Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Osteoarthritis Cartilage ; 32(8): 938-949, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38782253

RESUMO

OBJECTIVE: Traumatic meniscal injuries can cause acute pain, hemarthrosis (bleeding into the joint), joint immobility, and post-traumatic osteoarthritis (PTOA). However, the exact mechanism(s) by which PTOA develops following meniscal injuries is unknown. Since meniscus tears commonly coincide with hemarthrosis, investigating the direct effects of blood and its constituents on meniscus tissue is warranted. The goal of this study was to determine the direct effects of blood and blood components on meniscus tissue catabolism. METHODS: Porcine meniscus explants or primary meniscus cells were exposed to whole blood or various fractions of blood for 3 days to simulate blood exposure following injury. Explants were then washed and cultured for an additional 3 days prior to collection for biochemical analyses. RESULTS: Whole blood increased matrix metalloproteinase (MMP) activity. Fractionation experiments revealed blood-derived red blood cells did not affect meniscus catabolism. Conversely, viable mononuclear leukocytes induced MMP activity, nitric oxide (NO) production, and loss of tissue sulfated glycosaminoglycan (sGAG) content, suggesting that these cells are mediating meniscus catabolism. CONCLUSIONS: These findings highlight the potential challenges of meniscus healing in the presence of hemarthrosis and the need for further research to elucidate the in vivo effects of blood and blood-derived mononuclear leukocytes due to both hemarthrosis and blood-derived therapeutics.


Assuntos
Leucócitos Mononucleares , Meniscos Tibiais , Animais , Suínos , Leucócitos Mononucleares/metabolismo , Meniscos Tibiais/metabolismo , Óxido Nítrico/metabolismo , Lesões do Menisco Tibial/metabolismo , Glicosaminoglicanos/metabolismo , Metaloproteinases da Matriz/metabolismo , Células Cultivadas , Menisco/metabolismo , Sangue/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33758095

RESUMO

Osteoarthritis (OA) is a painful and debilitating condition of synovial joints without any disease-modifying therapies [A. M. Valdes, T. D. Spector, Nat. Rev. Rheumatol. 7, 23-32 (2011)]. We previously identified mechanosensitive PIEZO channels, PIEZO1 and PIEZO2, both expressed in articular cartilage, to function in chondrocyte mechanotransduction in response to injury [W. Lee et al., Proc. Natl. Acad. Sci. U.S.A. 111, E5114-E5122 (2014); W. Lee, F. Guilak, W. Liedtke, Curr. Top. Membr. 79, 263-273 (2017)]. We therefore asked whether interleukin-1-mediated inflammatory signaling, as occurs in OA, influences Piezo gene expression and channel function, thus indicative of maladaptive reprogramming that can be rationally targeted. Primary porcine chondrocyte culture and human osteoarthritic cartilage tissue were studied. We found that interleukin-1α (IL-1α) up-regulated Piezo1 in porcine chondrocytes. Piezo1 expression was significantly increased in human osteoarthritic cartilage. Increased Piezo1 expression in chondrocytes resulted in a feed-forward pathomechanism whereby increased function of Piezo1 induced excess intracellular Ca2+ at baseline and in response to mechanical deformation. Elevated resting state Ca2+ in turn rarefied the F-actin cytoskeleton and amplified mechanically induced deformation microtrauma. As intracellular substrates of this OA-related inflammatory pathomechanism, in porcine articular chondrocytes exposed to IL-1α, we discovered that enhanced Piezo1 expression depended on p38 MAP-kinase and transcription factors HNF4 and ATF2/CREBP1. CREBP1 directly bound to the proximal PIEZO1 gene promoter. Taken together, these signaling and genetic reprogramming events represent a detrimental Ca2+-driven feed-forward mechanism that can be rationally targeted to stem the progression of OA.


Assuntos
Condrócitos/metabolismo , Interleucina-1alfa/metabolismo , Canais Iônicos/genética , Mecanotransdução Celular/imunologia , Osteoartrite/imunologia , Fator 2 Ativador da Transcrição/metabolismo , Animais , Cálcio/metabolismo , Cartilagem Articular/citologia , Cartilagem Articular/imunologia , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos/imunologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Canais Iônicos/metabolismo , Mecanotransdução Celular/genética , Osteoartrite/genética , Osteoartrite/patologia , Cultura Primária de Células , Regiões Promotoras Genéticas/genética , Sus scrofa , Regulação para Cima/imunologia
3.
Curr Rheumatol Rep ; 25(2): 35-46, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36479669

RESUMO

PURPOSE OF REVIEW: Meniscus injury often leads to joint degeneration and post-traumatic osteoarthritis (PTOA) development. Therefore, the purpose of this review is to outline the current understanding of biomechanical and biological repercussions following meniscus injury and how these changes impact meniscus repair and PTOA development. Moreover, we identify key gaps in knowledge that must be further investigated to improve meniscus healing and prevent PTOA. RECENT FINDINGS: Following meniscus injury, both biomechanical and biological alterations frequently occur in multiple tissues in the joint. Biomechanically, meniscus tears compromise the ability of the meniscus to transfer load in the joint, making the cartilage more vulnerable to increased strain. Biologically, the post-injury environment is often characterized by an increase in pro-inflammatory cytokines, catabolic enzymes, and immune cells. These multi-faceted changes have a significant interplay and result in an environment that opposes tissue repair and contributes to PTOA development. Additionally, degenerative changes associated with OA may cause a feedback cycle, negatively impacting the healing capacity of the meniscus. Strides have been made towards understanding post-injury biological and biomechanical changes in the joint, their interplay, and how they affect healing and PTOA development. However, in order to improve clinical treatments to promote meniscus healing and prevent PTOA development, there is an urgent need to understand the physiologic changes in the joint following injury. In particular, work is needed on the in vivo characterization of the temporal biomechanical and biological changes that occur in patients following meniscus injury and how these changes contribute to PTOA development.


Assuntos
Artroplastia do Joelho , Cartilagem Articular , Menisco , Osteoartrite , Humanos , Osteoartrite/etiologia , Osteoartrite/metabolismo , Menisco/lesões , Citocinas/metabolismo , Artroplastia do Joelho/efeitos adversos , Cartilagem Articular/metabolismo
4.
Connect Tissue Res ; 61(3-4): 322-337, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31661326

RESUMO

Purpose: Meniscal injury and loss of meniscus tissue lead to osteoarthritis development. Therefore, novel biologic strategies are needed to enhance meniscus tissue repair. The purpose of this study was to identify a favorable culture medium for both bone marrow-derived mesenchymal stem cells (MSCs) and meniscal tissue, and to establish a novel meniscus tissue defect model that could be utilized for in vitro screening of biologics to promote meniscus repair.Materials and Methods: In parallel, we analyzed the biochemical properties of MSC - seeded meniscus-derived matrix (MDM) scaffolds and meniscus repair model explants cultured in different combinations of serum, dexamethasone (Dex), and TGF-ß. Next, we combined meniscus tissue and MSC-seeded MDM scaffolds into a novel meniscus tissue defect model to evaluate the effects of chondrogenic and meniscal media on the tissue biochemical properties and repair strength.Results: Serum-free medium containing TGF-ß and Dex was the most promising formulation for experiments with MSC-seeded scaffolds, whereas serum-containing medium was the most effective for meniscus tissue composition and integrative repair. When meniscus tissue and MSC-seeded MDM scaffolds were combined into a defect model, the chondrogenic medium (serum-free with TGF-ß and Dex) enhanced the production of proteoglycans and promoted integrative repair of meniscus tissue. As well, cross-linked scaffolds improved repair over the MDM slurry.Conclusions: The meniscal tissue defect model established in this paper can be used to perform in vitro screening to identify and optimize biological treatments to enhance meniscus tissue repair prior to conducting preclinical animal studies.


Assuntos
Células da Medula Óssea , Matriz Extracelular/química , Meniscos Tibiais/química , Células-Tronco Mesenquimais , Modelos Biológicos , Lesões do Menisco Tibial , Alicerces Teciduais/química , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Técnicas de Cultura de Células , Feminino , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Suínos , Lesões do Menisco Tibial/metabolismo , Lesões do Menisco Tibial/patologia
5.
Int J Mol Sci ; 21(1)2019 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-31861690

RESUMO

Meniscal injuries, particularly in the avascular zone, have a low propensity for healing and are associated with the development of osteoarthritis. Current meniscal repair techniques are limited to specific tear types and have significant risk for failure. In previous work, we demonstrated the ability of meniscus-derived matrix (MDM) scaffolds to augment the integration and repair of an in vitro meniscus defect. The objective of this study was to determine the effects of percent composition and dehydrothermal (DHT) or genipin cross-linking of MDM bioscaffolds on primary meniscus cellular responses and integrative meniscus repair. In all scaffolds, the porous microenvironment allowed for exogenous cell infiltration and proliferation, as well as endogenous meniscus cell migration. The genipin cross-linked scaffolds promoted extracellular matrix (ECM) deposition and/or retention. The shear strength of integrative meniscus repair was improved with increasing percentages of MDM and genipin cross-linking. Overall, the 16% genipin cross-linked scaffolds were most effective at enhancing integrative meniscus repair. The ability of the genipin cross-linked scaffolds to attract endogenous meniscus cells, promote glycosaminoglycan and collagen deposition, and enhance integrative meniscus repair reveals that these MDM scaffolds are promising tools to augment meniscus healing.


Assuntos
Matriz Extracelular/metabolismo , Iridoides/farmacologia , Menisco/citologia , Engenharia Tecidual/métodos , Animais , Proliferação de Células , Células Cultivadas , Feminino , Menisco/efeitos dos fármacos , Menisco/metabolismo , Resistência ao Cisalhamento , Suínos , Alicerces Teciduais
6.
Connect Tissue Res ; 58(3-4): 305-316, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27813662

RESUMO

PURPOSE: Meniscus tears are a common knee injury and are associated with the development of post-traumatic osteoarthritis (OA). The purpose of this study is to evaluate potential OA mediators in the synovial fluid and serum of meniscus tear subjects compared to those in the synovial fluid of radiographic non-OA control knees. MATERIALS AND METHODS: Sixteen subjects with an isolated unilateral meniscus injury and six subjects who served as reference controls (knee Kellgren-Lawrence grade 0-1) were recruited. Twenty-one biomarkers were measured in serum from meniscus tear subjects and in synovial fluid from both groups. Meniscus tear subjects were further stratified by tear type to assess differences in biomarker levels. RESULTS: Synovial fluid total matrix metalloproteinase (MMP) activity and prostaglandin E2 (PGE2) were increased 25-fold and 290-fold, respectively, in meniscus tear subjects as compared to reference controls (p < 0.05). Synovial fluid MMP activity and PGE2 concentrations were positively correlated in meniscus tear subjects (R = 0.83, p < 0.0001). In meniscus tear subjects, synovial fluid levels of MMP activity, MMP-2, MMP-3, sGAG, COMP, IL-6, and PGE2 were higher than serum levels (p < 0.05). Subjects with complex meniscus tears had higher synovial fluid MMP-10 (p < 0.05) and reduced serum TNFα and IL-8 (p < 0.05) compared to other tear types. CONCLUSIONS: Given the degradative and pro-inflammatory roles of MMP activity and PGE2, these molecules may alter the biochemical environment of the joint. Our findings suggest that modulation of PGE2 signaling, MMP activity, or both following a meniscus injury may be targets to promote meniscus repair and prevent OA development.


Assuntos
Dinoprostona/metabolismo , Metaloproteinases da Matriz/metabolismo , Menisco/lesões , Menisco/metabolismo , Líquido Sinovial/metabolismo , Adulto , Biomarcadores/sangue , Demografia , Feminino , Humanos , Traumatismos do Joelho/sangue , Traumatismos do Joelho/enzimologia , Masculino , Metaloproteinases da Matriz/sangue , Pessoa de Meia-Idade
7.
Proc Natl Acad Sci U S A ; 111(47): E5114-22, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25385580

RESUMO

Diarthrodial joints are essential for load bearing and locomotion. Physiologically, articular cartilage sustains millions of cycles of mechanical loading. Chondrocytes, the cells in cartilage, regulate their metabolic activities in response to mechanical loading. Pathological mechanical stress can lead to maladaptive cellular responses and subsequent cartilage degeneration. We sought to deconstruct chondrocyte mechanotransduction by identifying mechanosensitive ion channels functioning at injurious levels of strain. We detected robust expression of the recently identified mechanosensitive channels, PIEZO1 and PIEZO2. Combined directed expression of Piezo1 and -2 sustained potentiated mechanically induced Ca(2+) signals and electrical currents compared with single-Piezo expression. In primary articular chondrocytes, mechanically evoked Ca(2+) transients produced by atomic force microscopy were inhibited by GsMTx4, a PIEZO-blocking peptide, and by Piezo1- or Piezo2-specific siRNA. We complemented the cellular approach with an explant-cartilage injury model. GsMTx4 reduced chondrocyte death after mechanical injury, suggesting a possible therapy for reducing cartilage injury and posttraumatic osteoarthritis by attenuating Piezo-mediated cartilage mechanotransduction of injurious strains.


Assuntos
Cartilagem Articular/fisiologia , Canais Iônicos/fisiologia , Estresse Mecânico , Animais , Sinalização do Cálcio , Condrócitos/fisiologia , Canais Iônicos/genética , Camundongos , RNA Interferente Pequeno
8.
FASEB J ; 28(6): 2525-37, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24577120

RESUMO

Point mutations in the calcium-permeable TRPV4 ion channel have been identified as the cause of autosomal-dominant human motor neuropathies, arthropathies, and skeletal malformations of varying severity. The objective of this study was to determine the mechanism by which TRPV4 channelopathy mutations cause skeletal dysplasia. The human TRPV4(V620I) channelopathy mutation was transfected into primary porcine chondrocytes and caused significant (2.6-fold) up-regulation of follistatin (FST) expression levels. Pore altering mutations that prevent calcium influx through the channel prevented significant FST up-regulation (1.1-fold). We generated a mouse model of the TRPV4(V620I) mutation, and found significant skeletal deformities (e.g., shortening of tibiae and digits, similar to the human disease brachyolmia) and increases in Fst/TRPV4 mRNA levels (2.8-fold). FST was significantly up-regulated in primary chondrocytes transfected with 3 different dysplasia-causing TRPV4 mutations (2- to 2.3-fold), but was not affected by an arthropathy mutation (1.1-fold). Furthermore, FST-loaded microbeads decreased bone ossification in developing chick femora (6%) and tibiae (11%). FST gene and protein levels were also increased 4-fold in human chondrocytes from an individual natively expressing the TRPV4(T89I) mutation. Taken together, these data strongly support that up-regulation of FST in chondrocytes by skeletal dysplasia-inducing TRPV4 mutations contributes to disease pathogenesis.


Assuntos
Doenças do Desenvolvimento Ósseo/embriologia , Canalopatias/fisiopatologia , Folistatina/fisiologia , Canais de Cátion TRPV/genética , Animais , Doenças do Desenvolvimento Ósseo/genética , Embrião de Galinha , Condrócitos/metabolismo , Humanos , Camundongos , Mutação , Osteocondrodisplasias , Osteogênese/genética , Suínos , Regulação para Cima
9.
Curr Rheumatol Rep ; 16(10): 451, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25182679

RESUMO

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.


Assuntos
Cartilagem Articular/patologia , Condrócitos/patologia , Osteoartrite/patologia , Humanos
10.
Quant Imaging Med Surg ; 14(4): 2738-2746, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38617143

RESUMO

Background: Diffusion magnetic resonance imaging (MRI) allows for the quantification of water diffusion properties in soft tissues. The goal of this study was to characterize the 3D collagen fiber network in the porcine meniscus using high angular resolution diffusion imaging (HARDI) acquisition with both diffusion tensor imaging (DTI) and generalized q-sampling imaging (GQI). Methods: Porcine menisci (n=7) were scanned ex vivo using a three-dimensional (3D) HARDI spin-echo pulse sequence with an isotropic resolution of 500 µm at 7.0 Tesla. Both DTI and GQI reconstruction techniques were used to quantify the collagen fiber alignment and visualize the complex collagen network of the meniscus. The MRI findings were validated with conventional histology. Results: DTI and GQI exhibited distinct fiber orientation maps in the meniscus using the same HARDI acquisition. We found that crossing fibers were only resolved with GQI, demonstrating the advantage of GQI over DTI to visualize the complex collagen fiber orientation in the meniscus. Furthermore, the MRI findings were consistent with conventional histology. Conclusions: HARDI acquisition with GQI reconstruction more accurately resolves the complex 3D collagen architecture of the meniscus compared to DTI reconstruction. In the future, these technologies have the potential to nondestructively assess both normal and abnormal meniscal structure.

11.
J Orthop Res ; 41(7): 1618-1623, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36515295

RESUMO

Orthopaedic research, and biomedical research in general, has made enormous strides to develop treatments for conditions long thought to be inevitable or untreatable; however, there is growing concern about the quality of published research. Considerable efforts have been made to improve overall research quality, integrity, and rigor, including meaningful proposals focused on transparency of reporting, appropriate use of statistics, and reporting of negative results. However, we believe that there is another key component to rigor and reproducibility that is not discussed sufficiently-analytical validation and quality control (QC). In this commentary, we discuss QC and method validation principles and practices that are systematically applied in the clinical laboratory setting to verify and monitor the analytical performance of quantitative assays, and the utility of applying similar practices to biochemical assays in the orthopaedic research setting. This commentary includes (1) recommendations for validation and QC practices, including examples of assay performance limitations uncovered by validation experiments performed in our laboratory, and (2) a description of an ongoing QC program developed to monitor the ongoing performance of commonly used assays in our lab. We hope that this commentary and the examples presented here will be thought-provoking and inspire further discussion and adaptation of analytical validation and QC procedures to advance our shared pursuit of high-quality, rigorous, and reproducible orthopaedic research.


Assuntos
Pesquisa Biomédica , Ortopedia , Reprodutibilidade dos Testes , Controle de Qualidade , Projetos de Pesquisa
12.
Osteoarthr Cartil Open ; 5(3): 100376, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37719442

RESUMO

Objective: To examine the effects of a 6-month weight loss intervention on physical function, inflammatory biomarkers, and metabolic biomarkers in both those with and without osteoarthritis (OA). Design: 59 individuals ≥60 years old with obesity and a functional impairment were enrolled into this IRB approved clinical trial and randomized into one of two 6-month weight loss arms: a higher protein hypocaloric diet or a standard protein hypocaloric diet. All participants were prescribed individualized 500-kcal daily-deficit diets, with a goal of 10% weight loss. Additionally, participants participated in three, low-intensity, exercise sessions per week. Physical function, serum biomarkers and body composition data were assessed at the baseline and 6-month timepoints. Statistical analyses assessed the relationships between biomarkers, physical function, body composition, and OA status as a result of the intervention. Results: No group effects of dietary intervention were detected on any outcome measures (multiple p â€‹> â€‹0.05). During the 6-month trial, participants lost 6.2 â€‹± â€‹4.0% of their bodyweight (p â€‹< â€‹0.0001) and experienced improved physical function on the Short-Performance-Physical-Battery (p â€‹< â€‹0.0001), 8-foot-up-and-go (p â€‹< â€‹0.0001), and time to complete 10-chair-stands (p â€‹< â€‹0.0001). Adiponectin concentrations (p â€‹= â€‹0.0480) were elevated, and cartilage oligomeric matrix protein (COMP) concentrations (p â€‹< â€‹0.0001) were reduced; further analysis revealed that reductions in serum COMP concentrations were greater in OA-negative individuals. Conclusions: These results suggest that weight loss in older adults with and without OA may provide a protective effect to cartilage and OA. In particular, OA-negative individuals may be able to mitigate changes associated with OA through weight loss.

14.
Am J Sports Med ; 50(7): 1997-2006, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35482438

RESUMO

BACKGROUND: Posttraumatic inflammation after joint injury, ranging from sprains to articular fracture, contributes to the development of arthritis, and the administration of interleukin 1 (IL-1) receptor antagonist (IL-1Ra) is a potential intervention to mitigate this response. Although IL-1Ra mitigates cartilage degenerative changes induced by IL-1, lidocaine is used for local pain management in acute joint injury. Intra-articular delivery of both drugs in combination would be a novel and possibly disease-modifying treatment. However, it is not known whether the interaction with lidocaine at clinical concentrations (1%) would alter the efficacy of IL-1Ra to protect cartilage from the catabolic effects of IL-1. HYPOTHESIS: Treatment of articular cartilage with IL-1Ra in combination with a clinically relevant concentration of lidocaine (1%) will inhibit the catabolic effects of IL-1α in a manner similar to treatment with IL-1Ra alone. STUDY DESIGN: Controlled laboratory study. METHODS: Fresh porcine cartilage explants were harvested, challenged with IL-1α, and incubated for 72 hours with IL-1Ra or a combination of IL-1Ra and lidocaine. The primary outcome was total sulfated glycosaminoglycan (sGAG) release. Additional experiments assessed the effect of storage temperature and premixing of IL-1Ra and lidocaine on sGAG release. All explants were histologically assessed for cartilage degradation using a modified Mankin grading scale. RESULTS: The combination of IL-1Ra and lidocaine, premixed at various time points and stored at room temperature or 4°C, was as effective as IL-1Ra alone at inhibiting IL-1α-mediated sGAG release. Mankin histopathology scores supported these findings. CONCLUSION: Our hypothesis was supported, and results indicated that the combination of IL-1Ra and lidocaine was as efficacious as IL-1Ra treatment alone in acutely mitigating biological cartilage injury due to IL-1α in an explant model. CLINICAL SIGNIFICANCE: The combination of IL-1Ra and lidocaine is stable when reagents are stored in advance of administration at varying temperatures, providing clinically relevant information about storage of medications. The ability to premix and store this drug combination for intra-articular delivery may provide a novel treatment after joint injury to provide pain relief and block inflammation-induced catabolism of joint tissues.


Assuntos
Doenças das Cartilagens , Cartilagem Articular , Animais , Doenças das Cartilagens/patologia , Cartilagem Articular/patologia , Humanos , Inflamação/patologia , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Lidocaína/metabolismo , Lidocaína/farmacologia , Suínos
15.
Front Bioeng Biotechnol ; 10: 837619, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35299636

RESUMO

Meniscus injuries are highly prevalent, and both meniscus injury and subsequent surgery are linked to the development of post-traumatic osteoarthritis (PTOA). Although the pathogenesis of PTOA remains poorly understood, the inflammatory cytokine IL-1 is elevated in synovial fluid following acute knee injuries and causes degradation of meniscus tissue and inhibits meniscus repair. Dynamic mechanical compression of meniscus tissue improves integrative meniscus repair in the presence of IL-1 and dynamic tensile strain modulates the response of meniscus cells to IL-1. Despite the promising observed effects of physiologic mechanical loading on suppressing inflammatory responses of meniscus cells, there is a lack of knowledge on the global effects of loading on meniscus transcriptomic profiles. In this study, we compared two established models of physiologic mechanical stimulation, dynamic compression of tissue explants and cyclic tensile stretch of isolated meniscus cells, to identify conserved responses to mechanical loading. RNA sequencing was performed on loaded and unloaded meniscus tissue or isolated cells from inner and outer zones, with and without IL-1. Overall, results from both models showed significant modulation of inflammation-related pathways with mechanical stimulation. Anti-inflammatory effects of loading were well-conserved between the tissue compression and cell stretch models for inner zone; however, the cell stretch model resulted in a larger number of differentially regulated genes. Our findings on the global transcriptomic profiles of two models of mechanical stimulation lay the groundwork for future mechanistic studies of meniscus mechanotransduction, which may lead to the discovery of novel therapeutic targets for the treatment of meniscus injuries.

16.
Connect Tissue Res ; 52(6): 523-33, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21787135

RESUMO

Obesity is one of the primary risk factors for osteoarthritis. Increased adiposity is associated not only with alterations in joint loading, but also with increased systemic and joint concentrations of adipose tissue-derived cytokines, or "adipokines", that promote a state of chronic, low-grade inflammation that may act in concert with other cytokines in the joint to increase joint degeneration. However, the direct effect of adipokines, such as leptin, visfatin, and interleukin-6 (IL-6), on joint tissues, such as articular cartilage and meniscus, are not fully understood. In this study, we examined the hypothesis that these adipokines act synergistically with interleukin-1 (IL-1) to increase catabolism and the production of proinflammatory mediators in cartilage and meniscus. Explants of porcine cartilage and meniscus were treated with physiologically relevant concentrations of leptin, IL-6, or visfatin, alone or in combination with IL-1. Visfatin and IL-1 promoted the catabolic degradation of both cartilage and meniscus, as evidenced by increased metalloproteinase activity, nitric oxide production, and proteoglycan release. However, leptin or IL-6 at physiologic concentrations had no effect on the breakdown of these tissues. These findings suggest that the effects of obesity-induced osteoarthritis may not be through a direct effect of leptin or IL-6 on cartilaginous tissues, but support a potential role for increased visfatin levels in this regard. These data provide an important first step in understanding the role of adipokines in regulating cartilage and meniscus metabolism; however, these adipokines may have different effects in the context of the whole joint and must be evaluated further.


Assuntos
Adipocinas/farmacologia , Cartilagem/efeitos dos fármacos , Cartilagem/metabolismo , Meniscos Tibiais/efeitos dos fármacos , Meniscos Tibiais/metabolismo , Animais , Feminino , Glicosaminoglicanos/metabolismo , Humanos , Interleucina-1/farmacologia , Interleucina-6/farmacologia , Leptina/farmacologia , Metaloproteinases da Matriz/metabolismo , Nicotinamida Fosforribosiltransferase/farmacologia , Óxido Nítrico/biossíntese , Receptores de Interleucina/metabolismo , Solubilidade/efeitos dos fármacos , Sus scrofa , Líquido Sinovial/efeitos dos fármacos , Líquido Sinovial/metabolismo
17.
J Orthop Res ; 39(10): 2177-2186, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33325039

RESUMO

Meniscus injuries are common and a major cause of long-term joint degeneration and disability. Current treatment options are limited, so novel regenerative therapies or tissue engineering strategies are urgently needed. The development of new therapies is hindered by a lack of knowledge regarding the cellular biology of the meniscus and a lack of well-established methods for studying meniscus cells in vitro. The goals of this study were to (1) establish baseline expression profiles and dedifferentiation patterns of inner and outer zone primary meniscus cells, and (2) evaluate the utility of poly(ethylene glycol) diacrylate (PEGDA) and gelatin methacrylate (GelMA) polymer hydrogels to reverse dedifferentiation trends for long-term meniscus cell culture. Using reverse transcription-quantitative polymerase chain reaction, we measured expression levels of putative meniscus phenotype marker genes in freshly isolated meniscus tissue, tissue explant culture, and monolayer culture of inner and outer zone meniscus cells from porcine knees to establish baseline dedifferentiation characteristics, and then compared these expression levels to PEGDA/GelMA embedded passaged meniscus cells. COL1A1 showed robust upregulation, while CHAD, CILP, and COMP showed downregulation with monolayer culture. Expression levels of COL2A1, ACAN, and SOX9 were surprisingly similar between inner and outer zone tissue and were found to be less sensitive as markers of dedifferentiation. When embedded in PEGDA/GelMA hydrogels, expression levels of meniscus cell phenotype genes were significantly modulated by varying the ratio of polymer components, allowing these materials to be tuned for phenotype restoration, meniscus cell culture, and tissue engineering applications.


Assuntos
Materiais Biocompatíveis , Menisco , Animais , Células Cultivadas , Gelatina , Hidrogéis , Fenótipo , Suínos , Engenharia Tecidual/métodos
18.
Cartilage ; 13(2_suppl): 1602S-1607S, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-31609141

RESUMO

OBJECTIVES: The utilization of viral vectors to deliver genes of interest directly to meniscus cells and promote long-term modulation of gene expression may prove useful to enhance meniscus repair and regeneration. The objective of this study was to optimize and compare the potential of lentivirus (LV) and adeno-associated virus (AAV) to deliver transgenes to meniscus cells in both intact meniscus tissue and isolated primary cells in monolayer. DESIGN: Porcine meniscus tissue explants and primary meniscus cells in monolayer were transduced with LV or self-complementary AAV2 (scAAV2) encoding green fluorescent protein (GFP). Following transduction, explants were enzymatically digested to isolate meniscus cells, and monolayer cells were trypsinized. Isolated cells were analyzed by flow cytometry to determine percent transduction. RESULTS: LV and scAAV2 showed a high transduction efficiency in monolayer meniscus cells. scAAV2 was most effective at transducing cells within intact meniscus tissue but the efficiency was less than 20%. Outer zone meniscus cells were more readily transduced by both LV and scAAV2 than the inner zone cells. Higher virus titers and higher cell density resulted in improved transduction efficiency. Polybrene was necessary for the highest transduction efficiency with LV, but it reduced scAAV2 transduction. CONCLUSIONS: Both LV and scAAV2 efficiently transduce primary meniscus cells but only scAAV2 can modestly transduce cells embedded in meniscus tissue. This work lays the foundation for viral gene transfer to be utilized to deliver bioactive transgenes or gene editing machinery, which can induce long-term and tunable expression of therapeutic proteins from tissue-engineered constructs for meniscus repair and regeneration.


Assuntos
Dependovirus , Menisco , Dependovirus/genética , Dependovirus/metabolismo , Edição de Genes , Lentivirus/genética , Engenharia Tecidual
19.
J Biomech ; 129: 110771, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34627074

RESUMO

Changes in cartilage structure and composition are commonly observed during the progression of osteoarthritis (OA). Importantly, quantitative magnetic resonance imaging (MRI) methods, such as T1rho relaxation imaging, can noninvasively provide in vivo metrics that reflect changes in cartilage composition and therefore have the potential for use in early OA detection. Changes in cartilage mechanical properties are also hallmarks of OA cartilage; thus, measurement of cartilage mechanical properties may also be beneficial for earlier OA detection. However, the relative predictive ability of compositional versus mechanical properties in detecting OA has yet to be determined. Therefore, we developed logistic regression models predicting OA status in an ex vivo environment using several mechanical and compositional metrics to assess which metrics most effectively predict OA status. Specifically, in this study the compositional metric analyzed was the T1rho relaxation time, while the mechanical metrics analyzed were the stiffness and recovery (defined as a measure of how quickly cartilage returns to its original shape after loading) of the cartilage. Cartilage recovery had the best predictive ability of OA status both alone and in a multivariate model including the T1rho relaxation time. These findings highlight the potential of cartilage recovery as a non-invasive marker of in vivo cartilage health and motivate future investigation of this metric clinically.


Assuntos
Cartilagem Articular , Osteoartrite do Joelho , Benchmarking , Biomarcadores , Cartilagem Articular/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética
20.
Arthritis Res Ther ; 23(1): 280, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34736523

RESUMO

BACKGROUND: Anterior cruciate ligament (ACL) and meniscus tears are common knee injuries. Despite the high rate of post-traumatic osteoarthritis (PTOA) following these injuries, the contributing factors remain unclear. In this study, we characterized the immune cell profiles of normal and injured joints at the time of ACL and meniscal surgeries. METHODS: Twenty-nine patients (14 meniscus-injured and 15 ACL-injured) undergoing ACL and/or meniscus surgery but with a normal contralateral knee were recruited. During surgery, synovial fluid was aspirated from both normal and injured knees. Synovial fluid cells were pelleted, washed, and stained with an antibody cocktail consisting of fluorescent antibodies for cell surface proteins. Analysis of immune cells in the synovial fluid was performed by polychromatic flow cytometry. A broad spectrum immune cell panel was used in the first 10 subjects. Based on these results, a T cell-specific panel was used in the subsequent 19 subjects. RESULTS: Using the broad spectrum immune cell panel, we detected significantly more total viable cells and CD3 T cells in the injured compared to the paired normal knees. In addition, there were significantly more injured knees with T cells above a 500-cell threshold. Within the injured knees, CD4 and CD8 T cells were able to be differentiated into subsets. The frequency of total CD4 T cells was significantly different among injury types, but no statistical differences were detected among CD4 and CD8 T cell subsets by injury type. CONCLUSIONS: Our findings provide foundational data showing that ACL and meniscus injuries induce an immune cell-rich microenvironment that consists primarily of T cells with multiple T helper phenotypes. Future studies investigating the relationship between immune cells and joint degeneration may provide an enhanced understanding of the pathophysiology of PTOA following joint injury.


Assuntos
Lesões do Ligamento Cruzado Anterior , Reconstrução do Ligamento Cruzado Anterior , Cartilagem Articular , Menisco , Ligamento Cruzado Anterior/cirurgia , Lesões do Ligamento Cruzado Anterior/cirurgia , Humanos , Articulação do Joelho , Imageamento por Ressonância Magnética , Líquido Sinovial
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA