Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Eur Cell Mater ; 41: 546-557, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34008855

ABSTRACT

Marrow stimulation, including subchondral drilling and microfracture, is the most commonly performed cartilage repair strategy, whereby the subchondral bone plate is perforated to release marrow-derived cells into a cartilage defect to initiate repair. Novel scaffolds and therapeutics are being designed to enhance and extend the positive short-term outcomes of this marrow stimulation. However, the translation of these newer treatments is hindered by bony abnormalities, including bone resorption, intralesional osteophytes, and bone cysts, that can arise after marrow stimulation. In this study, three different marrow stimulation approaches - microfracture, subchondral drilling and needle-puncture - were evaluated in a translationally relevant large-animal model, the Yucatan minipig. The objective of the study was to determine which method of marrow access (malleted awl, drilled Kirschner wire or spring-loaded needle) best preserved the underlying subchondral bone. Fluorochrome labels were injected at the time of surgery and 2 weeks post-surgery to capture bone remodelling over the first 4 weeks. Comprehensive outcome measures included cartilage indentation testing, histological grading, microcomputed tomography and fluorochrome imaging. Findings indicated that needle-puncture devices best preserved the underlying subchondral bone relative to other marrow access approaches. This may relate to the degree of bony compaction occurring with marrow access, as the Kirschner wire approach, which consolidated bone the most, induced the most significant bone damage with marrow stimulation. This study provided basic scientific evidence in support of updated marrow stimulation techniques for preclinical and clinical practice.


Subject(s)
Bone Remodeling/physiology , Bone and Bones/physiology , Animals , Cartilage, Articular/physiology , Male , Models, Animal , Osteophyte/physiopathology , Swine , Swine, Miniature
2.
Eur Cell Mater ; 41: 370-380, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33763848

ABSTRACT

Back and neck pain have become primary reasons for disability and healthcare spending globally. While the causes of back pain are multifactorial, intervertebral disc degeneration is frequently cited as a primary source of pain. The annulus fibrosus (AF) and nucleus pulposus (NP) subcomponents of the disc are common targets for regenerative therapeutics. However, disc degeneration is also associated with degenerative changes to adjacent spinal tissues, and successful regenerative therapies will likely need to consider and address the pathology of adjacent spinal structures beyond solely the disc subcomponents. This review summarises the current state of knowledge in the field regarding associations between back pain, disc degeneration, and degeneration of the cartilaginous and bony endplates, the AF-vertebral body interface, the facet joints and spinal muscles, in addition to a discussion of regenerative strategies for treating pain and degeneration from a whole motion segment perspective.


Subject(s)
Intervertebral Disc Degeneration/pathology , Intervertebral Disc/pathology , Regeneration/physiology , Animals , Annulus Fibrosus/pathology , Back Pain/pathology , Humans , Nucleus Pulposus/pathology
3.
Eur Cell Mater ; 41: 40-51, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33411938

ABSTRACT

The repair of focal cartilage defects remains one of the foremost issues in the field of orthopaedics. Chondral defects may arise from a variety of joint pathologies and left untreated, will likely progress to osteoarthritis. Current repair techniques, such as microfracture, result in short-term clinical improvements but have poor long-term outcomes. Emerging scaffold-based repair strategies have reported superior outcomes compared to microfracture and motivate the development of new biomaterials for this purpose. In this study, unique composite implants consisting of a base porous reinforcing component (woven poly(ε-caprolactone)) infiltrated with 1 of 2 hydrogels (self-assembling peptide or thermo-gelling hyaluronan) or bone marrow aspirate were evaluated. The objective was to evaluate cartilage repair with composite scaffold treatment compared to the current standard of care (microfracture) in a translationally relevant large animal model, the Yucatan minipig. While many cartilage-repair studies have shown some success in vivo, most are short term and not clinically relevant. Informed by promising 6-week findings, a 12-month study was carried out and those results are presented here. To aid in comparisons across platforms, several structural and functionally relevant outcome measures were performed. Despite positive early findings, the long-term results indicated less than optimal structural and mechanical results with respect to cartilage repair, with all treatment groups performing worse than the standard of care. This study is important in that it brings much needed attention to the importance of performing translationally relevant long-term studies in an appropriate animal model when developing new clinical cartilage repair approaches.


Subject(s)
Cartilage, Articular , Animals , Biocompatible Materials , Cartilage, Articular/surgery , Disease Models, Animal , Hyaluronic Acid , Swine , Swine, Miniature
4.
Eur Cell Mater ; 38: 23-34, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31393594

ABSTRACT

Articular cartilage is a specialised tissue that has a relatively homogenous endogenous cell population but a diverse extracellular matrix (ECM), with depth-dependent mechanical properties. Repair of this tissue remains an elusive clinical goal, with biological interventions preferred to arthroplasty in younger patients. Osteochondral transplantation (OCT) has emerged for the treatment of cartilage defects and osteoarthritis. Fresh allografts stored at 4 °C have been utilised, though matrix and cell viability loss remains an issue. To address this, several studies have developed media formulations to maintain cartilage explants in vitro. One promising factor for these applications is sprifermin, a human-recombinant fibroblast growth factor-18, which stimulates chondrocyte proliferation and matrix synthesis and is in clinical trials for the treatment of osteoarthritis. The study hypothesis was that addition of sprifermin during storage would maintain the unique depth-dependent mechanical profile of articular cartilage explants, a feature not often evaluated. Explants were maintained for up to 6 weeks with or without a weekly 24 h exposure to sprifermin (100 ng/mL) and the compressive modulus was assessed. Results showed that sprifermin-treated samples maintained their depth-dependent mechanical profile through 3 weeks, whereas untreated samples lost their mechanical integrity over 1 week of culture. Sprifermin also affected ECM balance by maintaining the levels of extracellular collagen and suppressing matrix metalloproteinase production. These findings support the use of sprifermin as a medium additive for OCT allografts during in vitro storage and present a potential mechanism where sprifermin may impact a functional characteristic of articular cartilage in repair strategies.


Subject(s)
Cartilage, Articular/drug effects , Compressive Strength , Fibroblast Growth Factors/pharmacology , Animals , Cartilage, Articular/chemistry , Cartilage, Articular/metabolism , Cattle , Cells, Cultured , Collagen/metabolism , Glycosaminoglycans/metabolism , Humans , Matrix Metalloproteinases/metabolism , Recombinant Proteins/pharmacology
5.
Osteoarthritis Cartilage ; 27(12): 1860-1869, 2019 12.
Article in English | MEDLINE | ID: mdl-31419488

ABSTRACT

OBJECTIVES: The objective of this study was to perform a quantitative analysis of the structural and functional alterations in the intervertebral disc during in vivo degeneration, using emerging tools that enable rigorous assessment from the microscale to the macroscale, as well as to correlate these outcomes with noninvasive, clinically relevant imaging parameters. DESIGN: Degeneration was induced in a rabbit model by puncturing the annulus fibrosus (AF) with a 16-gauge needle. 2, 4, 8, and 12 weeks following puncture, degenerative changes in the discs were evaluated via magnetic resonance imaging (MRI), whole motion segment biomechanics, atomic force microscopy, histology and polarized light microscopy, immunohistochemistry, biochemical content, and second harmonic generation imaging. RESULTS: Following puncture, degeneration was evident through marked changes in whole disc structure and mechanics. Puncture acutely compromised disc macro and microscale mechanics, followed by progressive stiffening and remodeling. Histological analysis showed substantial anterior fibrotic remodeling and osteophyte formation, as well as an overall reduction in disc height, and disorganization and infolding of the AF lamellae into the NP space. Increases in NP collagen content and aggrecan breakdown products were also noted within 4 weeks. On MRI, NP T2 was reduced at all post-puncture time points and correlated significantly with microscale indentation modulus. CONCLUSION: This study defined the time dependent changes in disc structure-function relationships during IVD degeneration in a rabbit annular injury model and correlated degeneration severity with clinical imaging parameters. Our findings identified AF infolding and occupancy of the space as a principle mechanism of disc degeneration in response to needle puncture, and provide new insights to direct the development of novel therapeutics.


Subject(s)
Annulus Fibrosus/diagnostic imaging , Intervertebral Disc Degeneration/diagnostic imaging , Nucleus Pulposus/diagnostic imaging , Aggrecans/metabolism , Animals , Annulus Fibrosus/metabolism , Annulus Fibrosus/pathology , Annulus Fibrosus/physiopathology , Biomechanical Phenomena , Collagen/metabolism , Disease Models, Animal , Disease Progression , Immunohistochemistry , Intervertebral Disc/diagnostic imaging , Intervertebral Disc/metabolism , Intervertebral Disc/pathology , Intervertebral Disc/physiopathology , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/physiopathology , Magnetic Resonance Imaging , Microscopy, Atomic Force , Microscopy, Polarization , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Nucleus Pulposus/physiopathology , Punctures , Rabbits , Second Harmonic Generation Microscopy
6.
Sci Rep ; 7(1): 15765, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29150639

ABSTRACT

Total disc replacement with an engineered substitute is a promising avenue for treating advanced intervertebral disc disease. Toward this goal, we developed cell-seeded disc-like angle ply structures (DAPS) and showed through in vitro studies that these constructs mature to match native disc composition, structure, and function with long-term culture. We then evaluated DAPS performance in an in vivo rat model of total disc replacement; over 5 weeks in vivo, DAPS maintained their structure, prevented intervertebral bony fusion, and matched native disc mechanical function at physiologic loads in situ. However, DAPS rapidly lost proteoglycan post-implantation and did not integrate into adjacent vertebrae. To address this, we modified the design to include polymer endplates to interface the DAPS with adjacent vertebrae, and showed that this modification mitigated in vivo proteoglycan loss while maintaining mechanical function and promoting integration. Together, these data demonstrate that cell-seeded engineered discs can replicate many characteristics of the native disc and are a viable option for total disc arthroplasty.


Subject(s)
Tissue Engineering/methods , Total Disc Replacement , Animals , Cattle , Cells, Cultured , Male , Prosthesis Implantation , Rats , Subcutaneous Tissue/physiology
7.
Eur Cell Mater ; 34: 217-231, 2017 10 27.
Article in English | MEDLINE | ID: mdl-29076514

ABSTRACT

Mesenchymal stem cells (MSCs) display substantial cell-to-cell variation. This heterogeneity manifests among donors, among tissue sources, and within cell populations. Such pervasive variability complicates the use of MSCs in regenerative applications and may limit their therapeutic efficacy. Most conventional assays measure MSC properties in bulk and, as a consequence, mask this cell-to-cell variation. Recent studies have identified extensive variability amongst and within clonal MSC populations, in dimensions including functional differentiation capacity, molecular state (e.g. epigenetic, transcriptomic, and proteomic status), and biophysical properties. While the origins of these variations remain to be elucidated, potential mechanisms include in vivo micro-anatomical heterogeneity, epigenetic bistability, and transcriptional fluctuations. Emerging tools for single cell analysis of MSC gene and protein expression may yield further insight into the mechanisms and implications of single cell variation amongst these cells, and ultimately improve the clinical utility of MSCs in tissue engineering and regenerative medicine applications. This review outlines the dimensions across which MSC heterogeneity is present, defines some of the known mechanisms that govern this heterogeneity, and highlights emerging technologies that may further refine our understanding and improve our clinical application of this unique cell type.


Subject(s)
Cell Differentiation , Cellular Microenvironment , Mesenchymal Stem Cells/cytology , Single-Cell Analysis/methods , Animals , Gene Expression Profiling/methods , Humans , Mesenchymal Stem Cells/metabolism , Proteomics/methods , Regenerative Medicine/methods , Tissue Engineering/methods
8.
Osteoarthritis Cartilage ; 25(1): 146-156, 2017 01.
Article in English | MEDLINE | ID: mdl-27568573

ABSTRACT

OBJECTIVE: The objective of this study was to establish a large animal model that recapitulates the spectrum of intervertebral disc degeneration that occurs in humans and which is suitable for pre-clinical evaluation of a wide range of experimental therapeutics. DESIGN: Degeneration was induced in the lumbar intervertebral discs of large frame goats by either intradiscal injection of chondroitinase ABC (ChABC) over a range of dosages (0.1U, 1U or 5U) or subtotal nucleotomy. Radiographs were used to assess disc height changes over 12 weeks. Degenerative changes to the discs and endplates were assessed via magnetic resonance imaging (MRI), semi-quantitative histological grading, microcomputed tomography (µCT), and measurement of disc biomechanical properties. RESULTS: Degenerative changes were observed for all interventions that ranged from mild (0.1U ChABC) to moderate (1U ChABC and nucleotomy) to severe (5U ChABC). All groups showed progressive reductions in disc height over 12 weeks. Histological scores were significantly increased in the 1U and 5U ChABC groups. Reductions in T2 and T1ρ, and increased Pfirrmann grade were observed on MRI. Resorption and remodeling of the cortical boney endplate adjacent to ChABC-injected discs also occurred. Spine segment range of motion (ROM) was greater and compressive modulus was lower in 1U ChABC and nucleotomy discs compared to intact. CONCLUSIONS: A large animal model of disc degeneration was established that recapitulates the spectrum of structural, compositional and biomechanical features of human disc degeneration. This model may serve as a robust platform for evaluating the efficacy of therapeutics targeted towards varying degrees of disc degeneration.


Subject(s)
Disease Models, Animal , Intervertebral Disc Degeneration/pathology , Animals , Chondroitin ABC Lyase/pharmacology , Diskectomy, Percutaneous , Goat Diseases/pathology , Goats , Humans , Intervertebral Disc/drug effects , Intervertebral Disc/surgery , Intervertebral Disc Degeneration/diagnostic imaging , Male , Radiography , X-Ray Microtomography
9.
Osteoarthritis Cartilage ; 24(8): 1330-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27063441

ABSTRACT

Meniscal lesions are common problems in orthopaedic surgery and sports medicine, and injury or loss of the meniscus accelerates the onset of knee osteoarthritis (OA). Despite a variety of therapeutic options in the clinics, there is a critical need for improved treatments to enhance meniscal repair. In this regard, combining gene-, cell-, and tissue engineering-based approaches is an attractive strategy to generate novel, effective therapies to treat meniscal lesions. In the present work, we provide an overview of the tools currently available to improve meniscal repair and discuss the progress and remaining challenges for potential future translation in patients.


Subject(s)
Genetic Therapy , Tissue Engineering , Humans , Menisci, Tibial , Meniscus , Tibial Meniscus Injuries , Wound Healing
10.
Osteoarthritis Cartilage ; 23(1): 134-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25241241

ABSTRACT

OBJECTIVE: Tissue engineering approaches for cartilage repair have focused on the use of mesenchymal stem cells (MSCs). For clinical success, MSCs must survive and produce extracellular matrix in the physiological context of the synovial joint, where low nutrient conditions engendered by avascularity, nutrient utilization, and waste production prevail. This study sought to delineate the role of microenvironmental stressors on MSC viability and functional capacity in three dimensional (3D) culture. DESIGN: We evaluated the impact of glucose and oxygen deprivation on the functional maturation of 3D MSC-laden agarose constructs. Since MSC isolation procedures result in a heterogeneous cell population, we also utilized micro-pellet culture to investigate whether clonal subpopulations respond to these microenvironmental stressors in a distinct fashion. RESULTS: MSC health and the functional maturation of 3D constructs were compromised by both glucose and oxygen deprivation. Importantly, glucose deprivation severely limited viability, and so compromised the functional maturation of 3D constructs to the greatest extent. The observation that not all cells died suggested there exists heterogeneity in the response of MSC populations to metabolic stressors. Population heterogeneity was confirmed through a series of studies utilizing clonally derived subpopulations, with a spectrum of matrix production and cell survival observed under conditions of metabolic stress. CONCLUSIONS: Our findings show that glucose deprivation has a significant impact on functional maturation, and that some MSC subpopulations are more resilient to metabolic challenge than others. These findings suggest that pre-selection of subpopulations that are resilient to metabolic challenge may improve in vivo outcomes.


Subject(s)
Cartilage , Cell Hypoxia , Glucose/deficiency , Mesenchymal Stem Cells/physiology , Tissue Culture Techniques/methods , Tissue Engineering/methods , Animals , Cattle , Cell Survival , Cells, Cultured
11.
Osteoarthritis Cartilage ; 22(9): 1282-90, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24999113

ABSTRACT

OBJECTIVE: A number of in vitro models of post-traumatic osteoarthritis (PTOA) have been developed to study the effect of mechanical overload on the processes that regulate cartilage degeneration. While such frameworks are critical for the identification therapeutic targets, existing technologies are limited in their throughput capacity. Here, we validate a test platform for high-throughput mechanical injury incorporating engineered cartilage. METHOD: We utilized a high-throughput mechanical testing platform to apply injurious compression to engineered cartilage and determined their strain and strain rate dependent responses to injury. Next, we validated this response by applying the same injury conditions to cartilage explants. Finally, we conducted a pilot screen of putative PTOA therapeutic compounds. RESULTS: Engineered cartilage response to injury was strain dependent, with a 2-fold increase in glycosaminoglycan (GAG) loss at 75% compared to 50% strain. Extensive cell death was observed adjacent to fissures, with membrane rupture corroborated by marked increases in lactate dehydrogenase (LDH) release. Testing of established PTOA therapeutics showed that pan-caspase inhibitor [Z-VAD-FMK (ZVF)] was effective at reducing cell death, while the amphiphilic polymer [Poloxamer 188 (P188)] and the free-radical scavenger [N-Acetyl-L-cysteine (NAC)] reduced GAG loss as compared to injury alone. CONCLUSIONS: The injury response in this engineered cartilage model replicated key features of the response of cartilage explants, validating this system for application of physiologically relevant injurious compression. This study establishes a novel tool for the discovery of mechanisms governing cartilage injury, as well as a screening platform for the identification of new molecules for the treatment of PTOA.


Subject(s)
Cartilage, Articular/injuries , Osteoarthritis/etiology , Tissue Engineering/methods , Acetylcysteine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/physiopathology , Caspase Inhibitors/pharmacology , Cattle , Cell Death/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Glycosaminoglycans/metabolism , High-Throughput Screening Assays/methods , Materials Testing/methods , Pilot Projects , Poloxamer/pharmacology , Stress, Mechanical
12.
Eur Cell Mater ; 27: 321-31, 2014 Jun 08.
Article in English | MEDLINE | ID: mdl-24908425

ABSTRACT

Mechanical signals regulate a multitude of cell functions and ultimately govern fibrous tissue growth, maintenance and repair. Such mechanotransduction processes often involve modulation of intracellular calcium concentration ([Ca2+]i). However, most studies interrogate these responses in cells in simplified culture systems, thereby removing potentially important inputs from the native extracellular microenvironment. The objective of this study was to test the hypothesis that the intracellular calcium response of meniscus fibrochondrocytes (MFCs) is dependent on both the microenvironmental context in which this perturbation is applied and on the tensile deformation. Using a custom micro-mechanical tester mounted on a confocal microscope, intracellular calcium activity in MFCs in response to incremental tissue strains (0, 3, 6 and 9 %) was monitored in situ (i.e., in the native tissues) on MFC-seeded aligned scaffolds and MFC-seeded silicone membranes. The [Ca2+]i regulation by MFCs within the native meniscus tissue microenvironment was considerably different from [Ca2+]i regulation by MFCs on either aligned nanofibrous scaffolds or flat silicone membranes. Additionally, increasing levels of tensile deformation resulted in a greater number of responding cells, both in situ and in vitro, while having no effects on temporal characteristics of [Ca2+]i signalling. Collectively, these findings have significant implications for mechanobiology of load-bearing fibrous tissues and their responses to injury and degeneration. In addition, from a tissue engineering perspective, the findings establish cellular benchmarks for maturing engineered constructs, where native tissue-like calcium mechano-regulation may be an important outcome parameter to achieve mechanical functionality comparable to native tissue.


Subject(s)
Calcium Signaling , Cellular Microenvironment , Chondrocytes/cytology , Chondrogenesis , Menisci, Tibial/cytology , Tensile Strength , Animals , Cattle , Chondrocytes/metabolism , Tissue Engineering/instrumentation , Tissue Engineering/methods , Tissue Scaffolds
13.
Eur Cell Mater ; 23: 425-40, 2012 Jun 10.
Article in English | MEDLINE | ID: mdl-22684531

ABSTRACT

Mesenchymal stem cells (MSCs) are a promising cell source for the treatment of musculoskeletal disease. However, MSC chondrogenesis in 3D culture generates constructs whose macroscopic (bulk) mechanical properties are inferior to constructs formed with chondrocytes. To investigate where and why these deficits in functionality arise, we assessed the local (microscopic) properties of cell-laden hydrogel constructs. Both chondrocyte- and MSC-laden constructs showed pronounced depth dependency, with ~3.5 and ~11.5 fold decreases in modulus from the surface to central regions, respectively. Importantly, in the surface region, properties were similar, suggesting that MSCs can produce matrix of mechanical equivalence to chondrocytes, but only in conditions of maximal nutrient support. Dynamic culture on an orbital shaker (which enhances diffusion) attenuated depth-dependent disparities in mechanics and improved the bulk properties compared to free swelling conditions (225 to 438 kPa for chondrocytes, 122 to 362 kPa for MSCs). However, properties in MSC-based constructs remained significantly lower due to persistent mechanical deficits in central regions. MSC viability in these central regions decreased markedly, with these changes apparent as early as day 21, while chondrocyte viability remained high. These findings suggest that, under optimal nutrient conditions, MSCs can undergo chondrogenesis and form functional tissue on par with that of the native tissue cell type. However, the lack of viability and matrix production in central regions suggests that chondrogenic MSCs do not yet fully recapitulate the advanced phenotype of the chondrocyte, a cell that is optimized to survive (and thrive) in a mechanically challenging and nutrient-poor environment.


Subject(s)
Cartilage , Cell Culture Techniques , Chondrocytes/cytology , Mesenchymal Stem Cells/cytology , Animals , Cartilage/cytology , Cartilage/growth & development , Cattle , Cell Differentiation , Cell Survival , Chondrogenesis , Hydrogels , Surface Properties , Tissue Engineering/methods
14.
J Tissue Eng Regen Med ; 4(1): 25-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19834956

ABSTRACT

The presence of a defect in mature articular cartilage can lead to degenerative changes of the joint. This is in part caused by the inability of cartilage to regenerate tissue that is capable of spanning a fissure or crack. In this study, we hypothesized that introduction of a biodegradable cell-seeded nanofibrous hydrogel, Puramatrix(), into a cartilage gap would facilitate the generation of a mechanically stable interface. The effects of chondrocyte incorporation within the hydrogel and supplementation with transforming growth factor-beta3 (TGFbeta3), a known regulator of cell growth and differentiation, on cartilage integration were examined mechanically and histologically as a function of cell density and incubation time. When supplemented with TGFbeta3, the cell-seeded hydrogel exhibited abundant matrix generation within the hydrogel and a corresponding increase in maximum push-out stress as compared to all other groups. Furthermore, initial cell seeding density affected interfacial strength in a time-dependent manner. This study suggests that a cell-seeded TGFbeta3-supplemented hydrogel can encourage integration between two opposing pieces of articular cartilage.


Subject(s)
Cartilage, Articular/physiology , Tissue Engineering/methods , Animals , Biomechanical Phenomena , Cartilage, Articular/drug effects , Cartilage, Articular/injuries , Cattle , Cell Survival , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/physiology , Chondrocytes/transplantation , Hydrogels , Models, Biological , Nanofibers , Regeneration/drug effects , Regeneration/physiology , Transforming Growth Factor beta3/pharmacology
15.
Osteoarthritis Cartilage ; 17(12): 1639-48, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19631307

ABSTRACT

OBJECTIVE: Engineering cartilage requires that a clinically relevant cell type be situated within a 3D environment that supports cell viability, the production and retention of cartilage-specific extracellular matrix (ECM), and eventually, the establishment of mechanical properties that approach that of the native tissue. In this study, we investigated the ability of bone marrow derived mesenchymal stem cells (MSCs) to undergo chondrogenesis in crosslinked methacrylated hyaluronic acid hydrogels (MeHA) of different macromer concentrations (1, 2, and 5%). DESIGN: Over a 6 week culture period under pro-chondrogenic conditions, we evaluated cartilage-specific gene expression, ECM deposition within constructs and released to the culture media, and mechanical properties in both compression and tension. Further, we examined early matrix assembly and long term histological features of the forming tissues, as well as the ability of macromolecules to diffuse within hydrogels as a function of MeHA macromer concentration. RESULTS: Findings from this study show that variations in macromer density influence MSC chondrogenesis in distinct ways. Increasing HA macromer density promoted chondrogenesis and matrix formation and retention, but yielded functionally inferior constructs due to limited matrix distribution throughout the construct expanse. In 1% MeHA constructs, the equilibrium compressive modulus reached 0.12MPa and s-GAG content reached nearly 3% of the wet weight, values that matched or exceeded those of control agarose constructs and that are 25 and 50% of native tissue levels, respectively. CONCLUSIONS: These data provide new insight into how early matrix deposition regulates long term construct development, and defines new parameters for optimizing the formation of functional MSC-based engineered articular cartilage using HA hydrogels.


Subject(s)
Cartilage, Articular/metabolism , Chondrogenesis/physiology , Extracellular Matrix/metabolism , Hyaluronic Acid/metabolism , Mesenchymal Stem Cells/metabolism , Tissue Engineering/methods , Animals , Biomechanical Phenomena , Cattle , Cells, Cultured , Hydrogels/metabolism , Mesenchymal Stem Cells/cytology
16.
Osteoarthritis Cartilage ; 17(3): 336-45, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18848784

ABSTRACT

OBJECTIVE: Injuries to the avascular regions of the meniscus fail to heal and so are treated by resection of the damaged tissue. This alleviates symptoms but fails to restore normal load transmission in the knee. Tissue engineering functional meniscus constructs for re-implantation may improve tissue repair. While numerous studies have developed scaffolds for meniscus repair, the most appropriate autologous cell source remains to be determined. In this study, we hypothesized that the debris generated from common meniscectomy procedures would possess cells with potential for forming replacement tissue. We also hypothesized that donor age and the disease status would influence the ability of derived cells to generate functional, fibrocartilaginous matrix. METHODS: Meniscus derived cells (MDCs) were isolated from waste tissue of 10 human donors (seven partial meniscectomies and three total knee arthroplasties) ranging in age from 18 to 84 years. MDCs were expanded in monolayer culture through passage 2 and seeded onto fiber-aligned biodegradable nanofibrous scaffolds and cultured in a chemically defined media. Mechanical properties, biochemical content, and histological features were evaluated over 10 weeks of culture. RESULTS: Results demonstrated that cells from every donor contributed to increasing biochemical content and mechanical properties of engineered constructs. Significant variability was observed in outcome parameters (cell infiltration, proteoglycan and collagen content, and mechanical properties) amongst donors, but these variations did not correlate with patient age or disease condition. Strong correlations were observed between the amount of collagen deposition within the construct and the tensile properties achieved. In scaffolds seeded with particularly robust cells, construct tensile moduli approached maxima of approximately 40 MPa over the 10-week culture period. CONCLUSIONS: This study demonstrates that cells derived from surgical debris are a potent cell source for engineered meniscus constructs. Results further show that robust growth is possible in MDCs from middle-aged and elderly patients, highlighting the potential for therapeutic intervention using autologous cells.


Subject(s)
Biocompatible Materials , Chondrocytes/cytology , Menisci, Tibial/surgery , Tensile Strength , Tissue Engineering/methods , Tissue Scaffolds/statistics & numerical data , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Arthroplasty , Biomechanical Phenomena , Cells, Cultured , Collagen/analysis , Female , Humans , Male , Medical Waste , Middle Aged , Tibial Meniscus Injuries , Time Factors , Tissue Scaffolds/chemistry , Young Adult
17.
Osteoarthritis Cartilage ; 16(9): 1074-82, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18353693

ABSTRACT

OBJECTIVE: The objective of this study was to determine the capacity of chondrocyte- and mesenchymal stem cell (MSC)-laden hydrogel constructs to achieve native tissue tensile properties when cultured in a chemically defined medium supplemented with transforming growth factor-beta3 (TGF-beta3). DESIGN: Cell-laden agarose hydrogel constructs (seeded with bovine chondrocytes or MSCs) were formed as prismatic strips and cultured in a chemically defined serum-free medium in the presence or absence of TGF-beta3. The effects of seeding density (10 vs 30 million cells/mL) and cell type (chondrocyte vs MSC) were evaluated over a 56-day period. Biochemical content, collagenous matrix deposition and localization, and tensile properties (ramp modulus, ultimate strain, and toughness) were assessed biweekly. RESULTS: Results show that the tensile properties of cell-seeded agarose constructs increase with time in culture. However, tensile properties (modulus, ultimate strain, and toughness) achieved on day 56 were not dependent on either the initial seeding density or the cell type employed. When cultured in medium supplemented with TGF-beta3, tensile modulus increased and plateaued at a level of 300-400 kPa for each cell type and starting cell concentration. Ultimate strain and toughness also increased relative to starting values. Collagen deposition increased in constructs seeded with both cell types and at both seeding densities, with exposure to TGF-beta3 resulting in a clear shift toward type II collagen deposition as determined by immunohistochemical staining. CONCLUSIONS: These findings demonstrate that the tensile properties, an important and often overlooked metric of cartilage development, increase with time in culture in engineered hydrogel-based cartilage constructs. Under the free-swelling conditions employed in the present study, tensile moduli and toughness did not match that of the native tissue, though significant time-dependent increases were observed with the inclusion of TGF-beta3. Of note, MSC-seeded constructs achieved tensile properties that were comparable to chondrocyte-seeded constructs, confirming the utility of this alternative cell source in cartilage tissue engineering. Further work, including both modulation of the chemical and mechanical culture environment, is required to optimize the deposition of collagen and its remodeling to achieve tensile properties in engineered constructs matching the native tissue.


Subject(s)
Cartilage/metabolism , Chondrocytes/metabolism , Hydrogels/pharmacology , Mesenchymal Stem Cells/metabolism , Tissue Engineering/methods , Transforming Growth Factor beta3/metabolism , Animals , Cartilage/pathology , Cattle , Cells, Cultured/metabolism , Chondrocytes/pathology , Chondrogenesis/physiology , Collagen/metabolism , Compressive Strength/physiology , Culture Techniques/methods , Immunohistochemistry , Mesenchymal Stem Cells/pathology , Transforming Growth Factor beta3/pharmacology , Weight-Bearing/physiology
18.
Osteoarthritis Cartilage ; 15(9): 1025-33, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17498976

ABSTRACT

OBJECTIVE: To determine whether the functional properties of tissue-engineered constructs cultured in a chemically-defined medium supplemented briefly with TGF-beta3 can be enhanced with the application of dynamic deformational loading. METHODS: Primary immature bovine cells (2-3 months old) were encapsulated in agarose hydrogel (2%, 30 x 10(6)cells/ml) and cultured in chemically-defined medium supplemented for the first 2 weeks with transforming growth factor beta 3 (TGF-beta3) (10 microg/ml). Physiologic deformational loading (1 Hz, 3 h/day, 10% unconfined deformation initially and tapering to 2% peak-to-peak deformation by day 42) was applied either concurrent with or after the period of TGF-beta3 supplementation. Mechanical and biochemical properties were evaluated up to day 56. RESULTS: Dynamic deformational loading applied concurrently with TGF-beta3 supplementation yielded significantly lower (-90%) overall mechanical properties when compared to free-swelling controls. In contrast, the same loading protocol applied after the discontinuation of the growth factor resulted in significantly increased (+10%) overall mechanical properties relative to free-swelling controls. Equilibrium modulus values reach 1306+/-79 kPa and glycosaminoglycan levels reach 8.7+/-1.6% w.w. during this 8-week period and are similar to host cartilage properties (994+/-280 kPa, 6.3+/-0.9% w.w.). CONCLUSIONS: An optimal strategy for the functional tissue engineering of articular cartilage, particularly to accelerate construct development, may incorporate sequential application of different growth factors and applied deformational loading.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/cytology , Stress, Mechanical , Tissue Engineering/methods , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/physiology , Cattle , Cell Culture Techniques , Chondrocytes/physiology , Collagen/analysis , Glycosaminoglycans/analysis , Models, Biological , Transforming Growth Factor beta3
19.
Biomech Model Mechanobiol ; 6(1-2): 113-25, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16691412

ABSTRACT

This study explored the biologic response of chondrocytes and mesenchymal stem cells (MSCs) to a dynamic mechanical loading regime. We developed a time-efficient methodology for monitoring regional changes in extracellular matrix gene transcription using reporter promoter constructs. Specifically, transfected cells were homogenously distributed throughout agarose hydrogel constructs, and spatial and temporal gene expression and the ability to form functional ECM were analyzed in response to dynamic mechanical stimuli. Theoretical analyses were used to predict the physical signals generated within the gel in response to these loading regimes. Using a custom compression bioreactor system, changes in aggrecan and type II collagen promoter activity in transfected chondrocyte-laden cylindrical constructs were evaluated in response to a range of loading frequencies and durations. In general, aggrecan promoter activity increased with increasing duration of loading, particularly in the outer annulus region. Interestingly, type II collagen promoter activity decreased in this annular region under identical loading conditions. In addition, we explored the role of mechanical compression in directing chondrogenic differentiation of MSCs by monitoring short-term aggrecan promoter activity. As an example of long-term utility, a specific loading protocol was applied to MSC-laden constructs for 5 days, and the resultant changes in glycosaminoglycan (GAG) production were evaluated over a 4-week period. This dynamic loading regime increased not only short-term aggrecan transcriptional activity but also GAG deposition in long-term culture. These results demonstrate the utility of a new reporter promoter system for optimizing loading protocols to improve the outcome of engineered chondrocyte- and MSC-laden cartilaginous constructs.


Subject(s)
Cartilage/metabolism , Chondrocytes/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation , Mesenchymal Stem Cells/metabolism , Transcription, Genetic , Weight-Bearing/physiology , Aggrecans/genetics , Animals , Bioreactors , Cattle , Cell Culture Techniques , Chondrocytes/cytology , Collagen Type II/genetics , Compressive Strength , Finite Element Analysis , Gels , Genes, Reporter , Glycosaminoglycans/metabolism , Luciferases, Renilla/metabolism , Mesenchymal Stem Cells/cytology , Promoter Regions, Genetic/genetics
20.
Osteoarthritis Cartilage ; 14(2): 179-89, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16257243

ABSTRACT

BACKGROUND: The developmental history of the chondrocyte results in a cell whose biosynthetic activities are optimized to maintain the concentration and organization of a mechanically functional cartilaginous extracellular matrix. While useful for cartilage tissue engineering studies, the limited supply of healthy autologous chondrocytes may preclude their clinical use. Consequently, multipotential mesenchymal stem cells (MSCs) have been proposed as an alternative cell source. OBJECTIVE: While MSCs undergo chondrogenesis, few studies have assessed the mechanical integrity of their forming matrix. Furthermore, efficiency of matrix formation must be determined in comparison to healthy chondrocytes from the same donor. Given the scarcity of healthy human tissue, this study determined the feasibility of isolating bovine chondrocytes and MSCs, and examined their long-term maturation in three-dimensional agarose culture. EXPERIMENTAL DESIGN: Bovine MSCs were seeded in agarose and induced to undergo chondrogenesis. Mechanical and biochemical properties of MSC-laden constructs were monitored over a 10-week period and compared to those of chondrocytes derived from the same group of animals maintained similarly. RESULTS: Our results show that while chondrogenesis does occur in MSC-laden hydrogels, the amount of the forming matrix and measures of its mechanical properties are lower than that produced by chondrocytes under the same conditions. Furthermore, some important properties, particularly glycosaminoglycan content and equilibrium modulus, plateau with time in MSC-laden constructs, suggesting that diminished capacity is not the result of delayed differentiation. CONCLUSIONS: These findings suggest that while MSCs do generate constructs with substantial cartilaginous properties, further optimization must be done to achieve levels similar to those produced by chondrocytes.


Subject(s)
Chondrocytes/cytology , Chondrogenesis , Mesenchymal Stem Cells/cytology , Animals , Biomechanical Phenomena , Cattle , Cell Culture Techniques , Cell Differentiation , Cell Survival , Cells, Cultured , Chondrocytes/metabolism , Collagen/analysis , Culture Media , DNA/analysis , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Mesenchymal Stem Cells/metabolism , Proteoglycans/analysis , Proteoglycans/metabolism , Sepharose , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...