Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Acta Biomater ; 55: 100-108, 2017 06.
Article in English | MEDLINE | ID: mdl-28433788

ABSTRACT

Nucleus pulposus (NP) cells are derived from the notochord and differ from neighboring cells of the intervertebral disc in phenotypic marker expression and morphology. Adult human NP cells lose this phenotype and morphology with age in a pattern that contributes to progressive disc degeneration and pathology. Select laminin-mimetic peptide ligands and substrate stiffnesses were examined for their ability to regulate human NP cell phenotype and biosynthesis through the expression of NP-specific markers aggrecan, N-cadherin, collagen types I and II, and GLUT1. Peptide-conjugated substrates demonstrated an ability to promote expression of healthy NP-specific markers, as well as increased biosynthetic activity. We show an ability to re-express markers of the juvenile NP cell and morphology through control of peptide presentation and stiffness on well-characterized polyacrylamide substrates. NP cells cultured on surfaces conjugated with α3 integrin receptor peptides P4 and P678, and on α2, α5, α6, ß1 integrin-recognizing peptide AG10, show increased expression of aggrecan, N-cadherin, and types I and II collagen, suggesting a healthier, more juvenile-like phenotype. Multi-cell cluster formation was also observed to be more prominent on peptide-conjugated substrates. These findings indicate a critical role for cell-matrix interactions with specific ECM-mimetic peptides in supporting and maintaining a healthy NP cell phenotype and bioactivity. STATEMENT OF SIGNIFICANCE: NP cells reside in a laminin-rich environment that deteriorates with age, including a loss of water content and changes in the extracellular matrix (ECM) structure that may lead to the development of a degenerated IVD. There is great interest in methods to re-express healthy, biosynthetically active NP cells using laminin-derived biomimetic peptides toward the goal of using autologous cell sources for tissue regeneration. Here, we describe a novel study utilizing several laminin mimetic peptides conjugated to polyacrylamide gels that are able to support an immature, healthy NP phenotype after culture on "soft" peptide gels. These findings can support future studies in tissue regeneration where cells may be directed to a desired regenerative phenotype using niche-specific ECM peptides.


Subject(s)
Antigens, Differentiation/metabolism , Gene Expression Regulation , Intervertebral Disc/metabolism , Laminin/chemistry , Peptides/chemistry , Acrylic Resins/chemistry , Adult , Aged , Aggrecans/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cells, Cultured , Collagen Type I/metabolism , Collagen Type II/metabolism , Female , Humans , Integrins/metabolism , Intervertebral Disc/cytology , Male , Middle Aged
2.
J Biomech Eng ; 139(2)2017 02 01.
Article in English | MEDLINE | ID: mdl-28024085

ABSTRACT

The function of the heart valve interstitial cells (VICs) is intimately connected to heart valve tissue remodeling and repair, as well as the onset and progression of valvular pathological processes. There is yet only very limited knowledge and extant models for the complex three-dimensional VIC internal stress-bearing structures, the associated cell-level biomechanical behaviors, and how they change under varying activation levels. Importantly, VICs are known to exist and function within the highly dynamic valve tissue environment, including very high physiological loading rates. Yet we have no knowledge on how these factors affect VIC function. To this end, we extended our previous VIC computational continuum mechanics model (Sakamoto, et al., 2016, "On Intrinsic Stress Fiber Contractile Forces in Semilunar Heart Valve Interstitial Cells Using a Continuum Mixture Model," J. Mech. Behav. Biomed. Mater., 54(244-258)). to incorporate realistic stress-fiber geometries, force-length relations (Hill model for active contraction), explicit α-smooth muscle actin (α-SMA) and F-actin expression levels, and strain rate. Novel micro-indentation measurements were then performed using cytochalasin D (CytoD), variable KCl molar concentrations, both alone and with transforming growth factor ß1 (TGF-ß1) (which emulates certain valvular pathological processes) to explore how α-SMA and F-actin expression levels influenced stress fiber responses under quasi-static and physiological loading rates. Simulation results indicated that both F-actin and α-SMA contributed substantially to stress fiber force generation, with the highest activation state (90 mM KCL + TGF-ß1) inducing the largest α-SMA levels and associated force generation. Validation was performed by comparisons to traction force microscopy studies, which showed very good agreement. Interestingly, only in the highest activation state was strain rate sensitivity observed, which was captured successfully in the simulations. These unique findings demonstrated that only VICs with high levels of αSMA expression exhibited significant viscoelastic effects. Implications of this study include greater insight into the functional role of α-SMA and F-actin in VIC stress fiber function, and the potential for strain rate-dependent effects in pathological states where high levels of α-SMA occur, which appear to be unique to the valvular cellular in vivo microenvironment.


Subject(s)
Heart Valves/cytology , Heart Valves/physiology , Mechanotransduction, Cellular/physiology , Models, Cardiovascular , Myocardial Contraction/physiology , Stress Fibers/physiology , Animals , Computer Simulation , Humans , Molecular Motor Proteins/physiology , Stress, Mechanical
3.
J Clin Invest ; 125(3): 1147-62, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25664850

ABSTRACT

Epithelial tumor metastasis is preceded by an accumulation of collagen cross-links that heighten stromal stiffness and stimulate the invasive properties of tumor cells. However, the biochemical nature of collagen cross-links in cancer is still unclear. Here, we postulated that epithelial tumorigenesis is accompanied by changes in the biochemical type of collagen cross-links. Utilizing resected human lung cancer tissues and a p21CIP1/WAF1-deficient, K-rasG12D-expressing murine metastatic lung cancer model, we showed that, relative to normal lung tissues, tumor stroma contains higher levels of hydroxylysine aldehyde-derived collagen cross-links (HLCCs) and lower levels of lysine aldehyde-derived cross-links (LCCs), which are the predominant types of collagen cross-links in skeletal tissues and soft tissues, respectively. Gain- and loss-of-function studies in tumor cells showed that lysyl hydroxylase 2 (LH2), which hydroxylates telopeptidyl lysine residues on collagen, shifted the tumor stroma toward a high-HLCC, low-LCC state, increased tumor stiffness, and enhanced tumor cell invasion and metastasis. Together, our data indicate that LH2 enhances the metastatic properties of tumor cells and functions as a regulatory switch that controls the relative abundance of biochemically distinct types of collagen cross-links in the tumor stroma.


Subject(s)
Adenocarcinoma/enzymology , Carcinoma, Squamous Cell/enzymology , Collagen/metabolism , Lung Neoplasms/enzymology , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/physiology , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Animals , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/secondary , Cell Line, Tumor , Cells, Cultured , Enzyme Induction , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice, 129 Strain , Mice, Transgenic , Neoplasm Transplantation , Promoter Regions, Genetic , STAT3 Transcription Factor/metabolism , Tumor Microenvironment , Up-Regulation
4.
Arthritis Rheumatol ; 67(5): 1286-94, 2015 May.
Article in English | MEDLINE | ID: mdl-25604429

ABSTRACT

OBJECTIVE: Mechanical factors play a critical role in the physiology and pathology of articular cartilage, although the mechanisms of mechanical signal transduction are not fully understood. We undertook this study to test the hypothesis that type VI collagen is necessary for mechanotransduction in articular cartilage by determining the effects of type VI collagen knockout on the activation of the mechano-osmosensitive, calcium-permeable channel TRPV4 (transient receptor potential vanilloid channel 4) as well as on osmotically induced chondrocyte swelling and pericellular matrix (PCM) mechanical properties. METHODS: Confocal laser scanning microscopy was used to image TRPV4-mediated calcium signaling and osmotically induced cell swelling in intact femora from 2- and 9-month-old wild-type (WT) and type VI collagen-deficient (Col6a1(-/-)) mice. Immunofluorescence-guided atomic force microscopy was used to map PCM mechanical properties based on the presence of perlecan. RESULTS: Hypo-osmotic stress-induced TRPV4-mediated calcium signaling was increased in Col6a1(-/-) mice relative to WT controls at 2 months. Col6a1(-/-) mice exhibited significantly increased osmotically induced cell swelling and decreased PCM moduli relative to WT controls at both ages. CONCLUSION: In contrast to our original hypothesis, type VI collagen was not required for TRPV4-mediated Ca(2+) signaling; however, knockout of type VI collagen altered the mechanical properties of the PCM, which in turn increased the extent of cell swelling and osmotically induced TRPV4 signaling in an age-dependent manner. These findings emphasize the role of the PCM as a transducer of mechanical and physicochemical signals, and they suggest that alterations in PCM properties, as may occur with aging or osteoarthritis, can influence mechanotransduction via TRPV4 or other ion channels.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Collagen Type VI/genetics , Extracellular Matrix/metabolism , Mechanotransduction, Cellular/genetics , Osmotic Pressure , TRPV Cation Channels/metabolism , Animals , Collagen Type VI/metabolism , Heparan Sulfate Proteoglycans/metabolism , Mice , Mice, Knockout , Microscopy, Confocal
5.
Curr Rheumatol Rep ; 16(10): 451, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25182679

ABSTRACT

Articular cartilage injuries and degenerative joint diseases are responsible for progressive pain and disability in millions of people worldwide, yet there is currently no treatment available to restore full joint functionality. As the tissue functions under mechanical load, an understanding of the physiologic or pathologic effects of biomechanical factors on cartilage physiology is of particular interest. Here, we highlight studies that have measured cartilage deformation at scales ranging from the macroscale to the microscale, as well as the responses of the resident cartilage cells, chondrocytes, to mechanical loading using in vitro and in vivo approaches. From these studies, it is clear that there exists a complex interplay among mechanical, inflammatory, and biochemical factors that can either support or inhibit cartilage matrix homeostasis under normal or pathologic conditions. Understanding these interactions is an important step toward developing tissue engineering approaches and therapeutic interventions for cartilage pathologies, such as osteoarthritis.


Subject(s)
Cartilage, Articular/pathology , Chondrocytes/pathology , Osteoarthritis/pathology , Humans
6.
Matrix Biol ; 39: 25-32, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25172825

ABSTRACT

Chondrocytes in articular cartilage are surrounded by a narrow pericellular matrix (PCM) that is both biochemically and biomechanically distinct from the extracellular matrix (ECM) of the tissue. While the PCM was first observed nearly a century ago, its role is still under investigation. In support of early hypotheses regarding its function, increasing evidence indicates that the PCM serves as a transducer of biochemical and biomechanical signals to the chondrocyte. Work over the past two decades has established that the PCM in adult tissue is defined biochemically by several molecular components, including type VI collagen and perlecan. On the other hand, the biomechanical properties of this structure have only recently been measured. Techniques such as micropipette aspiration, in situ imaging, computational modeling, and atomic force microscopy have determined that the PCM exhibits distinct mechanical properties as compared to the ECM, and that these properties are influenced by specific PCM components as well as disease state. Importantly, the unique relationships among the mechanical properties of the chondrocyte, PCM, and ECM in different zones of cartilage suggest that this region significantly influences the stress-strain environment of the chondrocyte. In this review, we discuss recent advances in the measurement of PCM mechanical properties and structure that further increase our understanding of PCM function. Taken together, these studies suggest that the PCM plays a critical role in controlling the mechanical environment and mechanobiology of cells in cartilage and other cartilaginous tissues, such as the meniscus or intervertebral disc.


Subject(s)
Cartilage, Articular/pathology , Extracellular Matrix/pathology , Osteoarthritis/pathology , Animals , Biomechanical Phenomena , Cartilage, Articular/metabolism , Collagen/metabolism , Extracellular Matrix/metabolism , Humans , Mechanotransduction, Cellular , Osteoarthritis/metabolism
7.
Arthritis Rheumatol ; 66(11): 3062-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25047145

ABSTRACT

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.


Subject(s)
Cartilage/pathology , Cell Differentiation , Drug Evaluation, Preclinical/methods , Osteoarthritis/pathology , Pluripotent Stem Cells/pathology , Animals , Antirheumatic Agents/therapeutic use , Cartilage/drug effects , Cartilage/metabolism , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Glycosaminoglycans/metabolism , High-Throughput Screening Assays/methods , In Vitro Techniques , Interleukin-1alpha/adverse effects , Interleukin-1alpha/pharmacology , Mice , Mice, Inbred C57BL , Osteoarthritis/chemically induced , Osteoarthritis/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism
8.
Proc Natl Acad Sci U S A ; 111(9): E798-806, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24550481

ABSTRACT

The ability to develop tissue constructs with matrix composition and biomechanical properties that promote rapid tissue repair or regeneration remains an enduring challenge in musculoskeletal engineering. Current approaches require extensive cell manipulation ex vivo, using exogenous growth factors to drive tissue-specific differentiation, matrix accumulation, and mechanical properties, thus limiting their potential clinical utility. The ability to induce and maintain differentiation of stem cells in situ could bypass these steps and enhance the success of engineering approaches for tissue regeneration. The goal of this study was to generate a self-contained bioactive scaffold capable of mediating stem cell differentiation and formation of a cartilaginous extracellular matrix (ECM) using a lentivirus-based method. We first showed that poly-L-lysine could immobilize lentivirus to poly(ε-caprolactone) films and facilitate human mesenchymal stem cell (hMSC) transduction. We then demonstrated that scaffold-mediated gene delivery of transforming growth factor ß3 (TGF-ß3), using a 3D woven poly(ε-caprolactone) scaffold, induced robust cartilaginous ECM formation by hMSCs. Chondrogenesis induced by scaffold-mediated gene delivery was as effective as traditional differentiation protocols involving medium supplementation with TGF-ß3, as assessed by gene expression, biochemical, and biomechanical analyses. Using lentiviral vectors immobilized on a biomechanically functional scaffold, we have developed a system to achieve sustained transgene expression and ECM formation by hMSCs. This method opens new avenues in the development of bioactive implants that circumvent the need for ex vivo tissue generation by enabling the long-term goal of in situ tissue engineering.


Subject(s)
Cell Differentiation/physiology , Chondrogenesis/physiology , Extracellular Matrix/physiology , Tissue Engineering/methods , Tissue Scaffolds/virology , Transduction, Genetic/methods , Analysis of Variance , Biomechanical Phenomena , DNA Primers/genetics , Flow Cytometry , Gene Transfer Techniques , Humans , Immunohistochemistry , Lentivirus , Mesenchymal Stem Cells/metabolism , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Polyesters , Polylysine , Regenerative Medicine/methods , Transforming Growth Factor beta3/genetics
9.
J Biomed Mater Res A ; 102(11): 3998-4008, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24375991

ABSTRACT

Macroscale scaffolds created from cartilage-derived matrix (CDM) demonstrate chondroinductive or chondro-inductive properties, but many fabrication methods do not allow for control of nanoscale architecture. In this regard, electrospun scaffolds have shown significant promise for cartilage tissue engineering. However, nanofibrous materials generally exhibit a relatively small pore size and require techniques such as multilayering or the inclusion of sacrificial fibers to enhance cellular infiltration. The objectives of this study were (1) to compare multilayer to single-layer electrospun poly(ɛ-caprolactone) (PCL) scaffolds for cartilage tissue engineering, and (2) to determine whether incorporation of CDM into the PCL fibers would enhance chondrogenesis by human adipose-derived stem cells (hASCs). PCL and PCL-CDM scaffolds were prepared by sequential collection of 60 electrospun layers from the surface of a grounded saline bath into a single scaffold, or by continuous electrospinning onto the surface of a grounded saline bath and harvest as a single-layer scaffold. Scaffolds were seeded with hASCs and evaluated over 28 days in culture. The predominant effects on hASCs of incorporation of CDM into scaffolds were to stimulate sulfated glycosaminoglycan synthesis and COL10A1 gene expression. Compared with single-layer scaffolds, multilayer scaffolds enhanced cell infiltration and ACAN gene expression. However, compared with single-layer constructs, multilayer PCL constructs had a much lower elastic modulus, and PCL-CDM constructs had an elastic modulus approximately 1% that of PCL constructs. These data suggest that multilayer electrospun constructs enhance homogeneous cell seeding, and that the inclusion of CDM stimulates chondrogenesis-related bioactivity.


Subject(s)
Adipose Tissue/metabolism , Cartilage , Extracellular Matrix/chemistry , Stem Cells/metabolism , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Adipose Tissue/cytology , Adult , Aggrecans/biosynthesis , Animals , Cartilage/chemistry , Cartilage/cytology , Cartilage/metabolism , Cells, Cultured , Collagen Type XI/biosynthesis , Elastic Modulus , Female , Gene Expression Regulation , Glycosaminoglycans/biosynthesis , Humans , Middle Aged , Nanofibers/chemistry , Polyesters/chemistry , Porosity , Stem Cells/cytology , Swine
10.
J Orthop Res ; 31(8): 1218-25, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23568545

ABSTRACT

Regional variations in the composition and architecture of the extracellular matrix (ECM) and pericellular matrix (PCM) of the knee meniscus play important roles in determining the local mechanical environment of meniscus cells. In this study, atomic force microscopy was used to spatially map the mechanical properties of matched ECM and perlecan-labeled PCM sites within the outer, middle, and inner porcine medial meniscus, and to evaluate the properties of the proximal surface of each region. The elastic modulus of the PCM was significantly higher in the outer region (151.4 ± 38.2 kPa) than the inner region (27.5 ± 8.8 kPa), and ECM moduli were consistently higher than region-matched PCM sites in both the outer (320.8 ± 92.5 kPa) and inner (66.1 ± 31.4 kPa) regions. These differences were associated with a higher proportion of aligned collagen fibers and lower glycosaminoglycan content in the outer region. Regional variations in the elastic moduli and some viscoelastic properties were observed on the proximal surface of the meniscus, with the inner region exhibiting the highest moduli overall. These results indicate that matrix architecture and composition play an important role in the regional micromechanical properties of the meniscus, suggesting that the local stress-strain environment of meniscal cells may vary significantly among the different regions.


Subject(s)
Extracellular Matrix/pathology , Menisci, Tibial/pathology , Microscopy, Atomic Force/methods , Animals , Biomechanical Phenomena , Collagen/metabolism , Elastic Modulus/physiology , Extracellular Matrix/metabolism , Female , Heparan Sulfate Proteoglycans/metabolism , Menisci, Tibial/metabolism , Menisci, Tibial/physiopathology , Swine
11.
Proc Natl Acad Sci U S A ; 109(47): 19172-7, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23115336

ABSTRACT

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.


Subject(s)
Cartilage/physiology , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Tissue Engineering/methods , Animals , Cell Differentiation/genetics , Cell Separation , Cellular Reprogramming/genetics , Chondrogenesis/genetics , Collagen Type II/metabolism , Elastic Modulus , Flow Cytometry , Gene Expression Regulation , Glycosaminoglycans/metabolism , Green Fluorescent Proteins/metabolism , Mice , Microscopy, Atomic Force , Organ Specificity , Sepharose , Wound Healing
12.
Biomech Model Mechanobiol ; 11(7): 1047-56, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22231673

ABSTRACT

Central to understanding mechanotransduction in the knee meniscus is the characterization of meniscus cell mechanics. In addition to biochemical and geometric differences, the inner and outer regions of the meniscus contain cells that are distinct in morphology and phenotype. This study investigated the regional variation in meniscus cell mechanics in comparison with articular chondrocytes and ligament cells. It was found that the meniscus contains two biomechanically distinct cell populations, with outer meniscus cells being stiffer (1.59 ± 0.19 kPa) than inner meniscus cells (1.07 ± 0.14 kPa). Additionally, it was found that both outer and inner meniscus cell stiffnesses were similar to ligament cells (1.32 ± 0.20 kPa), and articular chondrocytes showed the highest stiffness overall (2.51 ± 0.20 kPa). Comparison of compressibility characteristics of the cells showed similarities between articular chondrocytes and inner meniscus cells, as well as between outer meniscus cells and ligament cells. These results show that cellular biomechanics vary regionally in the knee meniscus and that meniscus cells are biomechanically similar to ligament cells. The mechanical properties of musculoskeletal cells determined in this study may be useful for the development of mathematical models or the design of experiments studying mechanotransduction in a variety of soft tissues.


Subject(s)
Cartilage, Articular/pathology , Chondrocytes/cytology , Ligaments/pathology , Menisci, Tibial/pathology , Animals , Biomechanical Phenomena , Cattle , Glycosaminoglycans/chemistry , Image Processing, Computer-Assisted , Lower Extremity/physiology , Lower Extremity/physiopathology , Microscopy, Atomic Force/methods , Phenotype , Poisson Distribution , Pressure , Stress, Mechanical
13.
Biomaterials ; 33(1): 109-19, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21959004

ABSTRACT

Adult stem cells from the dermal layer of skin are an attractive alternative to primary cells for meniscus engineering, as they may be easily obtained and used autologously. Recently, chondroinducible dermis cells from caprine skin have shown promising characteristics for cartilage tissue engineering. In this study, their multilineage differentiation capacity is determined, and methods of expanding and tissue engineering these cells are investigated. It was found that these cells could differentiate along adipogenic, osteogenic, and chondrogenic lineages, allowing them to be termed dermis isolated adult stem cells (DIAS cells). Focusing on cartilage tissue engineering, it was found that passaging these cells in chondrogenic medium and forming them into self-assembled tissue engineered constructs caused upregulation of collagen type II and COMP gene expression. Further investigation showed that applying transforming growth factor ß1 (TGF-ß1) or bone morphogenetic protein 2 (BMP-2) to DIAS constructs caused increased sulfated glycosaminoglycan content. Additionally, TGF-ß1 treatment caused significant increases in compressive properties and construct contraction. In contrast, BMP-2 treatment resulted in the largest constructs, but did not increase compressive properties. These results show that DIAS cells can be easily manipulated for cartilage tissue engineering strategies, and may also be a useful cell source for other mesenchymal tissues.


Subject(s)
Adult Stem Cells/cytology , Cartilage/cytology , Dermis/cytology , Tissue Engineering/methods , Adult Stem Cells/metabolism , Animals , Cartilage/metabolism , Cell Differentiation/physiology , Goats , Models, Biological , Reverse Transcriptase Polymerase Chain Reaction
14.
Tissue Eng Part A ; 18(5-6): 568-75, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21942992

ABSTRACT

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.


Subject(s)
Bioprosthesis , Chondrocytes/immunology , Chondrocytes/transplantation , Graft Rejection/immunology , Hyaline Cartilage/immunology , Menisci, Tibial/immunology , Tissue Engineering , Animals , B7-1 Antigen/immunology , B7-2 Antigen/immunology , Cattle , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Hyaline Cartilage/transplantation , Menisci, Tibial/transplantation , Rabbits , Transplantation, Heterologous , Transplantation, Homologous
15.
Tissue Eng Part C Methods ; 18(3): 235-43, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22029490

ABSTRACT

An abundant cell source is the cornerstone of most tissue engineering strategies, but extracting cells from the knee meniscus is hindered by its dense fibrocartilaginous matrix. Identifying a method to efficiently isolate meniscus cells is important, as it can reduce the cost and effort required to perform meniscus engineering research. In this study, six enzymatic digestion regimens used for cartilaginous cell isolation were used to isolate cells from the outer, middle, and inner regions of the bovine knee meniscus. Each regimen in each region was assessed in terms of cell yield, impact on cell phenotype, and cytotoxicity. All digestion regimens caused an overall upregulation of cartilage-specific genes Sox9, collagen type I (Col 1), collagen type II (Col 2), cartilage oligomeric matrix protein, and aggrecan (AGC) in cells from all meniscus regions, but was highest for cells isolated using 1075 U/mL of collagenase for 3 h (high collagenase). In response to isolation, outer meniscus cells showed highest upregulation of Sox9 and Col 1 genes, whereas greatest upregulation for middle meniscus cells was seen in Col 1 expression, and Col 2 expression for inner cells. Cell yield was highest in all regions when subjected to 45 min of 61 U/mL pronase followed by 3 h of 1075 U/mL collagenase (pronase/collagenase [P/C]) digestion regimen (outer: 6.57±0.37, middle: 12.77±1.41, inner: 22.17±1.47×10(6) cells/g tissue). The second highest cell yield was achieved using the low collagenase (LC) digestion regimen that applied 433 U/mL of collagenase for 18 h (outer: 1.95±0.54, middle: 3.3±4.4, inner: 6.06±2.44×10(6) cells/g tissue). Cytotoxicity analysis showed higher cell death in the LC group compared with the P/C group. Self-assembled constructs formed from LC-isolated cells were less dense than constructs formed from P/C-isolated cells, and P/C constructs showed higher glycosaminoglycan content and compressive moduli than LC constructs. All isolation methods tested resulted in similar phenotypic changes in meniscus cells from each region. These results indicate that, compared with other common isolation protocols, the P/C isolation method is able to more efficiently isolate meniscus cells from all regions that can produce tissue engineered constructs.


Subject(s)
Cell Separation/methods , Collagenases/pharmacology , Knee Joint/metabolism , Menisci, Tibial/cytology , Menisci, Tibial/drug effects , Tissue Engineering/methods , Animals , Cattle , Cell Count , Cells, Cultured , Collagenases/metabolism , Efficiency/drug effects , Knee Joint/cytology , Knee Joint/drug effects , Knee Joint/pathology , Menisci, Tibial/pathology , Necrosis/chemically induced , Phenotype , Proteolysis
16.
J Appl Physiol (1985) ; 111(6): 1590-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21903884

ABSTRACT

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.


Subject(s)
Glycosaminoglycans/physiology , Menisci, Tibial/physiology , Animals , Biomechanical Phenomena/physiology , Cattle , Chondroitin ABC Lyase/pharmacology , Compressive Strength/physiology , Extracellular Matrix/physiology , In Vitro Techniques , Menisci, Tibial/anatomy & histology , Menisci, Tibial/drug effects
17.
Biomacromolecules ; 7(6): 1968-75, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16768421

ABSTRACT

Degradation plays an important role in the evolution of the extracellular matrix secreted by chondrocytes encapsulated in PEG-based hydrogels. For this study, macromonomers were synthesized by methacrylating both ends of polycaprolactone-b-poly(ethylene glycol)-b-polycaprolactone (PEG-CAP) tri-block copolymers. These divinyl molecules were photopolymerized to form hydrogels with PEG-CAP crosslinks that were subsequently degraded upon exogenous addition of a lipase enzyme. The rate of degradation and subsequent mass loss depends on both the length of the polycaprolactone units and the concentration of enzyme. Control gels that did not receive lipase did not significantly degrade on the time scale of these experiments. A model was developed to predict mass loss using enzyme kinetics and a previously described statistical treatment of bulk network degradation. The model was used to predict mass loss profiles at the specific conditions used, and also to demonstrate the importance of potential changes in reaction rate and enzyme stability on temporal mass loss.


Subject(s)
Hydrogels/chemical synthesis , Lipase/chemistry , Models, Chemical , Polyesters/chemistry , Polyesters/chemical synthesis , Polyethylene Glycols/chemical synthesis , Hydrogels/chemistry , Kinetics , Molecular Structure , Molecular Weight , Photochemistry , Polyethylene Glycols/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...