Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Stem Cells ; 39(11): 1447-1456, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34427363

RESUMEN

Transient receptor potential vanilloid 4 (TRPV4) is a polymodal calcium-permeable cation channel that is highly expressed in cartilage and is sensitive to a variety of extracellular stimuli. The expression of this channel has been associated with the process of chondrogenesis in adult stem cells as well as several cell lines. Here, we used a chondrogenic reporter (Col2a1-GFP) in murine induced pluripotent stem cells (iPSCs) to examine the hypothesis that TRPV4 serves as both a marker and a regulator of chondrogenesis. Over 21 days of chondrogenesis, iPSCs showed significant increases in Trpv4 expression along with the standard chondrogenic gene markers Sox9, Acan, and Col2a1, particularly in the green fluorescent protein positive (GFP+) chondroprogenitor subpopulation. Increased gene expression for Trpv4 was also reflected by the presence of TRPV4 protein and functional Ca2+ signaling. Daily activation of TRPV4 using the specific agonist GSK1016790A resulted in significant increases in cartilaginous matrix production. An improved understanding of the role of TRPV4 in chondrogenesis may provide new insights into the development of new therapeutic approaches for diseases of cartilage, such as osteoarthritis, or channelopathies and hereditary disorders that affect cartilage during development. Harnessing the role of TRPV4 in chondrogenesis may also provide a novel approach for accelerating stem cell differentiation in functional tissue engineering of cartilage replacements for joint repair.


Asunto(s)
Condrogénesis , Células Madre Pluripotentes Inducidas , Canales Catiónicos TRPV , Animales , Cartílago/metabolismo , Diferenciación Celular , Células Cultivadas , Condrocitos , Condrogénesis/genética , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
2.
Stem Cells ; 37(1): 65-76, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30378731

RESUMEN

The differentiation of human induced pluripotent stem cells (hiPSCs) to prescribed cell fates enables the engineering of patient-specific tissue types, such as hyaline cartilage, for applications in regenerative medicine, disease modeling, and drug screening. In many cases, however, these differentiation approaches are poorly controlled and generate heterogeneous cell populations. Here, we demonstrate cartilaginous matrix production in three unique hiPSC lines using a robust and reproducible differentiation protocol. To purify chondroprogenitors (CPs) produced by this protocol, we engineered a COL2A1-GFP knock-in reporter hiPSC line by CRISPR-Cas9 genome editing. Purified CPs demonstrated an improved chondrogenic capacity compared with unselected populations. The ability to enrich for CPs and generate homogenous matrix without contaminating cell types will be essential for regenerative and disease modeling applications. Stem Cells 2019;37:65-76.


Asunto(s)
Sistemas CRISPR-Cas/genética , Condrogénesis/genética , Edición Génica/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Alelos , Diferenciación Celular , Humanos
3.
Proc Natl Acad Sci U S A ; 109(47): 19172-7, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23115336

RESUMEN

The development of regenerative therapies for cartilage injury has been greatly aided by recent advances in stem cell biology. Induced pluripotent stem cells (iPSCs) have the potential to provide an abundant cell source for tissue engineering, as well as generating patient-matched in vitro models to study genetic and environmental factors in cartilage repair and osteoarthritis. However, both cell therapy and modeling approaches require a purified and uniformly differentiated cell population to predictably recapitulate the physiological characteristics of cartilage. Here, iPSCs derived from adult mouse fibroblasts were chondrogenically differentiated and purified by type II collagen (Col2)-driven green fluorescent protein (GFP) expression. Col2 and aggrecan gene expression levels were significantly up-regulated in GFP+ cells compared with GFP- cells and decreased with monolayer expansion. An in vitro cartilage defect model was used to demonstrate integrative repair by GFP+ cells seeded in agarose, supporting their potential use in cartilage therapies. In chondrogenic pellet culture, cells synthesized cartilage-specific matrix as indicated by high levels of glycosaminoglycans and type II collagen and low levels of type I and type X collagen. The feasibility of cell expansion after initial differentiation was illustrated by homogenous matrix deposition in pellets from twice-passaged GFP+ cells. Finally, atomic force microscopy analysis showed increased microscale elastic moduli associated with collagen alignment at the periphery of pellets, mimicking zonal variation in native cartilage. This study demonstrates the potential use of iPSCs for cartilage defect repair and for creating tissue models of cartilage that can be matched to specific genetic backgrounds.


Asunto(s)
Cartílago/fisiología , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ingeniería de Tejidos/métodos , Animales , Diferenciación Celular/genética , Separación Celular , Reprogramación Celular/genética , Condrogénesis/genética , Colágeno Tipo II/metabolismo , Módulo de Elasticidad , Citometría de Flujo , Regulación de la Expresión Génica , Glicosaminoglicanos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Microscopía de Fuerza Atómica , Especificidad de Órganos , Sefarosa , Cicatrización de Heridas
4.
J Biomech Eng ; 134(1): 011011, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22482666

RESUMEN

Understanding structure-function relationships in the temporomandibular joint (TMJ) disc is a critical first step toward creating functional tissue replacements for the large population of patients suffering from TMJ disc disorders. While many of these relationships have been identified for the collagenous fraction of the disc, this same understanding is lacking for the next most abundant extracellular matrix component, sulfated glycosaminoglycans (GAGs). Though GAGs are known to play a major role in maintaining compressive integrity in GAG-rich tissues such as articular cartilage, their role in fibrocartilaginous tissues in which GAGs are much less abundant is not clearly defined. Therefore, this study investigates the contribution of GAGs to the regional viscoelastic compressive properties of the temporomandibular joint (TMJ) disc. Chondroitinase ABC (C-ABC) was used to deplete GAGs in five different disc regions, and the time course for >95% GAG removal was defined. The compressive properties of GAG depleted regional specimens were then compared to non-treated controls using an unconfined compression stress-relaxation test. Additionally, treated and non-treated specimens were assayed biochemically and histologically to confirm GAG removal. Compared to untreated controls, the only regions affected by GAG removal in terms of biomechanical properties were in the intermediate zone, the most GAG-rich portion of the disc. Without GAGs, all intermediate zone regions showed decreased tissue viscosity, and the intermediate zone lateral region also showed a 12.5% decrease in modulus of relaxation. However, in the anterior and posterior band regions, no change in compressive properties was observed following GAG depletion, though these regions showed the highest compressive properties overall. Although GAGs are not the major extracellular matrix molecule of the TMJ disc, they are responsible for some of the viscoelastic compressive properties of the tissue. Furthermore, the mechanical role of sulfated GAGs in the disc varies regionally in the tissue, and GAG abundance does not always correlate with higher compressive properties. Overall, this study found that sulfated GAGs are important to TMJ disc mechanics in the intermediate zone, an important finding for establishing design characteristics for future tissue engineering efforts.


Asunto(s)
Fuerza Compresiva , Glicosaminoglicanos/metabolismo , Disco de la Articulación Temporomandibular/metabolismo , Animales , Fenómenos Biomecánicos , Condroitín/metabolismo , Condroitina ABC Liasa/metabolismo , Dermatán Sulfato/metabolismo , Módulo de Elasticidad , Elasticidad , Femenino , Porcinos , Viscosidad
5.
Sci Adv ; 7(38): eabi5918, 2021 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-34524840

RESUMEN

Articular cartilage has unique load-bearing properties but has minimal capacity for intrinsic repair. Here, we used three-dimensional weaving, additive manufacturing, and autologous mesenchymal stem cells to create a tissue-engineered, bicomponent implant to restore hip function in a canine hip osteoarthritis model. This resorbable implant was specifically designed to function mechanically from the time of repair and to biologically integrate with native tissues for long-term restoration. A massive osteochondral lesion was created in the hip of skeletally mature hounds and repaired with the implant or left empty (control). Longitudinal outcome measures over 6 months demonstrated that the implant dogs returned to normal preoperative values of pain and function. Anatomical structure and functional biomechanical properties were also restored in the implanted dogs. Control animals never returned to normal and exhibited structurally deficient repair. This study provides clinically relevant evidence that the bicomponent implant may be a potential therapy for moderate hip osteoarthritis.

6.
Stem Cell Res Ther ; 9(1): 61, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29523190

RESUMEN

BACKGROUND: Intervertebral disc (IVD) degeneration is characterized by an early decrease in cellularity of the nucleus pulposus (NP) region, and associated extracellular matrix changes, reduced hydration, and progressive degeneration. Cell-based IVD therapy has emerged as an area of great interest, with studies reporting regenerative potential for many cell sources, including autologous or allogeneic chondrocytes, primary IVD cells, and stem cells. Few approaches, however, have clear strategies to promote the NP phenotype, in part due to a limited knowledge of the defined markers and differentiation protocols for this lineage. Here, we developed a new protocol for the efficient differentiation of human induced pluripotent stem cells (hiPSCs) into NP-like cells in vitro. This differentiation strategy derives from our knowledge of the embryonic notochordal lineage of NP cells as well as strategies used to support healthy NP cell phenotypes for primary cells in vitro. METHODS: An NP-genic phenotype of hiPSCs was promoted in undifferentiated hiPSCs using a stepwise, directed differentiation toward mesodermal, and subsequently notochordal, lineages via chemically defined medium and growth factor supplementation. Fluorescent cell imaging was used to test for pluripotency markers in undifferentiated cells. RT-PCR was used to test for potential cell lineages at the early stage of differentiation. Cells were checked for NP differentiation using immunohistochemistry and histological staining at the end of differentiation. To enrich notochordal progenitor cells, hiPSCs were transduced using lentivirus containing reporter constructs for transcription factor brachyury (T) promoter and green fluorescent protein (GFP) fluorescence, and then sorted on T expression based on GFP intensity by flow cytometry. RESULTS: Periods of pellet culture following initial induction were shown to promote the vacuolated NP cell morphology and NP surface marker expression, including CD24, LMα5, and Basp1. Enrichment of brachyury (T) positive cells using fluorescence-activated cell sorting was shown to further enhance the differentiation efficiency of NP-like cells. CONCLUSIONS: The ability to efficiently differentiate human iPSCs toward NP-like cells may provide insights into the processes of NP cell differentiation and provide a cell source for the development of new therapies for IVD diseases.


Asunto(s)
Condrocitos/citología , Células Madre Pluripotentes Inducidas/citología , Núcleo Pulposo/citología , Diferenciación Celular , Técnicas de Reprogramación Celular/métodos , Humanos
7.
Trends Mol Med ; 23(10): 917-931, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28887050

RESUMEN

Arthritis represents a family of complex joint pathologies responsible for the majority of musculoskeletal conditions. Nearly all diseases within this family, including osteoarthritis, rheumatoid arthritis, and juvenile idiopathic arthritis, are chronic conditions with few or no disease-modifying therapeutics available. Advances in genome engineering technology, most recently with CRISPR-Cas9, have revolutionized our ability to interrogate and validate genetic and epigenetic elements associated with chronic diseases such as arthritis. These technologies, together with cell reprogramming methods, including the use of induced pluripotent stem cells, provide a platform for human disease modeling. We summarize new evidence from genome-wide association studies and genomics that substantiates a genetic basis for arthritis pathogenesis. We also review the potential contributions of genome engineering in the development of new arthritis therapeutics.


Asunto(s)
Artritis , Sistemas CRISPR-Cas , Edición Génica/métodos , Genoma Humano , Estudio de Asociación del Genoma Completo , Medicina de Precisión/métodos , Animales , Artritis/genética , Artritis/terapia , Humanos
8.
Stem Cell Reports ; 8(5): 1202-1213, 2017 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-28457885

RESUMEN

Chronic inflammatory diseases such as arthritis are characterized by dysregulated responses to pro-inflammatory cytokines such as interleukin-1 (IL-1) and tumor necrosis factor α (TNF-α). Pharmacologic anti-cytokine therapies are often effective at diminishing this inflammatory response but have significant side effects and are used at high, constant doses that do not reflect the dynamic nature of disease activity. Using the CRISPR/Cas9 genome-engineering system, we created stem cells that antagonize IL-1- or TNF-α-mediated inflammation in an autoregulated, feedback-controlled manner. Our results show that genome engineering can be used successfully to rewire endogenous cell circuits to allow for prescribed input/output relationships between inflammatory mediators and their antagonists, providing a foundation for cell-based drug delivery or cell-based vaccines via a rapidly responsive, autoregulated system. The customization of intrinsic cellular signaling pathways in stem cells, as demonstrated here, opens innovative possibilities for safer and more effective therapeutic approaches for a wide variety of diseases.


Asunto(s)
Edición Génica/métodos , Factores Inmunológicos/genética , Células Madre Pluripotentes Inducidas/metabolismo , Trasplante de Células Madre/métodos , Animales , Artritis/terapia , Sistemas CRISPR-Cas , Cartílago/fisiología , Células Cultivadas , Retroalimentación Fisiológica , Factores Inmunológicos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Interleucina-1/genética , Interleucina-1/metabolismo , Ratones , Ratones Endogámicos C57BL , Regeneración , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
9.
Arthritis Rheumatol ; 69(5): 1111-1121, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27813286

RESUMEN

OBJECTIVE: Proinflammatory cytokines such as interleukin-1 (IL-1) are found in elevated levels in diseased or injured tissues and promote rapid tissue degradation while preventing stem cell differentiation. This study was undertaken to engineer inflammation-resistant murine induced pluripotent stem cells (iPSCs) through deletion of the IL-1 signaling pathway and to demonstrate the utility of these cells for engineering replacements for diseased or damaged tissues. METHODS: Targeted deletion of the IL-1 receptor type I (IL-1RI) gene in murine iPSCs was achieved using the RNA-guided, site-specific clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 genome engineering system. Clonal cell populations with homozygous and heterozygous deletions were isolated, and loss of receptor expression and cytokine signaling was confirmed by flow cytometry and transcriptional reporter assays, respectively. Cartilage was engineered from edited iPSCs and tested for its ability to resist IL-1-mediated degradation in gene expression, histologic, and biomechanical assays after a 3-day treatment with 1 ng/ml of IL-1α. RESULTS: Three of 41 clones isolated possessed the IL-1RI+/- genotype. Four clones possessed the IL-1RI-/- genotype, and flow cytometry confirmed loss of IL-1RI on the surface of these cells, which led to an absence of NF-κB transcription activation after IL-1α treatment. Cartilage engineered from homozygous null clones was resistant to cytokine-mediated tissue degradation. In contrast, cartilage derived from wild-type and heterozygous clones exhibited significant degradative responses, highlighting the need for complete IL-1 blockade. CONCLUSION: This work demonstrates proof-of-concept of the ability to engineer custom-designed stem cells that are immune to proinflammatory cytokines (i.e., IL-1) as a potential cell source for cartilage tissue engineering.


Asunto(s)
Sistemas CRISPR-Cas , Cartílago/efectos de los fármacos , Condrogénesis , Edición Génica/métodos , Células Madre Pluripotentes Inducidas/inmunología , Interleucina-1/farmacología , FN-kappa B/efectos de los fármacos , Receptores de Interleucina-1/genética , Animales , Cartílago/inmunología , Cartílago/metabolismo , Condrocitos/efectos de los fármacos , Condrocitos/inmunología , Condrocitos/metabolismo , Citometría de Flujo , Eliminación de Gen , Expresión Génica , Regulación de la Expresión Génica , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Ratones , FN-kappa B/inmunología , Ingeniería de Tejidos/métodos
10.
Tissue Eng Part A ; 21(7-8): 1261-74, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25517798

RESUMEN

The limited regenerative capacity of articular cartilage contributes to progressive joint dysfunction associated with cartilage injury or osteoarthritis. Cartilage tissue engineering seeks to provide a biological substitute for repairing damaged or diseased cartilage, but requires a cell source with the capacity for extensive expansion without loss of chondrogenic potential. In this study, we hypothesized that decreased expression of the cell cycle inhibitor p21 would enhance the proliferative and chondrogenic potential of differentiated induced pluripotent stem cells (iPSCs). Murine iPSCs were directed to differentiate toward the chondrogenic lineage with an established protocol and then engineered to express a short hairpin RNA (shRNA) to reduce the expression of p21. Cells expressing the p21 shRNA demonstrated higher proliferative potential during monolayer expansion and increased synthesis of glycosaminoglycans (GAGs) in pellet cultures. Furthermore, these cells could be expanded ∼150-fold over three additional passages without a reduction in the subsequent production of GAGs, while control cells showed reduced potential for GAG synthesis with three additional passages. In pellets from extensively passaged cells, knockdown of p21 attenuated the sharp decrease in cell number that occurred in control cells, and immunohistochemical analysis showed that p21 knockdown limited the production of type I and type X collagen while maintaining synthesis of cartilage-specific type II collagen. These findings suggest that manipulating the cell cycle can augment the monolayer expansion and preserve the chondrogenic capacity of differentiated iPSCs, providing a strategy for enhancing iPSC-based cartilage tissue engineering.


Asunto(s)
Cartílago/crecimiento & desarrollo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Técnicas de Silenciamiento del Gen , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Bromodesoxiuridina/metabolismo , Cartílago/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Condrogénesis , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo X/metabolismo , ADN/biosíntesis , Regulación de la Expresión Génica , Glicosaminoglicanos/metabolismo , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/citología , Ratones
11.
Arthritis Rheumatol ; 66(11): 3062-72, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25047145

RESUMEN

OBJECTIVE: The discovery of novel disease-modifying drugs for osteoarthritis (OA) is limited by the lack of adequate genetically defined cartilage tissues for application in high-throughput screening systems. We addressed this need by synthesizing cartilage from induced pluripotent stem cells (iPSCs) to establish and validate an in vitro model of OA. METHODS: Native or iPSC-derived mouse cartilage samples were treated with the cytokine interleukin-1α (IL-1α) for 3 days to model the inflammatory environment of OA. The biochemical content, mechanical properties, and gene expression of the resulting tissues were assayed. In addition, the inflammatory and catabolic environment of the media was assessed. To establish high-throughput capability, we used a 96-well plate format and conducted a screen of previously identified candidate OA drugs. Glycosaminoglycan (GAG) release into the medium was used as the primary output for screening. RESULTS: Treatment of iPSC-derived or native cartilage with IL-1α induced characteristic features of OA in a rapid and dose-dependent manner. In addition to the loss of GAGs and tissue mechanical properties, IL-1α treatment induced the expression of matrix metalloproteinases and increased the production of the inflammatory mediators nitric oxide and prostaglandin E2 . In the high-throughput screen validation, all candidate OA therapeutic agents provided some benefit, but only the NF-κB inhibitor SC514 effectively reduced cartilage loss in response to IL-1α. CONCLUSION: This work demonstrates the utility of iPSCs for studying cartilage pathology and provides a platform for identifying novel, patient-specific therapeutic agents that prevent cartilage degradation and modify the course of OA development.


Asunto(s)
Cartílago/patología , Diferenciación Celular , Evaluación Preclínica de Medicamentos/métodos , Osteoartritis/patología , Células Madre Pluripotentes/patología , Animales , Antirreumáticos/uso terapéutico , Cartílago/efectos de los fármacos , Cartílago/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Técnicas In Vitro , Interleucina-1alfa/efectos adversos , Interleucina-1alfa/farmacología , Ratones , Ratones Endogámicos C57BL , Osteoartritis/inducido químicamente , Osteoartritis/metabolismo , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo
12.
Arch Oral Biol ; 57(6): 599-606, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22129470

RESUMEN

OBJECTIVE: The complex movement of the temporomandibular joint (TMJ) disc during mastication is controlled in large part by the disc's attachments to the surrounding tissues. This study seeks to address the lack of available quantitative data characterizing the extracellular matrix composition of the discal attachments and how these properties compare to the disc. DESIGN: Porcine TMJ disc-attachment complexes were carefully dissected into six discal attachments and five TMJ disc regions. All samples were assayed biochemically for total collagen, glycosaminoglycan (GAG), DNA, and hydration. Additionally, histology was performed on the whole joint to investigate the anatomy of the disc-attachment complex, and to verify the regional distribution of matrix components. RESULTS: Quantitative biochemical assays showed that overall water content was fairly constant in all disc and attachment regions. Disc regions generally showed higher sulfated GAG and collagen content than the attachments. In contrast, the attachments contained greater DNA content than the disc. Histological staining supported the quantitative results and also indicated more elastic fibres to be present in the attachments than the disc. CONCLUSIONS: Although macroscopically the TMJ disc and its attachments form a seamless complex within the joint, a closer look at regional biochemical constituents reveals that these two components are distinct. Whilst the disc and attachments both contain the same major constituents, the relative amounts of these components vary based on the functional requirements of the tissue. These results can further understanding of both TMJ biology and pathology.


Asunto(s)
Disco de la Articulación Temporomandibular/anatomía & histología , Disco de la Articulación Temporomandibular/metabolismo , Análisis de Varianza , Animales , Colágeno/metabolismo , ADN/metabolismo , Femenino , Glicosaminoglicanos/metabolismo , Porcinos
13.
Tissue Eng Part A ; 18(5-6): 568-75, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21942992

RESUMEN

Immune rejection is a major concern for any allogeneic or xenogeneic graft. For in vivo investigations of cartilage tissue engineering strategies, small animal models such as the leporine model are commonly employed. Many studies report little to no immune rejection upon allogeneic or xenogeneic implantation of native articular and meniscal cartilages. This study investigated whether bovine and leporine articular chondrocytes (ACs) and meniscus cells (MCs) have immunoprivileged characteristics because of their ability to stimulate proliferation of leporine peripheral blood mononuclear cells (PBMCs) in vitro. After 6 days of co-culture, none of the cell types caused a proliferative response in the leporine PBMCs, indicating that these cells may not elicit immune rejection in vivo. Reverse transcriptase polymerase chain reaction analysis for major histocompatibility complex class (MHC) I and II and costimulation factors CD80 and CD86 revealed that all cell types produced messenger RNA for MHC I and II, but only some were CD80 or CD86 positive, and none were positive for both costimulation factors. Flow cytometry found that bovine MCs and ACs displayed MHC II (MCs: 32.5%, ACs: 14.4%), whereas only leporine ACs were MHC II positive (7.5%). Although present in isolated cells, MHC I and II were not observed in intact bovine or leporine hyaline cartilage or meniscus tissues. Despite some presence of MHC II and costimulation factors, none of the cell types studied were able to cause PBMC proliferation. These findings indicate that bovine and leporine MCs and ACs share a similar immunoprivileged profile, bolstering their use as allogeneic and xenogeneic cell sources for engineered cartilage.


Asunto(s)
Bioprótesis , Condrocitos/inmunología , Condrocitos/trasplante , Rechazo de Injerto/inmunología , Cartílago Hialino/inmunología , Meniscos Tibiales/inmunología , Ingeniería de Tejidos , Animales , Antígeno B7-1/inmunología , Antígeno B7-2/inmunología , Bovinos , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Cartílago Hialino/trasplante , Meniscos Tibiales/trasplante , Conejos , Trasplante Heterólogo , Trasplante Homólogo
14.
J Appl Physiol (1985) ; 111(6): 1590-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21903884

RESUMEN

High compressive properties of cartilaginous tissues are commonly attributed to the sulfated glycosaminoglycan (GAG) fraction of the extracellular matrix (ECM), but this relationship has not been directly measured in the knee meniscus, which shows regional variation in GAG content. In this study, biopsies from each meniscus region (outer, middle, and inner) were either subjected to chondroitinase ABC (CABC) to remove all sulfated GAGs or not. Compressive testing revealed that GAG depletion in the inner and middle meniscus regions caused a significant decrease in modulus of relaxation (58% and 41% decreases, respectively, at 20% strain), and all regions exhibited a significant decrease in viscosity (outer: 29%; middle: 58%; inner: 62% decrease). Tensile properties following CABC treatment were unaffected for outer and middle meniscus specimens, but the inner meniscus displayed significant increases in Young's modulus (41% increase) and ultimate tensile stress (40% increase) following GAG depletion. These findings suggest that, in the outer meniscus, GAGs contribute to increasing tissue viscosity, whereas in the middle and inner meniscus, where GAGs are most abundant, these molecules also enhance the tissue's ability to withstand compressive loads. GAGs in the inner meniscus also contribute to reducing the circumferential tensile properties of the tissue, perhaps due to the pre-stress on the collagen network from increased hydration of the ECM. Understanding the mechanical role of GAGs in each region of the knee meniscus is important for understanding meniscus structure-function relationships and creating design criteria for functional meniscus tissue engineering efforts.


Asunto(s)
Glicosaminoglicanos/fisiología , Meniscos Tibiales/fisiología , Animales , Fenómenos Biomecánicos/fisiología , Bovinos , Condroitina ABC Liasa/farmacología , Fuerza Compresiva/fisiología , Matriz Extracelular/fisiología , Técnicas In Vitro , Meniscos Tibiales/anatomía & histología , Meniscos Tibiales/efectos de los fármacos
15.
Stem Cells Dev ; 18(2): 283-92, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18454697

RESUMEN

The successful differentiation of human embryonic stem cells (hESCs) to fibrochondrocyte-like cells and characterization of these differentiated cells is a critical step toward tissue engineering of musculoskeletal fibrocartilages (e.g., knee meniscus, temporomandibular joint disc, and intervertebral disc). In this study, growth factors and primary cell cocultures were applied to hESC embryoid bodies (EBs) for 3 weeks and evaluated for their effect on the synthesis of critical fibrocartilage matrix components: glycosaminoglycans (GAG) and collagens (types I, II, and VI). Changes in surface markers (CD105, CD44, SSEA, PDGFR alpha) after the differentiation treatments were also analyzed. The study was conducted in three phases: (1) examination of growth factors (TGF-beta 3, BMP-2, BMP-4, BMP-6, PDGF-BB, sonic hedgehog protein); (2) comparison of two cocultures (primary chondrocytes or fibrochondrocytes); and (3) the combination of the most effective growth factor and coculture regimen. TGF-beta 3 with BMP-4 yielded EBs positive for collagens I, II, and VI, with up to 6.7- and 4.8-fold increases in GAG and collagen, respectively. Analysis of cell surface markers showed a significant increase in CD44 with the TGF-beta 3 + BMP-4 treatment compared to the controls. Coculture with fibrochondrocytes resulted in up to a 9.8-fold increase in collagen II production. The combination of the growth factors BMP-4 + TGF-beta 3 with the fibrochondrocyte coculture led to an increase in cell proliferation and GAG production compared to either treatment alone. This study determined two powerful treatments for inducing fibrocartilaginous differentiation of hESCs and provides a foundation for using flow cytometry to purify these differentiated cells.


Asunto(s)
Condrogénesis/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Animales , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Condrocitos/citología , Condrocitos/efectos de los fármacos , Técnicas de Cocultivo , Colágeno/metabolismo , Citometría de Flujo , Glicosaminoglicanos/metabolismo , Humanos , Inmunohistoquímica , Ratones , Factor de Crecimiento Derivado de Plaquetas/farmacología , Factor de Crecimiento Transformador beta3/farmacología
16.
J Biomech Eng ; 131(6): 061011, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19449965

RESUMEN

Human embryonic stem cells (hESCs) possess an immense potential in a variety of regenerative applications. A firm understanding of hESC mechanics, on the single cell level, may provide great insight into the role of biophysical forces in the maintenance of cellular phenotype and elucidate mechanical cues promoting differentiation along various mesenchymal lineages. Moreover, cellular biomechanics can provide an additional tool for characterizing stem cells as they follow certain differentiation lineages, and thus may aid in identifying differentiated hESCs, which are most suitable for tissue engineering. This study examined the viscoelastic properties of single undifferentiated hESCs, chondrogenically differentiated hESC subpopulations, mesenchymal stem cells (MSCs), and articular chondrocytes (ACs). hESC chondrogenesis was induced using either transforming growth factor-beta1 (TGF-beta1) or knock out serum replacer as differentiation agents, and the resulting cell populations were separated based on density. All cell groups were mechanically tested using unconfined creep cytocompression. Analyses of subpopulations from all differentiation regimens resulted in a spectrum of mechanical and morphological properties spanning the range of hESCs to MSCs to ACs. Density separation was further successful in isolating cellular subpopulations with distinct mechanical properties. The instantaneous and relaxed moduli of subpopulations from TGF-beta1 differentiation regimen were statistically greater than those of undifferentiated hESCs. In addition, two subpopulations from the TGF-beta1 group were identified, which were not statistically different from native articular chondrocytes in their instantaneous and relaxed moduli, as well as their apparent viscosity. Identification of a differentiated hESC subpopulation with similar mechanical properties as native chondrocytes may provide an excellent cell source for tissue engineering applications. These cells will need to withstand any mechanical stimulation regimen employed to augment the mechanical and biochemical characteristics of the neotissue. Density separation was effective at purifying distinct populations of cells. A differentiated hESC subpopulation was identified with both similar mechanical and morphological characteristics as ACs. Future research may utilize this cell source in cartilage regeneration efforts.


Asunto(s)
Diferenciación Celular/fisiología , Elasticidad , Células Madre Embrionarias/fisiología , Células Madre Mesenquimatosas/citología , Adulto , Cartílago Articular/citología , Cartílago Articular/fisiología , Condrocitos/citología , Condrocitos/fisiología , Células Madre Embrionarias/citología , Humanos , Células Madre Mesenquimatosas/fisiología , Factor de Crecimiento Transformador beta1/farmacología , Viscosidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA