Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Stem Cells ; 39(11): 1447-1456, 2021 11.
Article in English | MEDLINE | ID: mdl-34427363

ABSTRACT

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


Subject(s)
Chondrogenesis , Induced Pluripotent Stem Cells , TRPV Cation Channels , Animals , Cartilage/metabolism , Cell Differentiation , Cells, Cultured , Chondrocytes , Chondrogenesis/genetics , Induced Pluripotent Stem Cells/metabolism , Mice , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
2.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33758095

ABSTRACT

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.


Subject(s)
Chondrocytes/metabolism , Interleukin-1alpha/metabolism , Ion Channels/genetics , Mechanotransduction, Cellular/immunology , Osteoarthritis/immunology , Activating Transcription Factor 2/metabolism , Animals , Calcium/metabolism , Cartilage, Articular/cytology , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/immunology , Female , Gene Knockdown Techniques , Humans , Ion Channels/metabolism , Mechanotransduction, Cellular/genetics , Osteoarthritis/genetics , Osteoarthritis/pathology , Primary Cell Culture , Promoter Regions, Genetic/genetics , Sus scrofa , Up-Regulation/immunology
3.
Sci Rep ; 11(1): 1626, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33452305

ABSTRACT

Osteoarthritis is a debilitating disease characterized by cartilage degradation and altered cartilage mechanical properties. Furthermore, it is well established that obesity is a primary risk factor for osteoarthritis. The purpose of this study was to investigate the influence of obesity on the mechanical properties of murine knee cartilage. Two-month old wild type mice were fed either a normal diet or a high fat diet for 16 weeks. Atomic force microscopy-based nanoindentation was used to quantify the effective indentation modulus of medial femoral condyle cartilage. Osteoarthritis progression was graded using the OARSI system. Additionally, collagen organization was evaluated with picrosirius red staining imaged using polarized light microscopy. Significant differences between diet groups were assessed using t tests with p < 0.05. Following 16 weeks of a high fat diet, no significant differences in OARSI scoring were detected. However, we detected a significant difference in the effective indentation modulus between diet groups. The reduction in cartilage stiffness is likely the result of disrupted collagen organization in the superficial zone, as indicated by altered birefringence on polarized light microscopy. Collectively, these results suggest obesity is associated with changes in knee cartilage mechanical properties, which may be an early indicator of disease progression.


Subject(s)
Cartilage, Articular/metabolism , Collagen/metabolism , Elastic Modulus , Obesity/pathology , Animals , Cartilage, Articular/pathology , Diet, High-Fat , Disease Models, Animal , Glucose Tolerance Test , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Atomic Force , Obesity/complications , Obesity/metabolism , Osteoarthritis/etiology , Osteoarthritis/metabolism , Osteoarthritis/pathology , SOX9 Transcription Factor/metabolism
4.
Proc Natl Acad Sci U S A ; 116(6): 1992-1997, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30674675

ABSTRACT

Microarchitectural cues drive aligned fibrillar collagen deposition in vivo and in biomaterial scaffolds, but the cell-signaling events that underlie this process are not well understood. Utilizing a multicellular patterning model system that allows for observation of intracellular signaling events during collagen matrix assembly, we investigated the role of calcium (Ca2+) signaling in human mesenchymal stem cells (MSCs) during this process. We observed spontaneous Ca2+ oscillations in MSCs during fibrillar collagen assembly, and hypothesized that the transient receptor potential vanilloid 4 (TRPV4) ion channel, a mechanosensitive Ca2+-permeable channel, may regulate this signaling. Inhibition of TRPV4 nearly abolished Ca2+ signaling at initial stages of collagen matrix assembly, while at later times had reduced but significant effects. Importantly, blocking TRPV4 activity dramatically reduced aligned collagen fibril assembly; conversely, activating TRPV4 accelerated aligned collagen formation. TRPV4-dependent Ca2+ oscillations were found to be independent of pattern shape or subpattern cell location, suggesting this signaling mechanism is necessary for aligned collagen formation but not sufficient in the absence of physical (microarchitectural) cues that force multicellular alignment. As cell-generated mechanical forces are known to be critical to the matrix assembly process, we examined the role of TRPV4-mediated Ca2+ signaling in force generated across the load-bearing focal adhesion protein vinculin within MSCs using an FRET-based tension sensor. Inhibiting TRPV4 decreased tensile force across vinculin, whereas TRPV4 activation caused a dynamic unloading and reloading of vinculin. Together, these findings suggest TRPV4 activity regulates forces at cell-matrix adhesions and is critical to aligned collagen matrix assembly by MSCs.


Subject(s)
Calcium Signaling/physiology , Collagen/biosynthesis , Mesenchymal Stem Cells/metabolism , TRPV Cation Channels/metabolism , Vinculin/metabolism , Bone Marrow Cells , Calcium , Cell-Matrix Junctions/metabolism , Cellular Microenvironment , Extracellular Matrix , Focal Adhesions , Humans
5.
Arthritis Rheumatol ; 67(5): 1286-94, 2015 May.
Article in English | MEDLINE | ID: mdl-25604429

ABSTRACT

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


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Collagen Type VI/genetics , Extracellular Matrix/metabolism , Mechanotransduction, Cellular/genetics , Osmotic Pressure , TRPV Cation Channels/metabolism , Animals , Collagen Type VI/metabolism , Heparan Sulfate Proteoglycans/metabolism , Mice , Mice, Knockout , Microscopy, Confocal
6.
Naunyn Schmiedebergs Arch Pharmacol ; 388(4): 437-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25519495

ABSTRACT

Biomechanical factors play a critical role in regulating the physiology as well as the pathology of multiple joint tissues and have been implicated in the pathogenesis of osteoarthritis. Therefore, the mechanisms by which cells sense and respond to mechanical signals may provide novel targets for the development of disease-modifying osteoarthritis drugs (DMOADs). Transient receptor potential vanilloid 4 (TRPV4) is a Ca(2+)-permeable cation channel that serves as a sensor of mechanical or osmotic signals in several musculoskeletal tissues, including cartilage, bone, and synovium. The importance of TRPV4 in joint homeostasis is apparent in patients harboring TRPV4 mutations, which result in the development of a spectrum of skeletal dysplasias and arthropathies. In addition, the genetic knockout of Trpv4 results in the development of osteoarthritis and decreased osteoclast function. In engineered cartilage replacements, chemical activation of TRPV4 can reproduce many of the anabolic effects of mechanical loading to accelerate tissue growth and regeneration. Overall, TRPV4 plays a key role in transducing mechanical, pain, and inflammatory signals within joint tissues and thus is an attractive therapeutic target to modulate the effects of joint diseases. In pathological conditions in the joint, when the delicate balance of TRPV4 activity is altered, a variety of different tools could be utilized to directly or indirectly target TRPV4 activity.


Subject(s)
Joint Diseases/metabolism , TRPV Cation Channels/metabolism , Animals , Bone and Bones/metabolism , Cartilage/metabolism , Humans , Joint Diseases/drug therapy , Joints/metabolism , Pain/metabolism
7.
Proc Natl Acad Sci U S A ; 111(47): E5114-22, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385580

ABSTRACT

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.


Subject(s)
Cartilage, Articular/physiology , Ion Channels/physiology , Stress, Mechanical , Animals , Calcium Signaling , Chondrocytes/physiology , Ion Channels/genetics , Mice , RNA, Small Interfering
8.
Curr Rheumatol Rep ; 16(10): 451, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25182679

ABSTRACT

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


Subject(s)
Cartilage, Articular/pathology , Chondrocytes/pathology , Osteoarthritis/pathology , Humans
9.
FASEB J ; 28(6): 2525-37, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24577120

ABSTRACT

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.


Subject(s)
Bone Diseases, Developmental/embryology , Channelopathies/physiopathology , Follistatin/physiology , TRPV Cation Channels/genetics , Animals , Bone Diseases, Developmental/genetics , Chick Embryo , Chondrocytes/metabolism , Humans , Mice , Mutation , Osteochondrodysplasias , Osteogenesis/genetics , Swine , Up-Regulation
10.
Proc Natl Acad Sci U S A ; 111(4): 1316-21, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24474754

ABSTRACT

Mechanical loading of joints plays a critical role in maintaining the health and function of articular cartilage. The mechanism(s) of chondrocyte mechanotransduction are not fully understood, but could provide important insights into new physical or pharmacologic therapies for joint diseases. Transient receptor potential vanilloid 4 (TRPV4), a Ca(2+)-permeable osmomechano-TRP channel, is highly expressed in articular chondrocytes, and loss of TRPV4 function is associated with joint arthropathy and osteoarthritis. The goal of this study was to examine the hypothesis that TRPV4 transduces dynamic compressive loading in articular chondrocytes. We first confirmed the presence of physically induced, TRPV4-dependent intracellular Ca(2+) signaling in agarose-embedded chondrocytes, and then used this model system to study the role of TRPV4 in regulating the response of chondrocytes to dynamic compression. Inhibition of TRPV4 during dynamic loading prevented acute, mechanically mediated regulation of proanabolic and anticatabolic genes, and furthermore, blocked the loading-induced enhancement of matrix accumulation and mechanical properties. Furthermore, chemical activation of TRPV4 by the agonist GSK1016790A in the absence of mechanical loading similarly enhanced anabolic and suppressed catabolic gene expression, and potently increased matrix biosynthesis and construct mechanical properties. These findings support the hypothesis that TRPV4-mediated Ca(2+) signaling plays a central role in the transduction of mechanical signals to support cartilage extracellular matrix maintenance and joint health. Moreover, these insights raise the possibility of therapeutically targeting TRPV4-mediated mechanotransduction for the treatment of diseases such as osteoarthritis, as well as to enhance matrix formation and functional properties of tissue-engineered cartilage as an alternative to bioreactor-based mechanical stimulation.


Subject(s)
Chondrocytes/metabolism , Mechanotransduction, Cellular/physiology , TRPV Cation Channels/physiology , Animals , Cells, Cultured , Chondrocytes/cytology , Gene Expression Regulation , Sepharose , Swine
11.
Rare Dis ; 2(1): e962971, 2014.
Article in English | MEDLINE | ID: mdl-26942100

ABSTRACT

Transient Receptor Potential Vanilloid 4 (TRPV4) is a mechano- and osmosensitive cation channel that is highly expressed in chondrocytes, the cells in cartilage. A large number of mutations in TRPV4 have been linked to skeletal dysplasias, and the goal of this addendum is to shed light on the mechanisms by which mutations in TRPV4 can cause skeletal dysplasias by focusing on 3 recent publications. These papers suggest that skeletal dysplasia-causing TRPV4 mutations reprogram chondrocytes to increase follistatin production, which inhibits BMP signaling, thus slowing the process of endochondral ossification and leading to skeletal dysplasia. In spite of these important advances in our understanding of the disease mechanism, much remains to be elucidated. Nonetheless, these new data suggest that inhibiting aberrant TRPV4 activity in the cartilage may be a promising direction for therapeutic intervention.

12.
Arthritis Rheum ; 65(10): 2615-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23818303

ABSTRACT

OBJECTIVE: Obesity is an important risk factor for osteoarthritis (OA) and is associated with changes in both the biomechanical and inflammatory environments within the joint. However, the relationship between obesity and cartilage deformation is not fully understood. The goal of this study was to determine the effects of body mass index (BMI) on the magnitude of diurnal cartilage strain in the knee. METHODS: Three-dimensional maps of knee cartilage thickness were developed from 3T magnetic resonance images of the knees of asymptomatic age- and sex-matched subjects with normal BMI (18.5-24.9 kg/m2) or high BMI (25-31 kg/m2). Site-specific magnitudes of diurnal cartilage strain were determined using aligned images recorded at 8:00 AM and 4:00 PM on the same day. RESULTS: Subjects with high BMI had significantly thicker cartilage on both the patella and femoral groove, as compared to subjects with normal BMI. Diurnal cartilage strains were dependent on location in the knee joint, as well as BMI. Subjects with high BMI, compared to those with normal BMI, exhibited significantly higher compressive strains in the tibial cartilage. Cartilage thickness on both femoral condyles decreased significantly from the AM to the PM time point; however, there was no significant effect of BMI on diurnal cartilage strain in the femur. CONCLUSION: Increased BMI is associated with increased diurnal strains in articular cartilage of both the medial and lateral compartments of the knee. The increased cartilage strains observed in individuals with high BMI may, in part, explain the elevated risk of OA associated with obesity or may reflect alterations in the cartilage mechanical properties in subjects with high BMI.


Subject(s)
Body Mass Index , Cartilage, Articular/physiopathology , Circadian Rhythm/physiology , Knee Joint/physiopathology , Obesity/physiopathology , Weight-Bearing/physiology , Cartilage, Articular/pathology , Case-Control Studies , Female , Humans , Knee Joint/pathology , Magnetic Resonance Imaging , Male , Obesity/complications , Osteoarthritis, Knee/epidemiology , Osteoarthritis, Knee/physiopathology , Risk Factors , Stress, Mechanical
13.
J Orthop Res ; 31(7): 1039-45, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23483596

ABSTRACT

Cartilage degeneration with osteoarthritis (OA) is believed to involve the activities of interleukin-1 (IL-1), which exists as alpha and beta isoforms. The goal of this study was to measure the concentrations of both isoforms of IL-1 in the synovial fluid of normal and spontaneously osteoarthritic porcine knees, and to test the hypothesis that physiologic concentrations of IL-1α and IL-1ß exhibit different potencies in activating calcium signaling, the production of matrix metalloproteinases and nitric oxide, and the loss of proteoglycans and tissue mechanical properties in cartilage and meniscus. Median concentrations of IL-1α were 0.043 ng/ml with mild OA and 0.288 ng/ml with moderate OA, whereas IL-1ß concentrations were 0.109 ng/ml with mild OA and 0.122 ng/ml with moderate OA. Both isoforms induced calcium signaling in chondrocytes and meniscal cells at all concentrations. Overall, cartilage and meniscus catabolism was significantly more sensitive to IL-1α than IL-1ß at concentrations of 1 ng/ml or less, while few differences were observed between the two forms at 10 ng/ml. These data provide a range of physiologic IL-1 concentrations that can serve as a framework for the comparison of various in vitro studies, as well as providing further insight for the development of anti-cytokine therapies for OA.


Subject(s)
Cartilage, Articular/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Menisci, Tibial/metabolism , Osteoarthritis, Knee/metabolism , Synovial Fluid/metabolism , Animals , Biomechanical Phenomena , Calcium Signaling/physiology , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cells, Cultured , Disease Models, Animal , Female , Interleukin-1alpha/pharmacology , Interleukin-1beta/pharmacology , Matrix Metalloproteinases/metabolism , Menisci, Tibial/cytology , Menisci, Tibial/drug effects , Proteoglycans/metabolism , Severity of Illness Index , Swine
14.
J Biomech ; 46(3): 541-7, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23102493

ABSTRACT

Due to the biphasic viscoelastic nature of cartilage, joint loading may result in deformations that require times on the order of hours to fully recover. Thus, cartilaginous tissues may exhibit cumulative strain over the course of each day. The goal of this study was to assess the magnitude and spatial distribution of strain in the articular cartilage of the knee with daily activity. Magnetic resonance (MR) images of 10 asymptomatic subjects (six males and four females) with mean age of 29 years were obtained at 8:00 AM and 4:00 PM on the same day using a 3T magnet. These images were used to create 3D models of the femur, tibia, and patella from which cartilage thickness distributions were quantified. Cartilage thickness generally decreased from AM to PM in all areas except the patellofemoral groove and was associated with significant compressive strains in the medial condyle and tibial plateau. From AM to PM, cartilage of the medial tibial plateau exhibited a compressive strain of -5.1±1.0% (mean±SEM) averaged over all locations, while strains in the lateral plateau were slightly lower (-3.1±0.6%). Femoral cartilage showed an average strain of -1.9±0.6%. The findings of this study show that human knee cartilage undergoes diurnal changes in strain that vary with site in the joint. Since abnormal joint loading can be detrimental to cartilage homeostasis, these data provide a baseline for future studies investigating the effects of altered biomechanics on diurnal cartilage strains and cartilage physiology.


Subject(s)
Cartilage, Articular/physiology , Circadian Rhythm/physiology , Knee Joint/physiology , Activities of Daily Living , Adult , Cartilage, Articular/anatomy & histology , Female , Humans , Knee Joint/anatomy & histology , Male , Middle Aged , Weight-Bearing/physiology
15.
J Biomech Eng ; 134(10): 101005, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23083196

ABSTRACT

One of the primary limitations of cell therapy for myocardial infarction is the low survival of transplanted cells, with a loss of up to 80% of cells within 3 days of delivery. The aims of this study were to investigate the distribution of nutrients and oxygen in infarcted myocardium and to quantify how macromolecular transport properties might affect cell survival. Transmural myocardial infarction was created by controlled cryoablation in pigs. At 30 days post-infarction, oxygen and metabolite levels were measured in the peripheral skeletal muscle, normal myocardium, the infarct border zone, and the infarct interior. The diffusion coefficients of fluorescein or FITC-labeled dextran (0.3-70 kD) were measured in these tissues using fluorescence recovery after photobleaching. The vascular density was measured via endogenous alkaline phosphatase staining. To examine the influence of these infarct conditions on cells therapeutically used in vivo, skeletal myoblast survival and differentiation were studied in vitro under the oxygen and glucose concentrations measured in the infarct tissue. Glucose and oxygen concentrations, along with vascular density were significantly reduced in infarct when compared to the uninjured myocardium and infarct border zone, although the degree of decrease differed. The diffusivity of molecules smaller than 40 kD was significantly higher in infarct center and border zone as compared to uninjured heart. Skeletal myoblast differentiation and survival were decreased stepwise from control to hypoxia, starvation, and ischemia conditions. Although oxygen, glucose, and vascular density were significantly reduced in infarcted myocardium, the rate of macromolecular diffusion was significantly increased, suggesting that diffusive transport may not be inhibited in infarct tissue, and thus the supply of nutrients to transplanted cells may be possible. in vitro studies mimicking infarct conditions suggest that increasing nutrients available to transplanted cells may significantly increase their ability to survive in infarct.


Subject(s)
Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Oxygen/metabolism , Animals , Biological Transport , Cell Death , Cell Differentiation , Cell Hypoxia , Cell Line , Cell Proliferation , Diffusion , Glucose/metabolism , Mice , Myoblasts, Skeletal/pathology , Myocardium/pathology , Swine
16.
PLoS One ; 7(3): e33397, 2012.
Article in English | MEDLINE | ID: mdl-22448243

ABSTRACT

Mutation or loss of collagen VI has been linked to a variety of musculoskeletal abnormalities, particularly muscular dystrophies, tissue ossification and/or fibrosis, and hip osteoarthritis. However, the role of collagen VI in bone and cartilage structure and function in the knee is unknown. In this study, we examined the role of collagen VI in the morphology and physical properties of bone and cartilage in the knee joint of Col6a1(-/-) mice by micro-computed tomography (microCT), histology, atomic force microscopy (AFM), and scanning microphotolysis (SCAMP). Col6a1(-/-) mice showed significant differences in trabecular bone structure, with lower bone volume, connectivity density, trabecular number, and trabecular thickness but higher structure model index and trabecular separation compared to Col6a1(+/+) mice. Subchondral bone thickness and mineral content increased significantly with age in Col6a1(+/+) mice, but not in Col6a1(-/-) mice. Col6a1(-/-) mice had lower cartilage degradation scores, but developed early, severe osteophytes compared to Col6a1(+/+) mice. In both groups, cartilage roughness increased with age, but neither the frictional coefficient nor compressive modulus of the cartilage changed with age or genotype, as measured by AFM. Cartilage diffusivity, measured via SCAMP, varied minimally with age or genotype. The absence of type VI collagen has profound effects on knee joint structure and morphometry, yet minimal influences on the physical properties of the cartilage. Together with previous studies showing accelerated hip osteoarthritis in Col6a1(-/-) mice, these findings suggest different roles for collagen VI at different sites in the body, consistent with clinical data.


Subject(s)
Bone Density , Cartilage, Articular/physiopathology , Collagen Type VI/physiology , Knee Joint/physiopathology , Osteoarthritis/physiopathology , Animals , Elasticity , Female , Immunoenzyme Techniques , Male , Mice , Mice, Knockout , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Osteoarthritis/etiology , X-Ray Microtomography
17.
Biochem Biophys Res Commun ; 408(2): 230-5, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21463604

ABSTRACT

Osmotic stress is a potent regulator of biological function in many cell types, but its mechanism of action is only partially understood. In this study, we examined whether changes in extracellular osmolality can alter chromatin condensation and the rate of nucleocytoplasmic transport, as potential mechanisms by which osmotic stress can act. Transport of 10 kDa dextran was measured both within and between the nucleus and the cytoplasm using two different photobleaching methods. A mathematical model was developed to describe fluorescence recovery via nucleocytoplasmic transport. As osmolality increased, the diffusion coefficient of dextran decreased in the cytoplasm, but not the nucleus. Hyper-osmotic stress decreased nuclear size and increased nuclear lacunarity, indicating that while the nucleus was getting smaller, the pores and channels interdigitating the chromatin had expanded. The rate of nucleocytoplasmic transport was increased under hyper-osmotic stress but was insensitive to hypo-osmotic stress, consistent with the nonlinear osmotic properties of the nucleus. The mechanism of this osmotic sensitivity appears to be a change in the size and geometry of the nucleus, resulting in a shorter effective diffusion distance for the nucleus. These results may explain physical mechanisms by which osmotic stress can influence intracellular signaling pathways that rely on nucleocytoplasmic transport.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , Cytoplasm/metabolism , Active Transport, Cell Nucleus , Animals , Cells, Cultured , Chromatin/chemistry , Osmotic Pressure , Swine
18.
Ann N Y Acad Sci ; 1192: 404-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20392266

ABSTRACT

The critical discovery in the past two decades of the transient receptor potential (TRP) superfamily of ion channels has revealed the potential mechanisms by which cells sense diverse stimuli beyond the prototypical "five senses," identifying ion channels that are gated by heat, cold, mechanical loading, osmolarity, and other physical and chemical stimuli. TRP vanilloid 4 (TRPV4) is a Ca(2+)-permeable nonselective cation channel that appears to play a mechanosensory or osmosensory role in several musculoskeletal tissues. In articular cartilage, TRPV4 exhibits osmotic sensitivity, controlling cellular volume recovery, and other physiologic responses to osmotic stress. TRPV4 is expressed in both osteoblasts and osteoclasts, and the absence of TRPV4 prevents disuse-induced bone loss. TRPV4 activation promotes chondrogenesis by inducing SOX9 transcription, whereas a TRPV4 gain-of-function mutation leads to a developmental skeletal dysplasia, suggesting a critical role for TRPV4 in skeletal development. These studies provide mounting evidence for a regulatory role for the sensory channel TRPV4 in control of musculoskeletal tissues.


Subject(s)
Musculoskeletal System/metabolism , TRPV Cation Channels/physiology , Animals , Bone and Bones/metabolism , Bone and Bones/physiology , Humans , Mechanotransduction, Cellular/genetics , Mechanotransduction, Cellular/physiology , Osmosis/physiology , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/physiology
19.
Arthritis Rheum ; 60(10): 3028-37, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19790068

ABSTRACT

OBJECTIVE: Transient receptor potential vanilloid 4 (TRPV4) is a Ca(2+)-permeable channel that can be gated by tonicity (osmolarity) and mechanical stimuli. Chondrocytes, the cells in cartilage, respond to their osmotic and mechanical environments; however, the molecular basis of this signal transduction is not fully understood. This study was undertaken to demonstrate the presence and functionality of TRPV4 in chondrocytes. METHODS: TRPV4 protein expression was measured by immunolabeling and Western blotting. In response to TRPV4 agonist/antagonists, osmotic stress, and interleukin-1 (IL-1), changes in Ca(2+) signaling, cell volume, and prostaglandin E(2) (PGE(2)) production were measured in porcine chondrocytes using fluorescence microscopy, light microscopy, or immunoassay, respectively. RESULTS: TRPV4 was expressed abundantly at the RNA and protein levels. Exposure to 4alpha-phorbol 12,13-didecanoate (4alphaPDD), a TRPV4 activator, caused Ca(2+) signaling in chondrocytes, which was blocked by the selective TRPV4 antagonist, GSK205. Blocking TRPV4 diminished the chondrocytes' response to hypo-osmotic stress, reducing the fraction of Ca(2+) responsive cells, the regulatory volume decrease, and PGE(2) production. Ca(2+) signaling was inhibited by removal of extracellular Ca(2+) or depletion of intracellular stores. Specific activation of TRPV4 restored the defective regulatory volume decrease caused by IL-1. Chemical disruption of the primary cilium eliminated Ca(2+) signaling in response to either 4alphaPDD or hypo-osmotic stress. CONCLUSION: Our findings indicate that TRPV4 is present in articular chondrocytes, and chondrocyte response to hypo-osmotic stress is mediated by this channel, which involves both an extracellular Ca(2+) and intracellular Ca(2+) release. TRPV4 may also be involved in modulating the production or influence of proinflammatory molecules in response to osmotic stress.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Osmosis/physiology , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Cartilage, Articular/pathology , Cell Size , Cells, Cultured , Chondrocytes/pathology , Dinoprostone/metabolism , Interleukin-1/metabolism , Models, Animal , Phorbol Esters/pharmacology , Signal Transduction/physiology , Swine , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/drug effects
20.
J Biomech Eng ; 130(6): 061002, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19045531

ABSTRACT

Chondrocytes, the cells in articular cartilage, are enclosed within a pericellular matrix (PCM) whose composition and structure differ from those of the extracellular matrix (ECM). Since the PCM surrounds each cell, molecules that interact with the chondrocyte must pass through the pericellular environment. A quantitative understanding of the diffusional properties of the PCM may help in elucidating the regulatory role of the PCM in controlling transport to and from the chondrocyte. The diffusivities of fluorescently labeled 70 kDa and 500 kDa dextrans were quantified within the PCM of porcine articular cartilage using a newly developed mathematical model of scanning microphotolysis (SCAMP). SCAMP is a rapid line photobleaching method that accounts for out-of-plane bleaching attributable to high magnification. Data were analyzed by a best-fit comparison to simulations generated using a discretization of the diffusion-reaction equation in conjunction with the microscope-specific three-dimensional excitation and detection profiles. The diffusivity of the larger molecule (500 kDa dextran) was significantly lower than that of the smaller molecule (70 kDa dextran), and values were consistent with those reported previously using standard techniques. Furthermore, for both dextran sizes, the diffusion coefficient was significantly lower in the PCM than in the ECM; however, this difference was not detected in early-stage arthritic tissue. We have successfully modified the SCAMP technique to measure diffusion coefficients within the small volume of the PCM using confocal laser scanning microscopy. Our results support the hypothesis that diffusivity within the PCM of healthy articular cartilage is lower than that within the ECM, presumably due to differences in proteoglycan content.


Subject(s)
Cartilage, Articular/chemistry , Dextrans/chemistry , Extracellular Matrix/chemistry , Fluorescence Recovery After Photobleaching/methods , Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Models, Biological , Animals , Computer Simulation , Diffusion , Models, Chemical , Molecular Weight , Photolysis , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...