in relative gene expression levels. These findings illustrate the combinatorial factors of microscale materials properties and nanoscale molecular features to consider in the design of three-dimensional graphene scaffolds for tissue engineering applications.


Assuntos
Condrócitos/citologia , Condrogênese , Fibronectinas/metabolismo , Grafite/química , Engenharia Tecidual/instrumentação , Alicerces Teciduais/química , Animais , Adesão Celular , Moléculas de Adesão Celular , Técnicas de Cultura de Células , Condrócitos/metabolismo , Matriz Extracelular/química , Fibronectinas/química , Camundongos
19.
J Tissue Eng Regen Med ; 12(1): e579-e591, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-27690334

RESUMO

Intervertebral disc (IVD) degeneration is characterized by the loss of nucleus pulposus (NP), which is a common cause for lower back pain. Although, currently, there is no cure for the degenerative disc disease, stem cell therapy is increasingly being considered for its treatment. In this study, we investigated the feasibility and efficacy of human umbilical cord mesenchymal stem cells (MSCs) and chondroprogenitor cells (CPCs) derived from those cells to regenerate damaged IVD in a rabbit model. Transplanted cells survived, engrafted and dispersed into NP in situ. Significant improvement in the histology, cellularity, extracellular matrix proteins, and water and glycosaminoglycan contents in IVD recipients of CPCs was observed compared to MSCs. In addition, IVDs receiving CPCs exhibited higher expression of NP-specific human markers, SOX9, aggrecan, collagen 2, FOXF1 and KRT19. The novelty of the study is that in vitro differentiated CPCs derived from umbilical cord MSCs, demonstrated far greater capacity to regenerate damaged IVDs, which provides basis and impetus for stem cell based clinical studies to treat degenerative disc disease. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Disco Intervertebral/fisiopatologia , Regeneração , Cordão Umbilical/citologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Condrogênese , Feminino , Humanos , Disco Intervertebral/patologia , Degeneração do Disco Intervertebral/patologia , Degeneração do Disco Intervertebral/fisiopatologia , Degeneração do Disco Intervertebral/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Núcleo Pulposo/patologia , Núcleo Pulposo/fisiopatologia , Coelhos
20.
Acta Biomater ; 61: 41-53, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28782725

RESUMO

Cell-laden hydrogels are the primary building blocks for bioprinting, and, also termed bioinks, are the foundations for creating structures that can potentially recapitulate the architecture of articular cartilage. To be functional, hydrogel constructs need to unlock the regenerative capacity of encapsulated cells. The recent identification of multipotent articular cartilage-resident chondroprogenitor cells (ACPCs), which share important traits with adult stem cells, represents a new opportunity for cartilage regeneration. However, little is known about the suitability of ACPCs for tissue engineering, especially in combination with biomaterials. This study aimed to investigate the potential of ACPCs in hydrogels for cartilage regeneration and biofabrication, and to evaluate their ability for zone-specific matrix production. Gelatin methacryloyl (gelMA)-based hydrogels were used to culture ACPCs, bone marrow mesenchymal stromal cells (MSCs) and chondrocytes, and as bioinks for printing. Our data shows ACPCs outperformed chondrocytes in terms of neo-cartilage production and unlike MSCs, ACPCs had the lowest gene expression levels of hypertrophy marker collagen type X, and the highest expression of PRG4, a key factor in joint lubrication. Co-cultures of the cell types in multi-compartment hydrogels allowed generating constructs with a layered distribution of collagens and glycosaminoglycans. By combining ACPC- and MSC-laden bioinks, a bioprinted model of articular cartilage was generated, consisting of defined superficial and deep regions, each with distinct cellular and extracellular matrix composition. Taken together, these results provide important information for the use of ACPC-laden hydrogels in regenerative medicine, and pave the way to the biofabrication of 3D constructs with multiple cell types for cartilage regeneration or in vitro tissue models. STATEMENT OF SIGNIFICANCE: Despite its limited ability to repair, articular cartilage harbors an endogenous population of progenitor cells (ACPCs), that to date, received limited attention in biomaterials and tissue engineering applications. Harnessing the potential of these cells in 3D hydrogels can open new avenues for biomaterial-based regenerative therapies, especially with advanced biofabrication technologies (e.g. bioprinting). This study highlights the potential of ACPCs to generate neo-cartilage in a gelatin-based hydrogel and bioink. The ACPC-laden hydrogel is a suitable substrate for chondrogenesis and data shows it has a bias in directing cells towards a superficial zone phenotype. For the first time, ACPC-hydrogels are evaluated both as alternative for and in combination with chondrocytes and MSCs, using co-cultures and bioprinting for cartilage regeneration in vitro. This study provides important cues on ACPCs, indicating they represent a promising cell source for the next generation of cartilage constructs with increased biomimicry.


Assuntos
Bioimpressão/métodos , Cartilagem Articular/citologia , Hidrogéis/farmacologia , Tinta , Regeneração/efeitos dos fármacos , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Condrogênese/efeitos dos fármacos , Condrogênese/genética , Técnicas de Cocultura , Força Compressiva , DNA/metabolismo , Glicosaminoglicanos/metabolismo , Cavalos , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células-Tronco/efeitos dos fármacos , Sus scrofa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA