Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Tissue Eng Part A ; 29(5-6): 127-140, 2023 03.
Article in English | MEDLINE | ID: mdl-36458467

ABSTRACT

Objectives: Successful tissue regeneration requires a clinically viable source of mesenchymal stem cells (MSCs). We explored activin receptor-like kinase (ALK)-5 inhibitors to rapidly derive an MSC-like phenotype with high cartilage forming capacity from a xeno-free human embryonic cell line. Methods: Embryonic stem cell (ESC) lines (H9 and HADC100) were treated with the ALK-5 inhibitor SB431542; HADC100 cells were additionally treated with ALK-5 inhibitors SB525334 or GW788388. Cells were then seeded upon human fibronectin in the presence of fibroblast growth factor 2 (FGF2) in a serum-free medium. Flow cytometry was used to assess MSC markers (positive for CD73, CD90, and CD105; negative for CD34 and CD45). Differentiation status was assessed through quantitative polymerase chain reaction. Cartilage forming capacity was determined in high-density pellet cultures, in fibrin gels containing extracellular matrix (fibrin-ECM), and after implantation in ex vivo human osteoarthritic cartilage. Gene expression, histology, and immunostaining were used to assess cartilage phenotype, tissue regeneration, and integration. Results: Exposure to all three ALK-5 inhibitors lead to expression of mesodermal gene markers and differentiation into MSC-like cells (embryonic stem cell-derived mesenchymal stem cells [ES-MSCs]) based on surface marker expression. ES-MSC in pellet cultures or in fibrin-ECM gels expressed high levels of chondrogenic genes: COL2A1, ACAN, and COMP; and low levels of COL1A1 and RUNX2. Cell pellets or fibrin constructs implanted into ex vivo human osteoarthritic cartilage defects produced GAG-rich (safranin O positive) and collagen type II-positive neocartilage tissues that integrated well with native diseased tissue. Conclusions: We developed a protocol for rapid differentiation of xeno-free ESC into MSC-like cells with high cartilage forming capacity with potential for clinical applications. Impact statement Osteoarthritis (OA) is a common disease resulting in significant disability and no approved disease modifying treatment (other than total joint replacement). Embryonic stem cell-derived cell therapy has the potential to benefit patients with cartilage lesions leading to OA and may prevent or delay the need for total joint replacement.


Subject(s)
Human Embryonic Stem Cells , Mesenchymal Stem Cells , Receptor, Transforming Growth Factor-beta Type I , Humans , Cartilage , Cell Differentiation , Cells, Cultured , Chondrogenesis/genetics , Osteoarthritis/metabolism , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors
2.
Front Bioeng Biotechnol ; 10: 810705, 2022.
Article in English | MEDLINE | ID: mdl-35186903

ABSTRACT

Nanofibrous scaffolds fabricated via electrospinning have been proposed for meniscus tissue regeneration. However, the electrospinning process is slow, and can only generate scaffolds of limited thickness with densely packed fibers, which limits cell distribution within the scaffold. In this study, we explored whether pneumatospinning could produce thicker collagen type I fibrous scaffolds with higher porosity, that can support cell infiltration and neo-fibrocartilage tissue formation for meniscus tissue engineering. We pneumatospun scaffolds with solutions of collagen type I with thicknesses of approximately 1 mm in 2 h. Scanning electron microscopy revealed a mix of fiber sizes with diameters ranging from 1 to 30 µm. The collagen scaffold porosity was approximately 48% with pores ranging from 7.4 to 100.7 µm. The elastic modulus of glutaraldehyde crosslinked collagen scaffolds was approximately 45 MPa, when dry, which reduced after hydration to 0.1 MPa. Mesenchymal stem cells obtained from the infrapatellar fat pad were seeded in the scaffold with high viability (>70%). Scaffolds seeded with adipose-derived stem cells and cultured for 3 weeks exhibited a fibrocartilage meniscus-like phenotype (expressing COL1A1, COL2A1 and COMP). Ex vivo implantation in healthy bovine and arthritic human meniscal explants resulted in the development of fibrocartilage-like neotissues that integrated with the host tissue with deposition of glycosaminoglycans and collagens type I and II. Our proof-of-concept study indicates that pneumatospinning is a promising approach to produce thicker biomimetic scaffolds more efficiently that electrospinning, and with a porosity that supports cell growth and neo-tissue formation using a clinically relevant cell source.

3.
Cartilage ; 12(1): 112-120, 2021 01.
Article in English | MEDLINE | ID: mdl-30373381

ABSTRACT

OBJECTIVE: Mouse models are commonly used in research applications due to the relatively low cost, highly characterized strains, as well as the availability of many genetically modified phenotypes. In this study, we characterized an ex vivo murine osteochondral repair model using human infrapatellar fat pad (IPFP) progenitor cells. DESIGN: Femurs from euthanized mice were removed and clamped in a custom multidirectional vise to create cylindrical osteochondral defects 0.5 mm in diameter and 0.5 mm deep in both condyles. The IPFP contains progenitors that are a promising cell source for the repair of osteochondral defects. For proof of concept, human IPFP-derived progenitor cells, from osteoarthritic (OA) patients, cultured as pellets, were implanted into the defects and cultured in serum-free medium with TGFß3 for 3 weeks and then processed for histology and immunostaining. RESULTS: The custom multidirectional vise enabled reproducible creation of osteochondral defects in murine femoral condyles. Implantation of IPFP-derived progenitor cells led to development of cartilaginous tissue with Safranin O staining and deposition of collagen type II in the extracellular matrix. CONCLUSIONS: We showed feasibility in creating ex vivo osteochondral defects and demonstrated the regenerative potential of OA human IPFP-derived progenitors in mouse femurs. The murine model can be used to study the effects of aging and OA on tissue regeneration and to explore molecular mechanisms of cartilage repair using genetically modified mice.


Subject(s)
Adipose Tissue/cytology , Cartilage Diseases/therapy , Cartilage, Articular/transplantation , Stem Cell Transplantation/methods , Tissue Engineering/methods , Animals , Cartilage Diseases/etiology , Femur , Humans , Mice , Models, Biological , Patella/cytology , Proof of Concept Study , Stem Cells
4.
Sci Transl Med ; 12(567)2020 10 28.
Article in English | MEDLINE | ID: mdl-33115953

ABSTRACT

Meniscus tears are common knee injuries and a major osteoarthritis (OA) risk factor. Knowledge gaps that limit the development of therapies for meniscus injury and degeneration concern transcription factors that control the meniscus cell phenotype. Analysis of RNA sequencing data from 37 human tissues in the Genotype-Tissue Expression database and RNA sequencing data from meniscus and articular cartilage showed that transcription factor Mohawk (MKX) is highly enriched in meniscus. In human meniscus cells, MKX regulates the expression of meniscus marker genes, OA-related genes, and other transcription factors, including Scleraxis (SCX), SRY Box 5 (SOX5), and Runt domain-related transcription factor 2 (RUNX2). In mesenchymal stem cells (MSCs), the combination of adenoviral MKX (Ad-MKX) and transforming growth factor-ß3 (TGF-ß3) induced a meniscus cell phenotype. When Ad-MKX-transduced MSCs were seeded on TGF-ß3-conjugated decellularized meniscus scaffold (DMS) and inserted into experimental tears in meniscus explants, they increased glycosaminoglycan content, extracellular matrix interconnectivity, cell infiltration into the DMS, and improved biomechanical properties. Ad-MKX injection into mouse knee joints with experimental OA induced by surgical destabilization of the meniscus suppressed meniscus and cartilage damage, reducing OA severity. Ad-MKX injection into human OA meniscus tissue explants corrected pathogenic gene expression. These results identify MKX as a previously unidentified key transcription factor that regulates the meniscus cell phenotype. The combination of Ad-MKX with TGF-ß3 is effective for differentiation of MSCs to a meniscus cell phenotype and useful for meniscus repair. MKX is a promising therapeutic target for meniscus tissue engineering, repair, and prevention of OA.


Subject(s)
Cartilage, Articular , Homeodomain Proteins/metabolism , Meniscus , Mesenchymal Stem Cells , Osteoarthritis , Animals , Homeodomain Proteins/genetics , Mice , Phenotype , Transcription Factors
5.
Nanomedicine (Lond) ; 15(25): 2517-2538, 2020 10.
Article in English | MEDLINE | ID: mdl-32975146

ABSTRACT

The knee menisci are critical to the long-term health of the knee joint. Because of the high incidence of injury and degeneration, replacing damaged or lost meniscal tissue is extremely clinically relevant. The multiscale architecture of the meniscus results in unique biomechanical properties. Nanofibrous scaffolds are extremely attractive to replicate the biochemical composition and ultrastructural features in engineered meniscus tissue. We review recent advances in electrospinning to generate nanofibrous scaffolds and the current state-of-the-art of electrospun materials for meniscal regeneration. We discuss the importance of cellular function for meniscal tissue engineering and the application of cells derived from multiple sources. We compare experimental models necessary for proof of concept and to support translation. Finally, we discuss future directions and potential for technological innovations.


Subject(s)
Meniscus , Nanofibers , Tissue Engineering , Tissue Scaffolds
6.
Proc Natl Acad Sci U S A ; 117(6): 3135-3143, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31980519

ABSTRACT

The objective of this study was to examine FoxO expression and FoxO function in meniscus. In menisci from human knee joints with osteoarthritis (OA), FoxO1 and 3 expression were significantly reduced compared with normal menisci from young and old normal donors. The expression of FoxO1 and 3 was also significantly reduced in mouse menisci during aging and OA induced by surgical meniscus destabilization or mechanical overuse. Deletion of FoxO1 and combined FoxO1, 3, and 4 deletions induced abnormal postnatal meniscus development in mice and these mutant mice spontaneously displayed meniscus pathology at 6 mo. Mice with Col2Cre-mediated deletion of FoxO3 or FoxO4 had normal meniscus development but had more severe aging-related damage. In mature AcanCreERT2 mice, the deletion of FoxO1, 3, and 4 aggravated meniscus lesions in all experimental OA models. FoxO deletion suppressed autophagy and antioxidant defense genes and altered several meniscus-specific genes. Expression of these genes was modulated by adenoviral FoxO1 in cultured human meniscus cells. These results suggest that FoxO1 plays a key role in meniscus development and maturation, and both FoxO1 and 3 support homeostasis and protect against meniscus damage in response to mechanical overuse and during aging and OA.


Subject(s)
Forkhead Box Protein O1 , Forkhead Box Protein O3 , Knee Joint/metabolism , Meniscus/metabolism , Osteoarthritis/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Female , Forkhead Box Protein O1/analysis , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O3/analysis , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Humans , Male , Meniscus/growth & development , Mice , Mice, Knockout , Middle Aged , Young Adult
7.
Connect Tissue Res ; 61(2): 229-243, 2020 03.
Article in English | MEDLINE | ID: mdl-31134817

ABSTRACT

Purpose: Scaffold-free cartilage tissue engineering circumvents issues with scaffold seeding, potential toxicity response, and impaired host integration. However, precisely controlling and maintaining a scaffold-free construct shape have been challenging. We explored the feasibility of microneedle arrays to print tissue using cellular microspheroids as building blocks.Materials and Methods: Human embryonic-derived mesenchymal stem cells or infrapatellar fat pad mesenchymal stem cells were used to create microspheroids of 500 µm in diameter, which were assembled on microneedle arrays in a predefined arrangement using a robotic system under computer vision. Microspheroids on microneedles were cultured to permit fusion into a tissue construct. Infrapatellar fat pad mesenchymal stem cell constructs were either implanted into chondral defects created in human osteoarthritic cartilage explants or maintained on the microneedle array for 3 weeks. Embryonic-derived mesenchymal stem cell constructs were designed to be press-fit into 3 mm subchondral defects in New Zealand White rabbits and maintained for up to 8 weeks to assess retention, early tissue repair, and more mature cartilage regeneration.Results: Microspheroids of both cell types fused together in culture to form neotissues of predefined shape and size. Infrapatellar fat pad mesenchymal stem cell neotissues expressed high levels of chondrogenic genes and integrated with the surrounding osteoarthritic host cartilage. Embryonic-derived mesenchymal stem cell constructs generated chondrogenic neotissue in vivo as early as 2 weeks and more mature tissue by 8 weeks with increased glycosaminoglycan deposition.Conclusions: We constructed defined scaffold-free shapes by bioprinting and fusing microspheroids. Proof of concept was shown in the repair of ex vivo osteoarthritic human cartilage and in vivo rabbit osteochondral (OC) defects.


Subject(s)
Cartilage , Chondrogenesis , Human Embryonic Stem Cells/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Osteoarthritis , Robotic Surgical Procedures , Tissue Engineering , Aged , Animals , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Female , Human Embryonic Stem Cells/pathology , Humans , Male , Mesenchymal Stem Cells/pathology , Middle Aged , Needles , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/therapy , Rabbits
8.
J Orthop Res ; 36(7): 1947-1958, 2018 07.
Article in English | MEDLINE | ID: mdl-29411909

ABSTRACT

Avascular (Avas) meniscus regeneration remains a challenge, which is partly a consequence of our limited knowledge of the cells that maintain this tissue region. In this study, we utilized microarrays to characterize gene expression profiles of intact human Avas meniscus tissue and of cells following culture expansion. Using these data, we examined various 3D culture conditions to redifferentiate Avas cells toward the tissue phenotype. RNA was isolated from either the tissue directly or following cell isolation and 2 weeks in monolayer culture. RNA was hybridized on human genome arrays. Differentially expressed (DE) genes were identified by ranking analysis. DAVID pathway analysis was performed and visualized using STRING analysis. Quantitative PCR (qPCR) on additional donor menisci (tissues and cells) were used to validate array data. Avas cells cultured in 3D were subjected to qPCR to compare with the array-generated data. A total of 387 genes were DE based on differentiation state (>3-fold change; p < 0.01). In Avas-cultured cells, the upregulated pathways included focal adhesion, ECM-receptor interaction, regulation of actin cytoskeleton, and PDGF Signaling. In 3D-cultured Avas cells, TGFß1 or combinations of TGFß1 and BMP6 were most effective to promote an Avas tissue phenotype. THBS2 and THBS4 expression levels were identified as a means to denote meniscus cell phenotype status. We identified the key gene expression profiles, new markers and pathways involved in characterizing the Avas meniscus phenotype in the native state and during in vitro dedifferentiation and redifferentiation. These data served to screen 3D conditions to generate meniscus-like neotissues. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1947-1958, 2018.


Subject(s)
Gene Expression Profiling , Menisci, Tibial/metabolism , Menisci, Tibial/pathology , Tissue Engineering/methods , Actin Cytoskeleton , Actins/metabolism , Adult , Chondrocytes/metabolism , Collagen Type II/metabolism , Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Female , Genome, Human , Humans , Male , Menisci, Tibial/anatomy & histology , Meniscus , Oligonucleotide Array Sequence Analysis , Phenotype , Platelet-Derived Growth Factor/metabolism , Polymerase Chain Reaction , Signal Transduction , Tissue Array Analysis , Tissue Banks , Transcriptome , Young Adult
9.
Tissue Eng Part A ; 24(1-2): 81-93, 2018 01.
Article in English | MEDLINE | ID: mdl-28463545

ABSTRACT

Hydrogel and electrospun scaffold materials support cell attachment and neotissue development and can be tuned to structurally and mechanically resemble native extracellular matrix by altering either electrospun fiber or hydrogel properties. In this study, we examined meniscus tissue generation from different human cell sources including meniscus cells derived from vascular and avascular regions, human bone marrow-derived mesenchymal stem cells, synovial cells, and cells from the infrapatellar fat pad (IPFP). All cells were seeded onto aligned electrospun collagen type I scaffolds and were optionally encapsulated in a tricomponent hydrogel. Single or multilayered constructs were generated and cultivated in defined medium with selected growth factors for 2 weeks. Cell viability, cell morphology, and gene-expression profiles were monitored using confocal microscopy, scanning electron microscopy, and quantitative polymerase chain reaction (qPCR), respectively. Multilayered constructs were examined with histology, immunohistochemistry, qPCR, and for tensile mechanical properties. For all cell types, TGFß1 and TGFß3 treatment increased COL1A1, COMP, Tenascin C (TNC), and Scleraxis (SCX) gene expression and deposition of collagen type I protein. IPFP cells generated meniscus-like tissues with higher meniscogenic gene expression, mechanical properties, and better cell distribution compared to other cell types studied. We show proof of concept that electrospun collagen scaffolds support neotissue formation and IPFP cells have potential for use in cell-based meniscus regeneration strategies.


Subject(s)
Collagen/chemistry , Meniscus/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Adult , Cell Count , Cells, Cultured , Female , Humans , Male , Microscopy, Electron, Scanning
10.
Connect Tissue Res ; 58(3-4): 259-270, 2017.
Article in English | MEDLINE | ID: mdl-27925477

ABSTRACT

PURPOSE: Meniscus contains heterogeneous populations of cells that have not been fully characterized. Cell phenotype is often lost during culture; however, culture expansion is typically required for tissue engineering. We examined and compared cell-surface molecule expression levels on human meniscus cells from the vascular and avascular regions and articular chondrocytes while documenting changes during culture-induced dedifferentiation. MATERIALS AND METHODS: Expressions of 16 different surface molecules were examined by flow cytometry after monolayer culture for 24 h, 1 week, and 2 weeks. Menisci were also immunostained to document the spatial distributions of selected surface molecules. RESULTS: Meniscus cells and chondrocytes exhibited several similarities in surface molecule profiles with dynamic changes during culture. A greater percentage of meniscal cells were positive for CD14, CD26, CD49c, and CD49f compared to articular chondrocytes. Initially, more meniscal cells from the vascular region were positive for CD90 compared to cells from the avascular region or chondrocytes. Cells from the vascular region also expressed higher levels of CD166 and CD271 compared to cells from the avascular region. CD90, CD166, and CD271-positive cells were predominately perivascular in location. However, CD166-positive cells were also located in the superficial layer and in the adjacent synovial and adipose tissue. CONCLUSIONS: These surface marker profiles provide a target phenotype for differentiation of progenitors in tissue engineering. The spatial location of progenitor cells in meniscus is consistent with higher regenerative capacity of the vascular region, while the surface progenitor subpopulations have potential to be utilized in tears created in the avascular region.


Subject(s)
Meniscus/cytology , Tissue Engineering/methods , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Female , Fluorescence , Humans , Male , Meniscus/blood supply , Middle Aged , Phenotype , Stem Cells/cytology , Stem Cells/metabolism , Time Factors , Transcriptome , Young Adult
11.
Tissue Eng Part A ; 22(5-6): 436-48, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26842062

ABSTRACT

The self-healing capacity of an injured meniscus is limited to the vascularized regions and is especially challenging in the inner avascular regions. As such, we investigated the use of human meniscus cell-seeded electrospun (ES) collagen type I scaffolds to produce meniscal tissue and explored whether these cell-seeded scaffolds can be implanted to repair defects created in meniscal avascular tissue explants. Human meniscal cells (derived from vascular and avascular meniscal tissue) were seeded on ES scaffolds and cultured. Constructs were evaluated for cell viability, gene expression, and mechanical properties. To determine potential for repair of meniscal defects, human meniscus avascular cells were seeded and cultured on aligned ES collagen scaffolds for 4 weeks before implantation. Surgical defects resembling "longitudinal tears" were created in the avascular zone of bovine meniscus and implanted with cell-seeded collagen scaffolds and cultured for 3 weeks. Tissue regeneration and integration were evaluated by histology, immunohistochemistry, mechanical testing, and magentic resonance imaging. Ex vivo implantation with cell-seeded collagen scaffolds resulted in neotissue that was significantly better integrated with the native tissue than acellular collagen scaffolds or untreated defects. Human meniscal cell-seeded ES collagen scaffolds may therefore be useful in facilitating meniscal repair of avascular meniscus tears.


Subject(s)
Collagen/pharmacology , Meniscus/pathology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Wound Healing/drug effects , Animals , Cattle , Cells, Cultured , Collagen/ultrastructure , Disease Models, Animal , Elastic Modulus/drug effects , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Humans , Magnetic Resonance Imaging , Phenotype , Tensile Strength/drug effects
12.
Tissue Eng Part A ; 22(3-4): 286-94, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26729061

ABSTRACT

Retinal ganglion cells (RGCs) are responsible for the transfer of signals from the retina to the brain. As part of the central nervous system, RGCs are unable to regenerate following injury, and implanted cells have limited capacity to orient and integrate in vivo. During development, secreted guidance molecules along with signals from extracellular matrix and the vasculature guide cell positioning, for example, around the fovea, and axon outgrowth; however, these changes are temporally regulated and are not the same in the adult. Here, we combine electrospun cell transplantation scaffolds capable of RGC neurite guidance with thermal inkjet 3D cell printing techniques capable of precise positioning of RGCs on the scaffold surface. Optimal printing parameters are developed for viability, electrophysiological function and, neurite pathfinding. Different media, commonly used to promote RGC survival and growth, were tested under varying conditions. When printed in growth media containing both brain-derived neurotrophic factor (BDNF) and ciliary neurotrophic factor (CNTF), RGCs maintained survival and normal electrophysiological function, and displayed radial axon outgrowth when printed onto electrospun scaffolds. These results demonstrate that 3D printing technology may be combined with complex electrospun surfaces in the design of future retinal models or therapies.


Subject(s)
Neurites/metabolism , Printing, Three-Dimensional , Retinal Ganglion Cells , Tissue Scaffolds/chemistry , Animals , Cells, Cultured , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism
13.
NMR Biomed ; 28(12): 1754-1762, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26527298

ABSTRACT

Bone water exists in different states with the majority bound to the organic matrix and to mineral, and a smaller fraction in 'free' form in the pores of cortical bone. In this study, we aimed to develop and evaluate ultrashort-TE (UTE) MRI techniques for the assessment of T2*, T1 and concentration of collagen-bound and pore water in cortical bone using a 3-T clinical whole-body scanner. UTE MRI, together with an isotope study using tritiated and distilled water (THO-H2O) exchange, as well as gravimetric analysis, were performed on ten sectioned bovine bone samples. In addition, 32 human cortical bone samples were prepared for comparison between the pore water concentration measured with UTE MRI and the cortical porosity derived from micro-computed tomography (µCT). A short T2* of 0.27 ± 0.03 ms and T1 of 116 ± 6 ms were observed for collagen-bound water in bovine bone. A longer T2* of 1.84 ± 0.52 ms and T1 of 527 ± 28 ms were observed for pore water in bovine bone. UTE MRI measurements showed a pore water concentration of 4.7-5.3% by volume and collagen-bound water concentration of 15.7-17.9% in bovine bone. THO-H2O exchange studies showed a pore water concentration of 5.9 ± 0.6% and collagen-bound water concentration of 18.1 ± 2.1% in bovine bone. Gravimetric analysis showed a pore water concentration of 6.3 ± 0.8% and collagen-bound water concentration of 19.2 ± 3.6% in bovine bone. A mineral water concentration of 9.5 ± 0.6% was derived in bovine bone with the THO-H2O exchange study. UTE-measured pore water concentration is highly correlated (R(2) = 0.72, p < 0.0001) with µCT porosity in the human cortical bone study. Both bovine and human bone studies suggest that UTE sequences could reliably measure collagen-bound and pore water concentration in cortical bone using a clinical scanner.


Subject(s)
Body Water/metabolism , Collagen/metabolism , Femur/metabolism , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , Animals , Cattle , Femur/anatomy & histology , Femur/diagnostic imaging , Porosity , Protein Binding , Radiography , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
14.
J Orthop Res ; 33(4): 572-83, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25640671

ABSTRACT

Meniscus injury and degeneration have been linked to the development of secondary osteoarthritis (OA). Therapies that successfully repair or replace the meniscus are, therefore, likely to prevent or delay OA progression. We investigated the novel approach of building layers of aligned polylactic acid (PLA) electrospun (ES) scaffolds with human meniscus cells embedded in extracellular matrix (ECM) hydrogel to lead to formation of neotissues that resemble meniscus-like tissue. PLA ES scaffolds with randomly oriented or aligned fibers were seeded with human meniscus cells derived from vascular or avascular regions. Cell viability, cell morphology, and gene expression profiles were monitored via confocal microscopy, scanning electron microscopy (SEM), and real-time polymerase chain reaction (PCR), respectively. Seeded scaffolds were used to produce multilayered constructs and were examined via histology and immunohistochemistry. Morphology and mechanical properties of PLA scaffolds (with and without cells) were influenced by fiber direction of the scaffolds. Both PLA scaffolds supported meniscus tissue formation with increased COL1A1, SOX9, and COMP, yet no difference in gene expression was found between random and aligned PLA scaffolds. Overall, ES materials, which possess mechanical strength of meniscus and can support neotissue formation, show potential for use in cell-based meniscus regeneration strategies.


Subject(s)
Biocompatible Materials/chemistry , Extracellular Matrix/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Materials Testing/methods , Menisci, Tibial/cytology , Tissue Engineering/methods , Tissue Scaffolds , Adult , Biomechanical Phenomena , Cell Survival , Cells, Cultured , Female , Humans , Male , Menisci, Tibial/ultrastructure , Tensile Strength , Young Adult
15.
Tissue Eng Part A ; 20(1-2): 264-74, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23962090

ABSTRACT

Interaction between chondrocytes and the cartilage extracellular matrix (ECM) is essential for maintaining the cartilage's role as a low-friction and load-bearing tissue. In this study, we examined the influence of cartilage zone-specific ECM on human articular chondrocytes (HAC) in two-dimensional and three-dimensional (3D) environments. Two culture systems were used. SYSTEM 1: HAC were cultured on cell-culture plates that had been precoated with the following ECM molecules for 7 days: decorin, biglycan, tenascin C (superficial zone), collagen type II, hyaluronan (HA) (middle and deep zones), and osteopontin (deep zone). Uncoated standard culture plates were used as controls. Expanded cells were examined for phenotypic changes using real-time polymerase chain reaction. In addition, expanded cells were placed into high-density pellet cultures for 14 days. Neocartilage formation was assessed via gene expression and histology evaluations. SYSTEM 2: HAC that were cultured on untreated plates and encapsulated in a 3D alginate scaffold were mixed with one of the zone-specific ECM molecules. Cell viability, gene expression, and histology assessments were conducted on 14-day-old tissues. In HAC monolayer culture, exposure to decorin, HA, and osteopontin increased COL2A1 and aggrecan messenger RNA (mRNA) levels compared with controls. Biglycan up-regulated aggrecan without a significant impact on COL2A1 expression; Tenascin C reduced COL2A1 expression. Neocartilage formed after preculture on tenascin C and collagen type II expressed higher COL2A1 mRNA compared with control pellets. Preculture of HAC on HA decreased both COL2A1 and aggrecan expression levels compared with controls, which was consistent with histology. Reduced proteoglycan 4 (PRG4) mRNA levels were observed in HAC pellets that had been precultured with biglycan and collagen type II. Exposing HAC to HA directly in 3D-alginate culture most effectively induced neocartilage formation, showing increased COL2A1 and aggrecan, and reduced COL1A1 compared with controls. Decorin treatments increased HAC COL2A1 mRNA levels. These data indicate that an appropriate exposure to cartilage-specific ECM proteins could be used to enhance cartilage formation and to even induce the formation of zone-specific phenotypes to improve cartilage regeneration.


Subject(s)
Cartilage, Articular/metabolism , Chondrogenesis , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Adult , Aggrecans/metabolism , Alginates/pharmacology , Cartilage, Articular/drug effects , Cartilage, Articular/growth & development , Cell Proliferation/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrogenesis/drug effects , Chondrogenesis/genetics , Collagen Type II/metabolism , Extracellular Matrix/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Humans , Male , Organ Specificity/drug effects , Organ Specificity/genetics , Phenotype , Proteoglycans/genetics , Proteoglycans/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Tissue Eng Part A ; 20(3-4): 683-92, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24028447

ABSTRACT

Chondrocytes have been generated in vitro from a range of progenitor cell types and by a number of strategies. However, achieving reconstitution of actual physiologically relevant, appropriately-laminated cartilage in situ that would be applicable to conditions, such as arthritis and cartilage degeneration remains elusive. This lack of success is multifactorial and includes limited cell source, decreased proliferation rate of mature chondrocytes, lack of maintenance of phenotype, reduced matrix synthesis, and poor integration with host tissue. We report an efficient approach for deriving mesenchymal chondroprogenitor cells from human embryonic stem cells. These cells generated tissue containing cartilage-specific matrix proteins that integrated in situ in a partial-thickness defect in ex vivo articular cartilage harvested from human arthritic joints. Given that stem cells provide a virtually inexhaustible supply of starting material and that our technique is easily scalable, cartilaginous tissue primed and grafted in this manner could be suitable for clinical translation.


Subject(s)
Arthritis/pathology , Cartilage, Articular/pathology , Cell Differentiation , Embryonic Stem Cells/cytology , Stem Cell Transplantation , Wound Healing , Biomarkers/metabolism , Cell Line , Chondrogenesis , Collagen Type II/metabolism , Embryonic Stem Cells/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Glycoproteins/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
17.
Acta Biomater ; 9(7): 7218-26, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23523536

ABSTRACT

Meniscus degeneration due to age or injury can lead to osteoarthritis. Although promising, current cell-based approaches show limited success. Here we present three-dimensional methacrylated gelatin (GelMA) scaffolds patterned via projection stereolithography to emulate the circumferential alignment of cells in native meniscus tissue. Cultured human avascular zone meniscus cells from normal meniscus were seeded on the scaffolds. Cell viability was monitored, and new tissue formation was assessed by gene expression analysis and histology after 2weeks in serum-free culture with transforming growth factor ß1 (10ngml(-1)). Light, confocal and scanning electron microscopy were used to observe cell-GelMA interactions. Tensile mechanical testing was performed on unseeded, fresh scaffolds and 2-week-old cell-seeded and unseeded scaffolds. 2-week-old cell-GelMA constructs were implanted into surgically created meniscus defects in an explant organ culture model. No cytotoxic effects were observed 3weeks after implantation, and cells grew and aligned to the patterned GelMA strands. Gene expression profiles and histology indicated promotion of a fibrocartilage-like meniscus phenotype, and scaffold integration with repair tissue was observed in the explant model. We show that micropatterned GelMA scaffolds are non-toxic, produce organized cellular alignment, and promote meniscus-like tissue formation. Prefabrication of GelMA scaffolds with architectures mimicking the meniscus collagen bundle organization shows promise for meniscal repair. Furthermore, the technique presented may be scaled up to repair larger defects.


Subject(s)
Lenses , Lighting/instrumentation , Menisci, Tibial/cytology , Menisci, Tibial/growth & development , Molecular Imprinting/instrumentation , Tissue Engineering/instrumentation , Tissue Scaffolds , Adolescent , Adult , Cells, Cultured , Equipment Design , Equipment Failure Analysis , Female , Humans , Male , Signal Processing, Computer-Assisted/instrumentation , Young Adult
18.
Orthop Nurs ; 32(1): 25-36; quiz 37-8, 2013.
Article in English | MEDLINE | ID: mdl-23344487

ABSTRACT

Osteoarthritis (OA) is a disease of the joint, and age is the major risk factor for its development. Clinical manifestation of OA includes joint pain, stiffness, and loss of mobility. Currently, no pharmacological treatments are available to treat this specific joint disease; only symptom-modifying drugs are available. Improvement in imaging technology, identification of biomarkers, and increased understanding of the molecular basis of OA will aid in detecting the early stages of disease. Yet the development of interventional strategies remains elusive and will be critical for effective prevention of OA-associated joint destruction. The potential of cell-based therapies may be applicable in improving joint function in mild to more advanced cases of OA. Ongoing studies to understand the basis of this disease will eventually lead to prevention and treatment strategies and will also be a key in reducing the social and economic burden of this disease. Nurses are advised to provide an integrative approach of disease assessment and management in OA patients' care with a focus on education and implementation. Knowledge and understanding of OA and how this affects the individual patient form the basis for such an integrative approach to all-round patient care and disease management.


Subject(s)
Osteoarthritis , Education, Nursing, Continuing , Female , Humans , Magnetic Resonance Imaging , Male , Nurse's Role , Osteoarthritis/diagnosis , Osteoarthritis/physiopathology , Osteoarthritis/therapy , Risk Factors
19.
Arthritis Rheum ; 65(2): 418-28, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23124445

ABSTRACT

OBJECTIVE: To identify novel genes and pathways specific to the superficial zone (SZ), middle zone (MZ), and deep zone (DZ) of normal articular cartilage. METHODS: Articular cartilage was obtained from the knees of 4 normal human donors. The cartilage zones were dissected on a microtome. RNA was analyzed on human genome arrays. The zone-specific DNA array data obtained from human tissue were compared to array data obtained from bovine cartilage. Genes differentially expressed between zones were evaluated using direct annotation for structural or functional features, and by enrichment analysis for integrated pathways or functions. RESULTS: The greatest differences in genome-wide RNA expression data were between the SZ and DZ in both human and bovine cartilage. The MZ, being a transitional zone between the SZ and DZ, thereby shared some of the same pathways as well as structural/functional features of the adjacent zones. Cellular functions and biologic processes that were enriched in the SZ relative to the DZ included, most prominently, extracellular matrix-receptor interactions, cell adhesion molecule functions, regulation of actin cytoskeleton, ribosome-related functions, and signaling aspects such as the IFN, IL4, Cdc42/Rac, and JAK/STAT signaling pathways. Two pathways were enriched in the DZ relative to the SZ, including PPARG and EGFR/SMRTE. CONCLUSION: These differences in cartilage zonal gene expression identify new markers and pathways that govern the unique differentiation status of chondrocyte subpopulations.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Gene Expression , Knee Joint/metabolism , Animals , Cartilage, Articular/cytology , Cattle , Chondrocytes/cytology , Humans , Knee Joint/cytology , Organ Specificity
20.
Tissue Eng Part A ; 18(17-18): 1784-92, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22536910

ABSTRACT

Dynamic loading and perfusion culture environments alone are known to enhance cartilage extracellular matrix (ECM) production in dedifferentiated articular chondrocytes. In this study, we explored whether a combination of these factors would enhance these processes over a free-swelling (FS) condition using adult human articular chondrocytes embedded in 2% alginate. The alginate constructs were placed into a bioreactor for perfusion (P) only (100 µL/per minute) or perfusion and dynamic compressive loading (PL) culture (20% for 1 h, at 0.5 Hz), each day. Control FS alginate gels were maintained in six-well static culture. Gene expression analysis was conducted on days 7 and 14, while cell viability, immunostaining, and mechanical property testing were performed on day 14 only. Total glycosaminoglycan (GAG) content and GAG synthesis were assessed after 14 days. Col2a1 mRNA expression levels were significantly higher (at least threefold; p<0.05) in both bioreactor conditions compared with FS by days 7 and 14. For all gene studies, no significant differences were seen between P and PL treatments. Aggrecan mRNA levels were not significantly altered in any condition although both GAG/DNA and (35)S GAG incorporation studies indicated higher GAG retention and synthesis in the FS treatment. Collagen type II protein deposition was low in all samples, link protein distribution was more diffuse in FS condition, and aggrecan deposition was located in the outer regions of the alginate constructs in both bioreactor conditions, yet more uniformly in the FS condition. Catabolic gene expression (matrix metalloproteinase 3 [MMP3] and inducible nitric oxide synthase [iNOS]) was higher in bioreactor conditions compared with FS, although iNOS expression levels decreased to approximately fourfold less than the FS condition by day 14. Our data indicate that conditions created in the bioreactor enhanced both anabolic and catabolic responses, similar to other loading studies. Perfusion was sufficient alone to promote this dual response. PL increased the deposition of aggrecan surrounding cells compared with the other conditions; however, overall low GAG retention in the bioreactor system was likely due to both perfusion and catabolic conditions created. Optimal conditions, which permit appropriate anabolic and catabolic processes for accumulation of ECM and tissue remodeling for neocartilage development, specifically for humans, are needed.


Subject(s)
Alginates/pharmacology , Cartilage, Articular/physiology , Chondrogenesis/drug effects , Hydrogels/pharmacology , Perfusion/methods , Adolescent , Adult , Bioreactors , Cartilage, Articular/cytology , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , DNA/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Glucuronic Acid/pharmacology , Glycosaminoglycans/metabolism , Hexuronic Acids/pharmacology , Humans , Immunohistochemistry , Male , Middle Aged , Weight-Bearing/physiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL