Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
J Bone Miner Res ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38887013

ABSTRACT

Knee osteoarthritis (OA), characterized by multiple joint tissue degenerations, remains a significant clinical challenge. Recent evidence suggests that crosstalk within the osteochondral unit may drive OA progression. While structural-biomechanical properties of bone and cartilage have been studied, potential interaction within the osteochondral unit in the context of OA has yet to be investigated. We performed comprehensive structural and biomechanical quantification of the cartilage, subchondral bone plate, and subchondral trabecular bone using 101 osteochondral cores collected from tibial plateaus of 12 control human cadavers (CT, 5 male/7 female) and 19 patients undergoing total knee replacement (OA, 6 male/13 female). For each sample, we quantified subchondral bone plate microstructure, plate-and-rod morphological properties of the subchondral trabecular bone using individual trabecula segmentation, and morphological and compositional properties of the articular cartilage. We also performed indentation testing on each compartment of the osteochondral unit to extract the respective structural-mechanical properties. Cartilage thickness was lower in moderate and severe OA regions, while OARSI score was higher only in severe OA regions. GAG content did not change in any OA region. Aggregate and shear moduli were lower only in severe OA regions, while permeability was lower only in moderate OA regions. In the subchondral bone plate, thickness and TMD were higher in moderate and severe OA regions. Tissue modulus of subchondral trabecular bone was lower in moderate OA regions despite a thicker and more mineralized subchondral bone plate; this deterioration was not observed in severe OA regions. Regression analysis revealed strong correlations between cartilage and subchondral trabecular bone properties in CT; these correlations were also found in moderate OA regions but were not observed in severe OA regions. In summary, our findings comprehensively characterize the human OA osteochondral unit. Importantly, uncoupling cartilage and subchondral bone structural-mechanical properties may be a hallmark of OA.


Knee osteoarthritis (OA) is a complex condition involving the degradation of joint tissues. To better understand OA progression, we investigated the interplay between different components of the joint. Our study focused on how cartilage, subchondral bone plate, and subchondral trabecular bone interact in human knee OA samples. We observed distinct changes in these tissues in moderate and severe OA regions compared to healthy joints. In moderate to severe OA, we found that cartilage thickness decreased while the subchondral bone plate thickened. Interestingly, the strength of the subchondral trabecular bone decreased only in moderate OA regions, not in severe OA. Moreover, our analysis revealed strong correlations between cartilage and subchondral trabecular bone properties in healthy joints and moderate OA regions. However, these correlations were absent in severe OA regions, indicating a disruption in the usual relationship between these tissues. Overall, our findings shed light on the structural and biomechanical changes occurring within the knee joint in OA. Understanding these changes may offer insights into potential therapeutic strategies for managing OA.

2.
Front Bioeng Biotechnol ; 11: 1225495, 2023.
Article in English | MEDLINE | ID: mdl-37711443

ABSTRACT

Electric fields find use in tissue engineering but also in sensor applications besides the broad classical application range. Accurate numerical models of electrical stimulation devices can pave the way for effective therapies in cartilage regeneration. To this end, the dielectric properties of the electrically stimulated tissue have to be known. However, knowledge of the dielectric properties is scarce. Electric field-based methods such as impedance spectroscopy enable determining the dielectric properties of tissue samples. To develop a detailed understanding of the interaction of the employed electric fields and the tissue, fine-grained numerical models based on tissue-specific 3D geometries are considered. A crucial ingredient in this approach is the automated generation of numerical models from biomedical images. In this work, we explore classical and artificial intelligence methods for volumetric image segmentation to generate model geometries. We find that deep learning, in particular the StarDist algorithm, permits fast and automatic model geometry and discretisation generation once a sufficient amount of training data is available. Our results suggest that already a small number of 3D images (23 images) is sufficient to achieve 80% accuracy on the test data. The proposed method enables the creation of high-quality meshes without the need for computer-aided design geometry post-processing. Particularly, the computational time for the geometrical model creation was reduced by half. Uncertainty quantification as well as a direct comparison between the deep learning and the classical approach reveal that the numerical results mainly depend on the cell volume. This result motivates further research into impedance sensors for tissue characterisation. The presented approach can significantly improve the accuracy and computational speed of image-based models of electrical stimulation for tissue engineering applications.

3.
ACS Biomater Sci Eng ; 8(6): 2564-2573, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35561285

ABSTRACT

During traumatic joint injuries, impact overloading can cause mechanical damage to the cartilage. In the following inflammation phase, excessive inflammatory cytokines (e.g., interleukin-1ß (IL-1ß)) can act on chondrocytes, causing over-proliferation, apoptosis, and extracellular matrix (ECM) degradation that can lead to osteoarthritis. This study investigated the combined effects of traumatic overloading and IL-1ß challenge on the metabolic activities of chondrocytes. Bovine cartilage explants underwent impact overloading followed by IL-1ß exposure at a physiologically relevant dosage (1 ng/mL). New click chemistry-based methods were developed to visualize and quantify the proliferation of in situ chondrocytes in a nondestructive manner without the involvement of histological sectioning or antibodies. Click chemistry-based methods were also employed to measure the ECM synthesis and degradation in cartilage explants. As the click reactions are copper-free and bio-orthogonal, i.e., with negligible cellular toxicity, cartilage ECM was cultured and studied for 6 weeks. Traumatic overloading induced significant cell death, mainly in the superficial zone. The high number of dead cells reduced the overall proliferation of chondrocytes as well as the synthesis of glycosaminoglycan (GAG) and collagen contents, but overloading alone had no effects on ECM degradation. IL-1ß challenge had little effect on cell viability, proliferation, or protein synthesis but induced over 40% GAG loss in 10 days and 61% collagen loss in 6 weeks. For the overloaded samples, IL-1ß induced greater degrees of degradation, with 68% GAG loss in 10 days and 80% collagen loss in 6 weeks. The results imply a necessary immediate ease of inflammation after joint injuries when trauma damage on cartilage is present. The new click chemistry methods could benefit many cellular and tissue engineering studies, providing convenient and sensitive assays of metabolic activities of cells in native three-dimensional (3D) environments.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cattle , Chondrocytes/metabolism , Chondrocytes/pathology , Click Chemistry , Inflammation/metabolism , Inflammation/pathology , Osteoarthritis/metabolism , Osteoarthritis/pathology
4.
ACS Nano ; 16(1): 1220-1230, 2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35015500

ABSTRACT

Molecular engineering of biological tissues using synthetic mimics of native matrix molecules can modulate the mechanical properties of the cellular microenvironment through physical interactions with existing matrix molecules, and in turn, mediate the corresponding cell mechanobiology. In articular cartilage, the pericellular matrix (PCM) is the immediate microniche that regulates cell fate, signaling, and metabolism. The negatively charged osmo-environment, as endowed by PCM proteoglycans, is a key biophysical cue for cell mechanosensing. This study demonstrated that biomimetic proteoglycans (BPGs), which mimic the ultrastructure and polyanionic nature of native proteoglycans, can be used to molecularly engineer PCM micromechanics and cell mechanotransduction in cartilage. Upon infiltration into bovine cartilage explant, we showed that localization of BPGs in the PCM leads to increased PCM micromodulus and enhanced chondrocyte intracellular calcium signaling. Applying molecular force spectroscopy, we revealed that BPGs integrate with native PCM through augmenting the molecular adhesion of aggrecan, the major PCM proteoglycan, at the nanoscale. These interactions are enabled by the biomimetic "bottle-brush" ultrastructure of BPGs and facilitate the integration of BPGs within the PCM. Thus, this class of biomimetic molecules can be used for modulating molecular interactions of pericellular proteoglycans and harnessing cell mechanosensing. Because the PCM is a prevalent feature of various cell types, BPGs hold promising potential for improving regeneration and disease modification for not only cartilage-related healthcare but many other tissues and diseases.


Subject(s)
Cartilage, Articular , Proteoglycans , Cattle , Animals , Biomimetics , Extracellular Matrix/metabolism , Mechanotransduction, Cellular , Chondrocytes/metabolism , Biophysics
5.
Matrix Biol ; 102: 1-19, 2021 08.
Article in English | MEDLINE | ID: mdl-34314838

ABSTRACT

This study queried the role of type V collagen in the post-natal growth of temporomandibular joint (TMJ) condylar cartilage, a hybrid tissue with a fibrocartilage layer covering a secondary hyaline cartilage layer. Integrating outcomes from histology, immunofluorescence imaging, electron microscopy and atomic force microscopy-based nanomechanical tests, we elucidated the impact of type V collagen reduction on TMJ condylar cartilage growth in the type V collagen haploinsufficiency and inducible knockout mice. Reduction of type V collagen led to significantly thickened collagen fibrils, decreased tissue modulus, reduced cell density and aberrant cell clustering in both the fibrous and hyaline layers. Post-natal growth of condylar cartilage involves the chondrogenesis of progenitor cells residing in the fibrous layer, which gives rise to the secondary hyaline layer. Loss of type V collagen resulted in reduced proliferation of these cells, suggesting a possible role of type V collagen in mediating the progenitor cell niche. When the knockout of type V collagen was induced in post-weaning mice after the start of physiologic TMJ loading, the hyaline layer exhibited pronounced thinning, supporting an interplay between type V collagen and occlusal loading in condylar cartilage growth. The phenotype in hyaline layer can thus be attributed to the impact of type V collagen on the mechanically regulated progenitor cell activities. In contrast, knee cartilage does not contain the progenitor cell population at post-natal stages, and develops normal structure and biomechanical properties with the loss of type V collagen. Therefore, in the TMJ, in addition to its established role in regulating the assembly of collagen I fibrils, type V collagen also impacts the mechanoregulation of progenitor cell activities in the fibrous layer. We expect such knowledge to establish a foundation for understanding condylar cartilage matrix development and regeneration, and to yield new insights into the TMJ symptoms in patients with classic Ehlers-Danlos syndrome, a genetic disease due to autosomal mutation of type V collagen.


Subject(s)
Cartilage, Articular , Collagen Type V , Animals , Biomechanical Phenomena , Cartilage , Humans , Hyalin , Mandibular Condyle , Mice , Temporomandibular Joint
6.
Genes (Basel) ; 13(1)2021 12 28.
Article in English | MEDLINE | ID: mdl-35052411

ABSTRACT

The proteoglycan-containing pericellular matrix (PCM) controls both the biophysical and biochemical microenvironment of osteocytes, which are the most abundant cells embedded and dispersed in bones. As a molecular sieve, osteocytic PCMs not only regulate mass transport to and from osteocytes but also act as sensors of external mechanical environments. The turnover of osteocytic PCM remains largely unknown due to technical challenges. Here, we report a novel imaging technique based on metabolic labeling and "click-chemistry," which labels de novo PCM as "halos" surrounding osteocytes in vitro and in vivo. We then tested the method and showed different labeling patterns in young vs. old bones. Further "pulse-chase" experiments revealed dramatic difference in the "half-life" of PCM of cultured osteocytes (~70 h) and that of osteocytes in vivo (~75 d). When mice were subjected to either 3-week hindlimb unloading or 7-week tibial loading (5.1 N, 4 Hz, 3 d/week), PCM half-life was shortened (~20 d) and degradation accelerated. Matrix metallopeptidase MMP-14 was elevated in mechanically loaded osteocytes, which may contribute to PCM degradation. This study provides a detailed procedure that enables semi-quantitative study of the osteocytic PCM remodeling in vivo and in vitro.


Subject(s)
Aging , Bone and Bones/metabolism , Extracellular Matrix/metabolism , Hindlimb Suspension/methods , Matrix Metalloproteinase 14/metabolism , Mechanotransduction, Cellular , Osteocytes/metabolism , Animals , Bone and Bones/cytology , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Osteocytes/cytology
7.
Matrix Biol ; 96: 1-17, 2021 02.
Article in English | MEDLINE | ID: mdl-33246102

ABSTRACT

In cartilage tissue engineering, one key challenge is for regenerative tissue to recapitulate the biomechanical functions of native cartilage while maintaining normal mechanosensitive activities of chondrocytes. Thus, it is imperative to discern the micromechanobiological functions of the pericellular matrix, the ~ 2-4 µm-thick domain that is in immediate contact with chondrocytes. In this study, we discovered that decorin, a small leucine-rich proteoglycan, is a key determinant of cartilage pericellular matrix micromechanics and chondrocyte mechanotransduction in vivo. The pericellular matrix of decorin-null murine cartilage developed reduced content of aggrecan, the major chondroitin sulfate proteoglycan of cartilage and a mild increase in collagen II fibril diameter vis-à-vis wild-type controls. As a result, decorin-null pericellular matrix showed a significant reduction in micromodulus, which became progressively more pronounced with maturation. In alignment with the defects of pericellular matrix, decorin-null chondrocytes exhibited decreased intracellular calcium activities, [Ca2+]i, in both physiologic and osmotically evoked fluidic environments in situ, illustrating impaired chondrocyte mechanotransduction. Next, we compared [Ca2+]i activities of wild-type and decorin-null chondrocytes following enzymatic removal of chondroitin sulfate glycosaminoglycans. The results showed that decorin mediates chondrocyte mechanotransduction primarily through regulating the integrity of aggrecan network, and thus, aggrecan-endowed negative charge microenvironment in the pericellular matrix. Collectively, our results provide robust genetic and biomechanical evidence that decorin is an essential constituent of the native cartilage matrix, and suggest that modulating decorin activities could improve cartilage regeneration.


Subject(s)
Cartilage, Articular/physiology , Decorin/genetics , Extracellular Matrix/metabolism , Loss of Function Mutation , Aggrecans/metabolism , Animals , Biomechanical Phenomena , Calcium Signaling , Cartilage, Articular/metabolism , Female , Male , Mechanotransduction, Cellular , Mice , Regeneration
8.
Acta Biomater ; 111: 267-278, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32428685

ABSTRACT

The pericellular matrix (PCM) of cartilage is a structurally distinctive microdomain surrounding each chondrocyte, and is pivotal to cell homeostasis and cell-matrix interactions in healthy tissue. This study queried if the PCM is the initiation point for disease or a casualty of more widespread matrix degeneration. To address this question, we queried the mechanical properties of the PCM and chondrocyte mechanoresponsivity with the development of post-traumatic osteoarthritis (PTOA). To do so, we integrated Kawamoto's film-assisted cryo-sectioning with immunofluorescence-guided AFM nanomechanical mapping, and quantified the microscale modulus of murine cartilage PCM and further-removed extracellular matrix. Using the destabilization of the medial meniscus (DMM) murine model of PTOA, we show that decreases in PCM micromechanics are apparent as early as 3 days after injury, and that this precedes changes in the bulk ECM properties and overt indications of cartilage damage. We also show that, as a consequence of altered PCM properties, calcium mobilization by chondrocytes in response to mechanical challenge (hypo-osmotic stress) is significantly disrupted. These aberrant changes in chondrocyte micromechanobiology as a consequence of DMM could be partially blocked by early inhibition of PCM remodeling. Collectively, these results suggest that changes in PCM micromechanobiology are leading indicators of the initiation of PTOA, and that disease originates in the cartilage PCM. This insight will direct the development of early detection methods, as well as small molecule-based therapies that can stop early aberrant remodeling in this critical cartilage microdomain to slow or reverse disease progression. STATEMENT OF SIGNIFICANCE: Post-traumatic osteoarthritis (PTOA) is one prevalent musculoskeletal disease that afflicts young adults, and there are no effective strategies for early detection or intervention. This study identifies that the reduction of cartilage pericellular matrix (PCM) micromodulus is one of the earliest events in the initiation of PTOA, which, in turn, impairs the mechanosensitive activities of chondrocytes, contributing to the vicious loop of cartilage degeneration. Rescuing the integrity of PCM has the potential to restore normal chondrocyte mechanosensitive homeostasis and to prevent further degradation of cartilage. Our findings enable the development of early OA detection methods targeting changes in the PCM, and treatment strategies that can stop early aberrant remodeling in this critical microdomain to slow or reverse disease progression.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Chondrocytes , Extracellular Matrix , Menisci, Tibial , Mice
9.
Biomolecules ; 10(2)2020 01 29.
Article in English | MEDLINE | ID: mdl-32013135

ABSTRACT

Perlecan/Hspg2, a large monomeric heparan sulfate proteoglycan, is found in the basement membrane and extracellular matrix, where it acts as a matrix scaffold, growth factor depot, and tissue barrier. Perlecan deficiency leads to skeletal dysplasia in Schwartz-Jampel Syndrome (SJS) and is a risk factor for osteoporosis. In the SJS-mimicking murine model (Hypo), inferior cortical bone quality and impaired mechanotransduction in osteocytes were reported. This study focused on trabecular bone, where perlecan deficiency was hypothesized to result in structural deficit and altered response to disuse and re-loading. We compared the Hypo versus WT trabecular bone in both axial and appendicular skeletons of 8-38-week-old male mice, and observed severe trabecular deficit in Hypo mice, approximately 50% reduction of Tb.BV/TV regardless of skeletal site and animal age. Defects in endochondral ossification (e.g., accelerated mineralization), increases in osteoclast activity, and altered differentiation of bone progenitor cells in marrow contributed to the Hypo phenotype. The Hypo trabecular bone deteriorated further under three-week hindlimb suspension as did the WT. Re-ambulation partially recovered the lost trabecular bone in Hypo, but not in WT mice. The novel finding that low-impact loading could counter detrimental disuse effects in the perlecan-deficient skeleton suggests a strategy to maintain skeletal health in SJS patients.


Subject(s)
Cancellous Bone/pathology , Heparan Sulfate Proteoglycans/deficiency , Heparan Sulfate Proteoglycans/genetics , Osteocytes/cytology , Animals , Femur/pathology , Hematopoietic Stem Cells/cytology , Heparan Sulfate Proteoglycans/physiology , Kyphosis , Male , Mechanotransduction, Cellular , Metabolism , Mice , Mice, Inbred C57BL , Osteoclasts/cytology , Osteogenesis , Phenotype , Risk Factors , Stress, Mechanical , Walking , X-Ray Microtomography
10.
J Mech Behav Biomed Mater ; 102: 103504, 2020 02.
Article in English | MEDLINE | ID: mdl-31670258

ABSTRACT

In this paper, the interstitial fluid flow in skeletal muscle endomysium was examined using an in-situ indentation testing in combination with theoretical modelling. The objective was to understand the transport properties of the three-dimensional and highly hierarchical muscular interstitial matrices, which play important roles in muscle-bone cross-talk and signaling during musculoskeletal development and maintenance. Gastrocnemius muscles from four 3-month old calves were harvested and subjected to a creeping test using a custom-designed device. The experiments, in combination with an anatomy-based theoretical model, were used to capture the spatial-temporal response of the skeletal muscle to external impacts. For the first time, the detailed load-induced interstitial fluid pressurization in the muscle endomyseal space was obtained. The relative contribution from the solid muscle fibers and the interstitial fluid to the temporal loading response was captured. The paper presented herein provides important information regarding the mechanical environment within the muscle tissue, which could help the future study of muscle's response to forces and its subsequent signaling to surrounding tissues in vivo.


Subject(s)
Extracellular Fluid , Muscle Fibers, Skeletal , Animals , Bone and Bones , Cattle , Muscle, Skeletal , Physical Phenomena
11.
Bone ; 131: 115078, 2020 02.
Article in English | MEDLINE | ID: mdl-31715337

ABSTRACT

Perlecan, a heparan sulfate proteoglycan, acts as a mechanical sensor for bone to detect external loading. Deficiency of perlecan increases the risk of osteoporosis in patients with Schwartz-Jampel Syndrome (SJS) and attenuates loading-induced bone formation in perlecan deficient mice (Hypo). Considering that intracellular calcium [Ca2+]i is an ubiquitous messenger controlling numerous cellular processes including mechanotransduction, we hypothesized that perlecan deficiency impairs bone's calcium signaling in response to loading. To test this, we performed real-time [Ca2+]i imaging on in situ osteocytes of adult murine tibiae under cyclic loading (8N). Relative to wild type (WT), Hypo osteocytes showed decreases in the overall [Ca2+]i response rate (-58%), calcium peaks (-33%), cells with multiple peaks (-53%), peak magnitude (-6.8%), and recovery speed to baseline (-23%). RNA sequencing and pathway analysis of tibiae from mice subjected to one or seven days of unilateral loading demonstrated that perlecan deficiency significantly suppressed the calcium signaling, ECM-receptor interaction, and focal adhesion pathways following repetitive loading. Defects in the endoplasmic reticulum (ER) calcium cycling regulators such as Ryr1/ryanodine receptors and Atp2a1/Serca1 calcium pumps were identified in Hypo bones. Taken together, impaired calcium signaling may contribute to bone's reduced anabolic response to loading, underlying the osteoporosis risk for the SJS patients.


Subject(s)
Calcium Signaling , Heparan Sulfate Proteoglycans , Animals , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Humans , Mechanotransduction, Cellular , Mice , Transcriptome/genetics
12.
ACS Nano ; 13(10): 11320-11333, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31550133

ABSTRACT

Joint biomechanical functions rely on the integrity of cartilage extracellular matrix. Understanding the molecular activities that govern cartilage matrix assembly is critical for developing effective cartilage regeneration strategies. This study elucidated the role of decorin, a small leucine-rich proteoglycan, in the structure and biomechanical functions of cartilage. In decorin-null cartilage, we discovered a substantial reduction of aggrecan content, the major proteoglycan of cartilage matrix, and mild changes in collagen fibril nanostructure. This loss of aggrecan resulted in significantly impaired biomechanical properties of cartilage, including decreased modulus, elevated hydraulic permeability, and reduced energy dissipation capabilities. At the cellular level, we found that decorin functions to increase the retention of aggrecan in the neo-matrix of chondrocytes, rather than to directly influence the biosynthesis of aggrecan. At the molecular level, we demonstrated that decorin significantly increases the adhesion between aggrecan and aggrecan molecules and between aggrecan molecules and collagen II fibrils. We hypothesize that decorin plays a crucial structural role in mediating the matrix integrity and biomechanical functions of cartilage by providing physical linkages to increase the adhesion and assembly of aggrecan molecules at the nanoscale.


Subject(s)
Aggrecans/chemistry , Decorin/chemistry , Extracellular Matrix/chemistry , Cartilage, Articular/chemistry , Nanostructures/chemistry , Proteoglycans/chemistry
13.
Sci Rep ; 9(1): 93, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30643177

ABSTRACT

Traumatic joint injuries often result in elevated proinflammatory cytokine (such as IL-1ß) levels in the joint cavity, which can increase the catabolic activities of chondrocytes and damage cartilage. This study investigated the early genetic responses of healthy in situ chondrocytes under IL-1ß attack with a focus on cell cycle and calcium signaling pathways. RNA sequencing analysis identified 2,232 significantly changed genes by IL-1ß, with 1,259 upregulated and 973 downregulated genes. Catabolic genes related to ECM degeneration were promoted by IL-1ß, consistent with our observations of matrix protein loss and mechanical property decrease during 24-day in vitro culture of cartilage explants. IL-1ß altered the cell cycle (108 genes) and Rho GTPases signaling (72 genes) in chondrocytes, while chondrocyte phenotypic shift was observed with histology, cell volume measurement, and MTT assay. IL-1ß inhibited the spontaneous calcium signaling in chondrocytes, a fundamental signaling event in chondrocyte metabolic activities. The expression of 24 genes from 6 calcium-signaling related pathways were changed by IL-1ß exposure. This study provided a comprehensive list of differentially expressed genes of healthy in situ chondrocytes in response to IL-1ß attack, which represents a useful reference to verify and guide future cartilage studies related to the acute inflammation after joint trauma.


Subject(s)
Calcium Signaling/drug effects , Cell Cycle , Chondrocytes/drug effects , Interleukin-1beta/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Animals , Cattle , Cells, Cultured , Gene Expression Profiling , Sequence Analysis, RNA
14.
FASEB J ; 33(4): 4675-4687, 2019 04.
Article in English | MEDLINE | ID: mdl-30601690

ABSTRACT

Intracellular calcium ([Ca2+]i) oscillation is a fundamental signaling response of cartilage cells under mechanical loading or osmotic stress. Chondrocytes are usually considered as nonexcitable cells with no spontaneous [Ca2+]i signaling. This study proved that chondrocytes can exhibit robust spontaneous [Ca2+]i signaling without explicit external stimuli. The intensity of [Ca2+]i peaks from individual chondrocytes maintain a consistent spatiotemporal pattern, acting as a unique "fingerprint" for each cell. Statistical analysis revealed lognormal distributions of the temporal parameters of [Ca2+]i peaks, as well as strong linear correlations between their means and sds. Based on these statistical findings, we hypothesized that the spontaneous [Ca2+]i peaks may result from an autocatalytic process and that [Ca2+]i oscillation is controlled by a threshold-regulating mechanism. To test these 2 mechanisms, we established a multistage biophysical model by assuming the spontaneous [Ca2+]i signaling of chondrocytes as a combination of deterministic and stochastic processes. The theoretical model successfully explained the lognormal distribution of the temporal parameters and the fingerprint feature of [Ca2+]i peaks. In addition, by using antagonists for 10 pathways, we revealed that the initiation of spontaneous [Ca2+]i peaks in chondrocytes requires the presence of extracellular Ca2+, and that the PLC-inositol 1,4,5-trisphosphate pathway, which controls the release of calcium from the endoplasmic reticulum, can affect the initiation of spontaneous [Ca2+]i peaks in chondrocytes. The purinoceptors and transient receptor potential vanilloid 4 channels on the plasma membrane also play key roles in the spontaneous [Ca2+]i signaling of chondrocytes. In contrast, blocking the T-type or L-type voltage-gated calcium channel promoted the spontaneous calcium signaling. This study represents a systematic effort to understand the features and initiation mechanisms of spontaneous [Ca2+]i signaling in chondrocytes, which are critical for chondrocyte mechanobiology.-Zhou, Y., Lv, M., Li, T., Zhang, T., Duncan, R., Wang, L., Lu, X. L. Spontaneous calcium signaling of cartilage cells: from spatiotemporal features to biophysical modeling.


Subject(s)
Calcium Signaling/physiology , Cartilage, Articular/metabolism , Animals , Calcium/metabolism , Cattle , Cell Membrane/metabolism , Chondrocytes/metabolism , Endoplasmic Reticulum/metabolism , Osmotic Pressure/physiology , Spatio-Temporal Analysis
15.
J Orthop Res ; 36(2): 730-738, 2018 02.
Article in English | MEDLINE | ID: mdl-28980722

ABSTRACT

Mechanical loading on articular cartilage can induce many physical and chemical stimuli on chondrocytes residing in the extracellular matrix (ECM). Intracellular calcium ([Ca2+ ]i ) signaling is among the earliest responses of chondrocytes to physical stimuli, but the [Ca2+ ]i signaling of in situ chondrocytes in loaded cartilage is not fully understood due to the technical challenges in [Ca2+ ]i imaging of chondrocytes in a deforming ECM. This study developed a novel bi-directional microscopy loading device that enables the record of transient [Ca2+ ]i responses of in situ chondrocytes in loaded cartilage. It was found that compressive loading significantly promoted [Ca2+ ]i signaling in chondrocytes with faster [Ca2+ ]i oscillations in comparison to the non-loaded cartilage. Seven [Ca2+ ]i signaling pathways were further investigated by treating the cartilage with antagonists prior to and/or during the loading. Removal of extracellular Ca2+ ions completely abolished the [Ca2+ ]i responses of in situ chondrocytes, suggesting the indispensable role of extracellular Ca2+ sources in initiating the [Ca2+ ]i signaling in chondrocytes. Depletion of intracellular Ca2+ stores, inhibition of PLC-IP3 pathway, and block of purinergic receptors on plasma membrane led to significant reduction in the responsive rate of cells. Three types of ion channels that are regulated by different physical signals, TRPV4 (osmotic and mechanical stress), T-type VGCCs (electrical potential), and mechanical sensitive ion channels (mechanical loading) all demonstrated critical roles in controlling the [Ca2+ ]i responses of in situ chondrocyte in the loaded cartilage. This study provided new knowledge about the [Ca2+ ]i signaling and mechanobiology of chondrocytes in its natural residing environment. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:730-738, 2018.


Subject(s)
Calcium Signaling , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Animals , Cartilage, Articular/cytology , Cattle , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Female , Ion Channels/metabolism , Male , Weight-Bearing
16.
J Biomech ; 60: 134-141, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28688538

ABSTRACT

This study aims to quantify the biomechanical properties of murine temporomandibular joint (TMJ) articular disc and condyle cartilage using AFM-nanoindentation. For skeletally mature, 3-month old mice, the surface of condyle cartilage was found to be significantly stiffer (306±84kPa, mean±95% CI) than those of the superior (85±23kPa) and inferior (45±12kPa) sides of the articular disc. On the disc surface, significant heterogeneity was also detected across multiple anatomical sites, with the posterior end being the stiffest and central region being the softest. Using SEM, this study also found that the surfaces of disc are composed of anteroposteriorly oriented collagen fibers, which are sporadically covered by thinner random fibrils. Such fibrous nature results in both an F-D3/2 indentation response, which is a typical Hertzian response for soft continuum tissue under a spherical tip, and a linear F-D response, which is typical for fibrous tissues, further signifying the high degree of tissue heterogeneity. In comparison, the surface of condyle cartilage is dominated by thinner, randomly oriented collagen fibrils, leading to Hertzian-dominated indentation responses. As the first biomechanical study of murine TMJ, this work will provide a basis for future investigations of TMJ tissue development and osteoarthritis in various murine TMJ models.


Subject(s)
Cartilage, Articular/physiology , Mandibular Condyle/physiology , Temporomandibular Joint/physiology , Animals , Fibrillar Collagens/physiology , Mice, Inbred C57BL , Microscopy, Atomic Force , Osteoarthritis/physiopathology
17.
Proc Natl Acad Sci U S A ; 113(50): 14360-14365, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911782

ABSTRACT

Osteoarthritis (OA) is the most common joint disease, characterized by progressive destruction of the articular cartilage. The surface of joint cartilage is the first defensive and affected site of OA, but our knowledge of genesis and homeostasis of this superficial zone is scarce. EGFR signaling is important for tissue homeostasis. Immunostaining revealed that its activity is mostly dominant in the superficial layer of healthy cartilage but greatly diminished when OA initiates. To evaluate the role of EGFR signaling in the articular cartilage, we studied a cartilage-specific Egfr-deficient (CKO) mouse model (Col2-Cre EgfrWa5/flox). These mice developed early cartilage degeneration at 6 mo of age. By 2 mo of age, although their gross cartilage morphology appears normal, CKO mice had a drastically reduced number of superficial chondrocytes and decreased lubricant secretion at the surface. Using superficial chondrocyte and cartilage explant cultures, we demonstrated that EGFR signaling is critical for maintaining the number and properties of superficial chondrocytes, promoting chondrogenic proteoglycan 4 (Prg4) expression, and stimulating the lubrication function of the cartilage surface. In addition, EGFR deficiency greatly disorganized collagen fibrils in articular cartilage and strikingly reduced cartilage surface modulus. After surgical induction of OA at 3 mo of age, CKO mice quickly developed the most severe OA phenotype, including a complete loss of cartilage, extremely high surface modulus, subchondral bone plate thickening, and elevated joint pain. Taken together, our studies establish EGFR signaling as an important regulator of the superficial layer during articular cartilage development and OA initiation.


Subject(s)
Arthritis, Experimental/metabolism , Cartilage, Articular/metabolism , ErbB Receptors/metabolism , Osteoarthritis/metabolism , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/prevention & control , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrogenesis , ErbB Receptors/deficiency , ErbB Receptors/genetics , Humans , Male , Mice , Mice, Knockout , Osteoarthritis/pathology , Osteoarthritis/prevention & control , Proteoglycans/metabolism , Signal Transduction
18.
Med Biol Eng Comput ; 54(12): 1893-1902, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27056409

ABSTRACT

Tyramine-substituted hyaluronic acid (HA-Tyr) hydrogels formed by the oxidative coupling reaction of hydrogen peroxide (H2O2) and horseradish peroxidase (HRP) have been used for cellular encapsulation and protein delivery. Crosslinking density and gelation time can be tuned by altering the H2O2 and HRP concentrations. Previous studies using HA-Tyr constructs report significant mechanical degradation after 21 days of culture. In this work, exogenous supplementation of HRP after initial gelation resulted in superior mechanical properties in acellular hydrogels and improved viability and proliferation in cell-laden constructs. Swelling of the acellular hydrogels was prevented in the samples receiving exogenous HRP. Monolayer studies showed no negative effect of relevant HRP concentrations on the viability of human adipose-derived stem cells (hASCs) and improved the viability of hASCs cultured with HRP and H2O2 compared to H2O2 alone. Taken together, this study demonstrates that HA-Tyr hydrogel properties could be modified by exogenous supplementation of HRP to tune scaffold degradation and improve cell viability by mitigating the negative effects of oxidative stress.


Subject(s)
Hyaluronic Acid/pharmacology , Hydrogels/pharmacology , Mechanical Phenomena , Stem Cells/cytology , Tyramine/pharmacology , Adipose Tissue/cytology , Cell Proliferation/drug effects , Cell Survival/drug effects , Elastic Modulus , Horseradish Peroxidase/metabolism , Humans , Hydrogen Peroxide/toxicity , Mechanical Phenomena/drug effects , Stem Cells/drug effects
19.
Soft Matter ; 12(4): 1158-69, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26599600

ABSTRACT

This study investigates the roles of two distinct features of ionically cross-linked polyelectrolyte networks - ionic cross-links and fixed charges - in determining their nanomechanical properties. The layer-by-layer assembled poly(allylamine hydrochloride)/poly(acrylic acid) (PAH/PAA) network is used as the model material. The densities of ionic cross-links and fixed charges are modulated through solution pH and ionic strength (IS), and the swelling ratio, elastic and viscoelastic properties are quantified via an array of atomic force microscopy (AFM)-based nanomechanical tools. The roles of ionic cross-links are underscored by the distinctive elastic and viscoelastic nanomechanical characters observed here. First, as ionic cross-links are highly sensitive to solution conditions, the instantaneous modulus, E0, exhibits orders-of-magnitude changes upon pH- and IS-governed swelling, distinctive from the rubber elasticity prediction based on permanent covalent cross-links. Second, ionic cross-links can break and self-re-form, and this mechanism dominates force relaxation of PAH/PAA under a constant indentation depth. In most states, the degree of relaxation is >90%, independent of ionic cross-link density. The importance of fixed charges is highlighted by the unexpectedly more elastic nature of the network despite low ionic cross-link density at pH 2.0, IS 0.01 M. Here, the complex is a net charged, loosely cross-linked, where the degree of relaxation is attenuated to ≈50% due to increased elastic contribution arising from fixed charge-induced Donnan osmotic pressure. In addition, this study develops a new method for quantifying the thickness of highly swollen polymer hydrogel films. It also underscores important technical considerations when performing nanomechanical tests on highly rate-dependent polymer hydrogel networks. These results provide new insights into the nanomechanical characters of ionic polyelectrolyte complexes, and lay the ground for further investigation of their unique time-dependent properties.


Subject(s)
Acrylates/chemistry , Allylamine/analogs & derivatives , Elasticity , Nanostructures/chemistry , Static Electricity , Allylamine/chemistry , Osmolar Concentration , Viscosity
20.
Ann Biomed Eng ; 44(4): 1138-47, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26219403

ABSTRACT

Calcium is a universal second messenger that mediates the metabolic activity of chondrocytes in articular cartilage. Spontaneous intracellular calcium ([Ca(2+)]i) oscillations, similar to those in neurons and myocytes, have recently been observed in chondrocytes. This study analyzed and compared the effects of different osmotic environments (hypertonic, hypotonic, and isotonic) on the spontaneous [Ca(2+)]i signaling of in situ chondrocytes residing in juvenile and adult cartilage explants. In spite of a lower cell density, a significantly higher percentage of chondrocytes in adult cartilage under all osmotic environments demonstrated spontaneous [Ca(2+)]i oscillations than chondrocytes in juvenile cartilage. For both juvenile and adult chondrocytes, hypotonic stress increased while hypertonic stress decreased the response rates. Furthermore, the spatiotemporal characteristics of the [Ca(2+)]i peaks vary in an age-dependent manner. In the hypotonic environment, the [Ca(2+)]i oscillation frequency of responsive adult cells is almost tripled whereas the juvenile cells respond with an increased duration and magnitude of each [Ca(2+)]i peak. Both juvenile and adult chondrocytes demonstrated significantly slower [Ca(2+)]i oscillations with longer rising and recovery time under the hypertonic condition. Taken together, these results shed new insights into the interplay between age and osmotic environment that may regulate the fundamental metabolism of chondrocytes.


Subject(s)
Calcium Signaling , Chondrocytes/metabolism , Osmolar Concentration , Age Factors , Animals , Cartilage, Articular/cytology , Cattle
SELECTION OF CITATIONS
SEARCH DETAIL
...