Skip to main content
APL Bioengineering logoLink to APL Bioengineering
. 2022 Jan 5;6(1):011501. doi: 10.1063/5.0068277

The role of mechanobiology in bone and cartilage model systems in characterizing initiation and progression of osteoarthritis

Tom Hodgkinson 1, Isabel N Amado 1, Fergal J O'Brien 1,2,3,1,2,3,1,2,3,a), Oran D Kennedy 1,2,3,1,2,3,1,2,3
PMCID: PMC12313331  PMID: 40746901

Abstract

Multifaceted changes in the mechanobiological environment of skeletal joints, at multiple length scales, are central to the development of diseases-like osteoarthritis (OA). Recent evidence demonstrates related mechanical alterations in both bone and cartilage tissues, with crosstalk between the tissues being an important factor in acute and chronic degenerative processes. However, recapitulating multicellular tissue systems in the laboratory to study the entire osteochondral unit remains challenging. Thus, the development of accurate and reproducible OA model systems and the selection of the most suitable model for individual experimental approaches are critical. This review first discusses recent progress in understanding mechanosensory processes in healthy and osteoarthritic joints. Subsequently, we review advancements in the development of in vitro and ex vivo model systems ranging from 2D monocultures through to joint organ-on-a-chip models. Use of these systems allows for the study of multiple cell types in controlled, reproducible, and dynamic environments, which can incorporate precisely controlled mechanical and biochemical stimuli, and biophysical cues. The way in which these models have, and will continue to, improve our ability to recapitulate complex mechanical/paracrine signaling pathways in osteochondral tissues is then discussed. As the accuracy of model systems advances, they will have a significant impact on both our understanding of the pathobiology of OA and in identifying and screening therapeutic targets to improve treatment of this complex disease.

INTRODUCTION

The healthy functioning of skeletal joints is often considered in terms of articular cartilage only. However, in addition to cartilage, the subchondral bone, synovial lining, ligaments, and other connective tissues form an integrated system that maintains form and function. Each of these tissues contributes to overall functionality and damage to any of them can impact joint integrity. Considering the joint at this level is crucial to understanding its function as well as its response to injury and disease—all of which are significantly influenced by the mechanical environment. The appendicular joints in the skeleton have evolved to allow almost frictionless articulation while bearing significant mechanical load.1 While physiological loading has long been known to be essential for healthy maintenance of joint homeostasis, supraphysiological conditions can drive degradative processes, such as osteoarthritis (OA).

OA is now considered to be a multifactorial disease of the whole joint.2–4 In cartilage, a cell-mediated shift from anabolism to catabolism by the resident cells, chondrocytes, drives the degeneration of the extracellular matrix (ECM). In the subchondral bone, dysregulation of bone remodeling and osteophyte formation are also part of the disease process. At present, the timing of, and relationship between, these phenomena from a pathophysiological perspective is not clear.1 It is known that the altered structure and composition of joint tissues often suggest that the physical microenvironment of resident cells themselves is changed. However, at a cellular level, understanding the mechanisms by which intrinsic properties of the ECM (e.g., stiffness) and extrinsic forces (e.g., compression/tension etc.) are transduced is non-trivial. Even in a single cell or tissue type (i.e., bone or cartilage) in isolation, the process is not straightforward. In OA, characteristic ECM degradation leads to localized changes in mechanical stress, driving cell stress responses, inflammation, and senescence/apoptosis. Eventually, a feedback loop is established whereby pathological cell phenotypes produce poor-quality ECM. This contributes to the degradation of remaining ECM and so drives further joint destruction.5 Thus, despite the diverse origins and complex etiology of OA, it is clear that the disease progression invariably involves multiple joint tissues as well as alterations in their mechanobiological properties.

Studying this multitissue dynamic joint system, and the complex manner in which it changes with injury and disease, remains challenging. Representative laboratory model systems have been used for many years, but a true unifying representative system has proved difficult to achieve.6 In vivo preclinical animal models provide many advantages over laboratory-based in vitro options as they allow the whole joint to be considered in the context of a native tissue and mechanical environment. Despite this, many issues surrounding their use remain; they are complex, costly, and time-consuming and require significant ethical consideration (adherence to 3R's principles). In addition, there are several biological concerns with their use, including differences between the human and animal model immune systems, manner of joint loading, and thus cell signaling pathways as well as mode of initiation of OA.7 The development of more accurate models would, therefore, be a significant contribution to this field of research.8 Conventional in vitro models show some benefits due to their well-established and reproducible methods and relatively user-friendly nature but often fail to recreate accurate cell environments. Recently, an increased appreciation of the importance of cell-level mechanobiology has emerged, and these considerations will likely be central to future developments. Alongside these advancements, the development of ex vivo or explant models that accurately recreate elements of OA pathology (in particular, the consideration of multiple tissues), including the mechanical environment, has led to a rapid expansion in their use. Explant models can be beneficial as they recapitulate aspects of in vivo conditions in the laboratory without some of the challenges of in vivo work. In particular, these systems may have value in short-term studies and drug screening applications. Advances in materials science and regenerative medicine will also likely feature heavily in the development of more complex (and ideally more representative) in vitro and ex vivo models that attempt to accurately recreate cell and tissue physiochemical microenvironments.6 This review will first discuss recent advances in the current understanding of mechanosensory processes in healthy and osteoarthritic joints and advancements made in the development of in vitro and ex vivo model systems ranging from two-dimensional (2D) monocultures through to joint organ-on-a-chip models. Finally, details of therapeutic targets identified and tested using these models will be discussed.

BASIC FORM AND FUNCTION OF THE OSTEOCHONDRAL TISSUES

Osteochondral joint tissues experience mechanical stress over a wide range throughout their lifetime.9 Bone is a key structural and protective element of the skeleton and is a complex, heterogeneous anisotropic material. The subchondral bone can be subdivided into several components, which combine to perform specific functions.10,11 The subchondral plate, directly adjacent to articular cartilage, primarily prevents shear forces from damaging joint tissues by dispersing them as compressive and tensile forces.12 Below the subchondral plate, the bone becomes a porous network of trabecular bone in which individual trabeculae are orientated along local “stress-lines,” as described by the “Wolff's law.”12,13 This load-sensitive structure is maintained through the coordinated action of resident bone cells: osteoblasts, osteoclasts, and osteocytes. In healthy bone, it is the resident (and most numerous and ubiquitous) osteocytes that “sense” when and where bone tissue needs to be removed.14–16 A pro-osteoclastogenic signaling cascade is produced, such that osteoclasts can carry out this task via resorption, and that tissue is then replaced via coupled osteoblast-mediated bone formation.17 This process is continuous, dynamic, and is aided and influenced by a range of specialized growth factors and hormones. Multiple mechanosensing mechanisms have been identified in these cells, including membrane channels, integrins, the cytoskeleton, and primary cilia.18–22

The structure of articular cartilage is also highly adapted to withstand mechanical loads, which it absorbs and disperses in an almost frictionless environment during movement. Cartilage achieves this through the anisotropic, hierarchical structure, and specialized composition of its ECM. The anisotropy of cartilage can be seen at the ECM level from the joint surface to the subchondral bone in its superficial, middle, and deep/calcified zones, and at the pericellular level (i.e., pericellular, territorial, and inter-territorial ECM).23,24 At the joint surface, superficial zone chondrocytes are flattened and aligned parallel to joint surfaces and produce a low-friction matrix rich in hyaluronan and lubricin. Deeper in the tissue, middle zone chondrocytes are larger/rounder, and type II collagen fibrils increase in thickness. Aggrecan is the most abundant hydrophilic proteoglycan in cartilage, and its concentration increases as a function of tissue depth.2,24–26 Other strongly hydrophilic proteoglycans and associated glycosaminoglycans allow cartilage to retain large quantities of water, which facilitates tissue resistance to compressive forces and decreased friction during movement. Many of the cellular mechanosensing mechanisms that exist in bone also exist in chondrocytes, including membrane channels, integrin activation, the cytoskeleton, and primary cilia, which have significant effects on anabolic and catabolic cell processes.27

During skeletal loading, complex gradients of mechanical stress and biochemical signals are generated that have profound impacts on cellular responses in the joint. The mechanisms of these important processes are yet to be fully understood.28,29 Furthermore, during the initiation and progression of injury/disease, it becomes even more important to understand these processes to develop new and more effective treatments.

MECHANISMS OF MECHANOTRANSDUCTION IN OSTEOCHONDRAL TISSUES

In order for cells in osteochondral tissues to respond to their physical environment, they must have the ability to sense specific parameters of the mechanical loads they sustain, such as its type, duration, and frequency. Multiple sensory mechanisms by which osteochondral cells achieve this have now been identified (Fig. 1) and are reviewed below.

FIG. 1.

FIG. 1.

Extracellular matrix environment and mechanosensory mechanisms in (a) cartilage and (b) bone. In cartilage, chondrocytes possess a number of cell surface receptors involved in mechanotransduction including integrins, syndecans CD44, and discoidin domain receptor 2. Mechanosensitive ion channels such as transient receptor potential cation channels, piezo-channels, and connexons. The primary cilium, a mechanosensory organelle, contains high concentrations of mechanosensing machinery central to cell responses to mechanical forces. Chondrocytes are immediately surrounded by the pericellular matrix (PCM), which is characterized by the expression of type VI collagen. The PCM acts as a force transducer for the cell and determines the local mechanical and biochemical environments. Moving out the PCM integrates with the territorial matrix (TCM), which contains fine type II collagen fibers, proteoglycans, and glycosaminoglycans like keratin sulfate (KS), chondroitin sulfate (ChS), heparin sulfate, hyaluronic acid, and aggrecan. The TCM acts with the PCM as a reservoir for growth factors, which are released or presented to cell surface receptors on mechanical deformation. The TCM integrates with the interterritorial matrix (ICM) containing large type II collagen fibers, along with non-fibrillar collagens like type IX collagen, which is associated with cartilage oligomeric matrix protein (COMP). The TCM also contains high levels of hyaluronic acid, aggrecan, and other proteoglycans, which retain high amounts of water within the ECM.

Mechanotransduction in bone tissue

By virtue of their function, joints experience loading as a complex combination of compression, tension, shear, and hydrostatic and osmotic pressures. At a basic level, compressive forces, generated by body weight, are transmitted through surface cartilage to the subchondral bone and then away from the joint surface.

The relatively high stiffness of the mineralized bone means that strains experienced by resident cells (osteocytes) in vivo are significantly lower than in the cartilage-of the order of 0.05% strain during normal activity and 0.2% during strenuous activity.30 Some mechanosensory mechanisms are common between bone and cartilage cells, including integrin activation, intracellular kinase cascades [e.g., mitogen-activated protein kinase (MAPK), extracellular signal-regulated protein kinase (ERK1/2)], intracellular calcium release, and membrane channel activation.31–33 As discussed above, osteocytes play a dominant role in mechanosensation in bone. Ablation of osteocyte populations significantly increases porosity in cortical bone and decreases overall sensitivity to mechanical loading.34 Integrin activation is central to bone cell responses to deformative and non-deformative loadings. In particular, β1 and β3 integrins are important for mechanosensing in osteoblasts and osteocytes (notably the same is true for chondrocytes—detailed below).35,36 Mice with osteoblast or osteocyte-specific dominant negative forms of β1 integrin or β1-ablation exhibit reduced bone mass and increased porosity due to increased osteoclast activity.37,38 In osteocytes, β1 integrin is preferentially expressed in the cell body, while β3 is predominately expressed along the cell processes.39,40 Aside from integrin signaling, hydrostatic pressure and fluid flow are critical mediators of bone remodeling. Hydrostatic pressures over a broad range (∼5 kPa–4 MPa) have been reported to influence bone cell behavior. Dynamic pressures ∼10 kPa have been shown to regulate 3′,5′-cyclic monophosphate (cAMP) and Cyclic guanosine monophosphate (cGMP) cellular accumulation, increase alkaline phosphatase activity, osteogenic gene expression, ECM mineralization, and promote resorptive-like phenotypes in osteoclasts.41,42 Dynamic pressures in the ∼30–100 kPa range promote anti-osteoclastogenic phenotypes in bone marrow cells. Even larger dynamic pressures (in the MPa range) have been shown to increase cell–cell and cell-matrix adhesions, promote actin reorganization in the cytoskeleton, and, in bone explant tissues, promote cell viability.43,44 Localized load-induced pressure fluctuations also generate low velocity fluid flow through the canicular network, which is now thought to be the primary mechanism by which osteocytes sense mechanical loading. Osteocyte responses to fluid flow require a functioning actin cytoskeleton and involve primary cilia, with loss of the cilia attenuating cell mechanosensitivity. On experiencing fluid flow, osteocyte signaling generally involves release of nitric oxide (NO), Adenosine triphosphate (ATP), Ca2+, and activation of ERK1/2, which, in turn, regulates numerous bone remodeling pathways including receptor activator of nuclear factor kappa-B ligand (RANKL) expression, cell proliferation, matrix metalloproteinase-13 (MMP13) expression, and osteogenic differentiation of MSCs.45–48 Key differences in osteocyte responses to oscillating and unidirectional flow have been reported.18,49–54 For example, unidirectional flow increases intracellular calcium through the release of intracellular stores and membrane channel activation, while no such activation of membrane channels has been reported in cells subjected to oscillatory flows.50,55

In addition to being highly mechanosensitive, osteocytes are also expert communicators. Despite being embedded in the mineralized matrix, osteocytes directly communicate with each other and with other local cell-types through gap junctions.56,57 This allows for the transmission of information from one part of the matrix to another, thus, in turn, allowing for the regulation of bone formation and turnover.58 Furthermore, gap junction phosphorylation and activity are, in part, mechanically regulated,59 and in the case of osteoblasts, they have been shown to open and become phosphorylated in response to fluid flow, resulting in increased ATP and prostaglandin release.59–61 While the blocking gap junction activity was found to prevent fluid flow-mediated production of osteopontin and osteocalcin.62 Taken together, these findings demonstrate that mechanotransduction is a central aspect of healthy bone function.

Mechanotransduction in cartilage tissue

Since cartilage covers the majority of articulating surfaces, it naturally takes the majority of the load imparted at the joint. At the cell level, chondrocytes perceive load-induced tissue deformation through a secreted pericellular matrix (PCM).63 The PCM is characterized by the presence of collagen type VI,64 perlecan,65 aggrecan,66 laminin,67 fibronectin,68 hyaluronan,69 biglycan,70 and type IX collagen.71 The PCM can modulate mechanical stress, osmotic pressure, and fluid-flow in the chondrocyte microenvironment, thus acting as a key regulator of mechanotransduction.72 Though significantly stiffer than the chondrocyte itself (Young's modulus ∼40–100 vs ∼0.5 kPa), the PCM is softer than the cartilage ECM surrounding it (∼0.1–2 MPa).73–76 The PCM and Territorial Matrix (TCM) also have a role as natural, mechanoresponsive growth factor reservoirs. For example, TGFβ and FGF bind to heparin sulfate domains in PCM/territorial matrix molecules, such as perlecan, and can be released to activate cell receptors by mechanical deformation.72,77,78

Chondrocytes also sense tissue deformation directly through integrin-matrix adhesions. Under physiological load, integrins initiate chondrogenic transcription through several mechanisms including the 3′,5′-cyclic monophosphate (cAMP) signaling cascade79 and actin cytoskeletal stress-mediated activation of protein kinase A (PKA).80 Subsequent activation of cAMP-response element binding (CREB) protein converges on SOX9 phosphorylation, driving chondrogenic gene expression.81–83 Additionally, integrin-associated kinases, such as Proto-oncogene tyrosine-protein kinase Src (SRC) and focal adhesion kinase (FAK), activate chondrogenic gene expression through ERK–MAPK-P38 signaling cascades.84 Cell deformation also activates membrane channels [PIEZO and transient receptor potential (TRPV) family], causing a Ca2+ influx into the cell.85 In particular, TRPV channels appear to have a significant role in physiological loading responses. Loss of TRPV4 leads to disruption of cartilage homeostasis and induction of OA,86,87 while in vitro, the addition of a TRPV4 agonist increased ECM production.86 Signaling downstream of TRPV4 has yet to be fully elucidated, but studies have shown G protein-coupled receptor (GPCR) pathway activation [phosphoinositide 3-kinase/Akt/forkhead box protein O (PI3K/Akt/FOXO)], which is thought to be involved in preventing cartilage damage and the onset of premature hypertrophy.88–90 Ca2+ influx also triggers ATP release through hemichannels (connexons) in the cell membrane and activates ERK1/2 through the anti-catabolic transactivator CITED2 (cbp/p300-interacting transactivator 2).91–95

In cartilage, as in bone, hydrostatic pressure and fluid flow also play a role in homeostasis of the local microenvironment. Due to the high-water content of cartilage, hydrostatic pressure bears approximately 90% of applied loads in the tissue.96–98 Pressures generated are generally in the 3–10 MPa range (but can reach 18 MPa at certain sites, such as hip joints). Hydrostatic pressure does not deform the tissue itself, but cell adhesion, in particular through α1β1 integrin, remains important for chondrocyte responses to hydrostatic pressure.99,100 Hydrostatic pressure, along with osmotic pressure, has significant effects on the activity of transmembrane ion channels and pumps, inhibiting Na/K pump and Na/K/2Cl transport activity but increasing Na/H pump activity and activating TRPV4.86,87,101–103 Pressure also activates the purinergic signaling pathway and drives a Ca2+ response through inositol-triphosphate-mediated release from sarcoendoplasmic reticulum stores (SERCs).104 Inhibition of SERCs, hemichannels, purine receptors, or extracellular ATP blocks cell responses to hydrostatic pressure.105,106 Other pathways involved in responses to hydrostatic pressure include estrogen receptor ERα-mediated activation of c-Jun N-terminal kinases (JNK) and increased transforming growth factor receptor (TGFR)I activation.107,108

Again, similar to bone, pressure variations in cartilage also generate interstitial fluid flow within cartilage, influencing chondrocyte biosynthetic activities.109–111 Numerous studies have shown that low fluid shear stress (∼2–10 dynes/cm2) has a chondroprotective effect and initiates repair mechanisms in chondrocytes, whereas high shear stress (∼10–20 dynes/cm2) can induce inflammation, cell death, and cartilage degradation.112–118 Chondrocyte responses to shear stress also appear to be time-dependent, with short duration (1–2 h) stimulation reducing catabolic responses while a longer stimulation (3–4 h) does not.118 Again in this case, shear stress appears to be detected by the primary cilia,86,87,119–121 which trigger mechanotransductive signaling cascades and ionic fluxes,120 a number of which converge on ERK1/2 and P38 signaling.122 Blocking ERK1/2 and P38 with small molecule inhibitors suppresses shear-related increases in ECM production and remodeling.

Pathological changes in osteochondral tissue and mechanotransduction in OA

Subchondral bone and cartilage undergo degenerative changes during OA progression (Fig. 2). One of the most pressing questions in the field is whether changes in the subchondral bone occur before or after cartilage changes, and whether changes in the two tissues are causally linked, or independently changing in parallel. Recent clinical studies have suggested that in fact, bone remodeling and composition changes occur prior to detectable changes in cartilage.123,124 In the case of post-traumatic OA (PTOA), in the initial aftermath of injury, a transient loss in subchondral bone has been observed through increased osteoclast activity. This period is then followed by an increase in bone formation, density, and volume as OA becomes more advanced.125–128 Interestingly, the bone formed in that period is often of poor quality, consistent with the idea that this is a rapid injury-response. Similarly, osteophyte formation occurs at the joint margins in later stages of disease, which causes pain and discomfort. These subchondral changes have long been hypothesized to play roles in disease progression through both biomechanical means and also biochemical bone-cartilage crosstalk.129,130 However, the precise mechanisms driving these processes remain elusive. It has long been known that subchondral porosities allow mass transport between bone and cartilage compartments in both healthy and OA joints.131–133 Increased osteoclastic activity is thought to increase plate porosity, at least in the initial stages of OA, facilitating crosstalk.125 This increased porosity is also linked to vascular invasion of the deep cartilage and aberrant chondrocyte hypertrophy.134–139 Studies using murine OA models support this idea, with both cartilage damage and vascular invasion coinciding with increased subchondral porosity and increasing in exchange of soluble factors.134,140 In addition to porosity, there is evidence that repetitive loading of joints creates subchondral bone microcracks, even in healthy joints.141,142 These microcracks also drive targeted remodeling, through localized osteocytes production of RANKL and decreased osteoprotegerin production.15,143,144 Further study is required, however, to understand these acute and chronic responses. As mentioned above, OA-associated accelerated bone remodeling is also connected to lower bone mineralization. OA osteoblasts appear to generate abnormal type I collagen, producing a homotrimer composed of α1 chains rather than the healthy heterotrimer formed from two α1 chains and one α2 chain.145,146 This abnormal collagen I production may contribute to deficient matrix mineralization.12 In addition to this, OA osteoblasts produce increased interleukin-6 (IL-6), prostaglandin E2 (PGE), and TGFβ; the latter drives increased Dickkopf-related protein 2, an inhibitor of mineralization.12,146,147 The significance of this shift in bone composition and structure to the overall disease progression needs further exploration. This deficient mineralization is continued as OA progresses and bone deposition and thickness increase, resulting in more bone being present in the area (sclerosis), but reduced subchondral ECM stiffness.148–151 Other animal and human studies have examined the effects of inhibiting bone remodeling on OA initiation and progression through the use of bisphosphonates.152–157 In these models, bisphosphonate treatment inhibits bone remodeling and attenuates degeneration of cartilage. However, to date in the clinic, while bisphosphonate treatment has been shown to be effective at inhibiting bone remodeling, they have not shown conclusive effects on modulating cartilage degeneration in OA patients.

FIG. 2.

FIG. 2.

Progression of osteoarthritis in cartilage and bone. In early stage OA, cartilage ECM degeneration by matrix degrading enzymes, such as matrix metalloproteinases, increases frictional, shear, and tensional stress on movement. Changes in cartilage ECM composition decrease tissue hydration, altering fluid flow and hydrostatic and osmotic pressures on loading. The subchondral bone plate decreases in thickness and increases in porosity, facilitating increases in bone-cartilage crosstalk and altering load distribution. These structural changes lead to the development of localized mechanical stress within the tissue, triggering the initiation of catabolic cell responses and mechano-inflammation. Senescent and apoptotic cells secrete disease-propagating molecules [senescence-associated secretory phenotype (SASP)]. The subchondral bone and calcified cartilage also become increasingly innervated and vascularized, contributing significantly to pain development and progression of inflammation. In late-stage OA, high friction, pathological ECM composition, and dehydration drive fissure cartilage fissure formation. Aberrant hypertrophic chondrocyte phenotypes are observed, and the calcified cartilage thickness increases. Sclerotic bone formation thickening of the subchondral bone plate occurs. Mechanical functionality of the joint is compromised, and inflammation and SASP-related secretion drive end stage degeneration and joint pain.

Osteocyte density and morphology are also altered in the subchondral bone of OA joints. The number of viable femoral head osteocytes is decreased in OA, while their markers are dysregulated, and cell–cell communication via gap junctions is decreased. These changes correspond to an increase in new bone formation and total bone volume, although as above—the mineral content and quality are altered.158–160 Recent reports have highlighted a fascinating link between OA and dysregulation of osteocyte remodeling of their perilacunar/canalicular channels (PLR).161–165 In these studies, MMP13 was selectively ablated in murine osteocytes, but not in chondrocytes.161 Not only these mice suppressed PLR in cortical and subchondral bone, but these changes also significantly impacted cartilage, reducing proteoglycan content, altering the production of type II collagen, aggrecan, and MMP13, and increasing the incidence of cartilage lesions. All of which are consistent with the development of early OA. These findings highlight a role for osteocyte-cartilage crosstalk, and in particular, a causal role for suppressed PLR in onset of OA.

In cartilage, OA-related degradation of the hydrated, proteoglycan-rich cartilage matrix leads to macroscopic disruption in the form of fissuring, and chondral flaps or tears.26,166 At the cellular level, PCM degradation is an early event during OA progression and has a significant impact on the mechanical environment of chondrocytes.167 Recent work showed that preventing this PCM degradation is sufficient to modulate overall disease progression.168 Several studies suggest that PCM degradation during OA leads to modulus reductions of between 30% and 50% and increased intracellular Ca2+ signaling.169,170 These changes in pericellular environment have been linked to cell organization changes observed in OA, whereby healthy columns of cells become disorganized clusters.171–174 Recently, this disorganization was used to categorize OA degradation, and chondrocyte PCM stiffness was measured at each stage. Strikingly, significant decreases in PCM stiffness were found between each stage of cellular disorganization and by extension OA progression.171 Intriguingly, the complete loss of PCM has been associated with the appearance of long cytoplasmic processes (>8 μm) on chondrocytes, which extend into the territorial ECM.175,176 Aside from the direct mechanical implications of decreased PCM stiffness, recent work indicates that such changes are also important in cellular responses to biochemical signaling. PCM degradation also increases chondrocyte exposure to abnormal ECM adhesion sites, specifically type II collagen fibrils (which are more highly expressed in the surrounding ECM rather than the healthy PCM). It is currently thought that this increased interaction, most notably through discoidin domain receptor 2 (DDR2), has significant effects on the cell metabolic process and cell signaling downstream of the receptor,177,178 including the upregulation of MMP13 expression.179,180

When joint injury or disease causes the development of an abnormal mechanical environment, chondrocytes receive damaging mechanical stimuli and driving catabolic and proinflammatory processes. Studies suggest that piezo-channels are activated in response to supraphysiological loading (strains of 13%–45%), leading to pathologically high intracellular Ca2+ concentrations and hyperactivation of downstream responses, cell damage, and apoptosis.181,182 Inhibition of piezo-channel activity protects chondrocytes during overload and reduces subsequent cell death.85 Despite its role in healthy responses to loading, recent work has shown that under pathological loading, TRPV4 can also trigger apoptotic responses in chondrocytes.183 Intracellularly, the transforming growth factor-β-activated kinase 1 (TAK1)-JNK2 cascade is critical to injury responses, acting as an upstream regulator and driving the expression of inflammatory markers and matrix catabolism.184,185

IN VITRO MODELS TO STUDY OA AND OSTEOCHONDRAL MECHANOBIOLOGY

Understanding these complex processes in both bone and cartilage tissues individually and in the complete osteochondral unit requires novel experimental approaches and/or a combination of in vitro, ex vivo, and in vivo models. In the sections below, we discuss current experimental models and recent developments, which will help researchers in this field unravel the mechanisms of this condition.

Two-dimensional (2D) culture systems to study osteochondral mechanobiology

Single cell type model systems

Two-dimensional culture systems are widely used to examine cell signaling, responses to stimuli, and screen therapeutics. Often, these cell culture models consider single cell populations in isolation (chondrocytes for cartilage; osteoblasts, osteocytes, and osteoclasts for bone). These approaches have utility for studying cell-level responses to stimuli due to the ability to tightly control experimental conditions, rapidly screen multiple experimental parameters and their relative low cost. In the context of the joint, 2D culture systems are commonly used to model OA environments through the supplementation of culture media with proinflammatory cytokines, such as IL-1β and tumor necrosis factor (TNF)-α. The simplicity of such approaches has allowed interrogation of many of the individual intracellular signaling pathways now known to be critical in joint anabolic and catabolic processes. Despite these benefits, these conventional approaches often neglect the significant impact of mechanical stimuli and environment in the osteochondral cell function. Therefore, in recent years, these models have progressed beyond static culture on stiff tissue culture plastic to allow investigators to control the cellular mechanical, along with the biochemical, culture environment. These include 2D hydrogels controlling stiffness and adhesion motifs (i.e., cells seeded onto hydrogel substrates), dynamic substrate tension/cell stretching systems, and systems allowing the generation of fluid flow/shear stress and hydrostatic/dynamic pressures (Fig. 3).186–193

FIG. 3.

FIG. 3.

Two-dimensional culture models to study mechanotransduction in osteochondral cells. (a) Uniaxial strain can be applied to cells in monolayer culture by seeding onto deformable membranes. In a typical setup, a vacuum can be applied pulling a polymer membrane uniformly around a loading post and creating a uniaxial strain in the stretched membrane. (b) Simple two-dimensional fluid flow experiments can be conducted in cone viscometer culture, which allows accurate establishment of defined fluid flow across the culture substrate. (c) Fluid flow chambers allow analysis of fluid flow across multiple substrates, which can include different cell types and culture setups simultaneously. Due to directional fluid flow, crosstalk between culture substrates may be limited. (d) Hydrostatic pressure can be applied to cultures through control of culture chamber pressurization. These systems can be used to apply constant or oscillatory hydrostatic pressures to cells in culture over relevant physiological and pathological ranges. (e) Microfluidic devices offer diverse options for steady state and oscillatory two-dimensional culture fluid flow analysis. Alongside this, through the chip design of culture chambers and channels, co-cultures and varied conditions can be achieved simultaneously to recreate in vivo conditions and probe pathology relevant signaling.

One simple system that delivers mechanical stimulation to 2D cell cultures involves seeding cells onto pneumatically or electromagnetically deformed membranes, which can deliver predetermined levels of strain over the designated culture surface. Chondrocytes stimulated in this way with physiological-like tensile strain (0.5 Hz, 10% strain for a duration of 24 h) decrease the expression of catabolic enzymes and increase the expression of anabolic markers such as aggrecan.194–196 Larger strains and durations were shown to have an opposing effect, increasing catabolic MMP-1, -3, -9, and -13 expression.195–197 Similar studies using 2D membrane-stretching systems seeded with bone cells identified signaling pathways involved in responses to stretch, which are directly translatable to more complex models and even the in vivo environment. For example, in osteocytes, cell stretching (5% elongation over 1–20 min) has an anti-apoptotic effects through ERK1/2 activation.198 In osteoblasts, numerous studies applying cyclical stretching have shown that osteoblastic maturation is accelerated, osteogenic gene expression is increased, and ECM deposition enhanced,199–201 while in osteoclasts, bone resorbing activity is increased by cyclic stretching in 2D cultures.202 These devices have been limited by their relatively low through-put capabilities. However, ongoing research is significantly increasing the throughput of these types of stretching systems.203,204

Aside from cell stretching, fluid flow systems allow the investigation of the effects of shear on osteochondral cells. These typically involve application of controlled unidirectional or oscillatory fluid movement in a cell culture chamber and range from rocking cultures and cone viscometers to complex microfluidic devices. In chondrocytes, cone viscometer experiments have helped to define flow conditions that elicit catabolic or anabolic responses and study morphological and molecular responses. For example, continuous, unidirectional laminar fluid flow (1.64 Pa) was found to significantly decrease the expression of type II collagen and aggrecan and increase nitrite levels in culture media, indicating cell stress.205 Higher shear stress regimes (3.5 Pa for 4 days) result in rounded chondrocyte morphology in comparison to static cultures,206 while shear stresses of 2 Pa were found to regulate IL-6, toll-like receptor (TLR)4, and caveolin-1 synthesis in a cyclooxygenase-2 (COX-2)-dependent manner.207 Similarly, the effects of fluid flow on bone cells have been studied in a variety of experimental systems.208,209 For example, in a parallel plate chamber, oscillatory flow was found to increase osteoblastic marker gene expression and modulate the activity of alkaline phosphatase (ALP) in mesenchymal stem cells (MSCs).210–212 Pairing these mechanostimulatory techniques with -omics analysis provides valuable insights into cell responses to similar stresses that might occur in vivo. For example, pairing oscillating fluid flow with transcriptomic microarray analysis in osteocyte-like cells (MLO-Y4) demonstrated that at 1 Pa peak shear stress for 2 h, ATP producing enzyme nucleoside-diphosphate kinase (NDK), calcium-binding calcyclin, and G-protein couple kinase 6 were all significantly upregulated.213 Such investigation has indicated that bone cells respond differentially to oscillatory and steady flow, with oscillatory flow conditions being advantageous for bone formation in vitro.212,214,215

Hydrostatic pressure can also be simulated in culture to study osteochondral cell responses. Applying intermittent hydrostatic pressure over a range (1–4 Hz, at 1, 5, and 10 MPa for 4 h per day for 4 days) through hydraulic loading was found to alter healthy and OA human chondrocyte phenotypes.216 Application of higher pressures (5 and 10 MPa) corresponded with the upregulation of aggrecan and type II collagen at gene and protein levels.216 This experimental approach also allows the simultaneous study of the effects of mechanical and biochemical stimuli by supplementation of culture media. For example, in a separate study, bone morphogenetic protein (BMP)-2 was applied to human OA chondrocytes, with and without the application of hydrostatic pressure (10 MPa, 1 Hz, 4 h a day for 4 days). Through this work, it was demonstrated that the growth factor and mechanical stimuli had complementary effects. BMP-2 was found to increase aggrecan, but when applied with hydrostatic pressure, an increase in type II collagen was also observed. The expression of the catabolic marker MMP-2 was also decreased with hydrostatic pressure application but not when BMP-2 was supplied alone.217 These results highlight how these systems can further our understanding of the mechanical and biochemical interplay required for healthy anabolic chondrocyte phenotypes and also the fine balance that exists between different stimuli. Interestingly, while physiological levels of hydrostatic pressure (5–10 MPa) have been shown to decrease catabolic marker genes like MMP-13 and a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5), high hydrostatic pressures in the range of 50 MPa (replicating joint overloading) have been shown to increase the expression of vascular endothelial growth factor (VEGF), which could be one of the mechanisms by which vessel invasion of the cartilage is increased in OA.218–220 Interstitial fluid flow in bone through the lacunar-canalicular system is important to maintain bone homeostasis. Similar to work with chondrocytes, simple cyclic hydrostatic pressure bioreactors can be used to investigate bone cell responses. Using such approaches, cyclic hydrostatic pressures in the range of 10–300 kPa (0.5–2 Hz) have been found to increase the expression of osteogenic markers like RUNX Family Transcription factor 2 (RUNX2) and osteopontin in MSCs.221 In vivo, the mode of mechanical stimulation may impact cell responses in a cell-type dependent manner. Comparisons of the effects of fluid flow and hydrostatic pressure on osteoblastic MC3T3 cells showed that the two modes of mechanical stimulation had differing effects, for example, fluid flow has increased ATP release and F-actin fiber formation, while hydrostatic pressure did not, despite both increasing COX-2 expression.222 This further highlights the need for careful selection of the most relevant mechanical stimuli for the investigation of specific signaling responses.

Co-culture model systems

2D co-culture systems allow for the investigation of multiple cell types cultured in shared environments and can help determine the complex bone-cartilage signaling crosstalk processes in osteochondral tissues. Critical parameters for co-cultures include the type of cells, culture media, order in which cells are cultured, and numbers/ratio of cell types. Even though optimization of these technical details can be time consuming, such co-cultures are valuable tools to systematically increase culture complexity and move closer to recapitulating real tissue microenvironments while retaining control over experimental conditions.223 These systems can involve direct cell–cell contact or cells not in direct contact but sharing culture environments, for example, through the use of cell culture inserts or microfluidic chambers. Such systems have been employed to determine, for example, that co-culturewith chondrocytes can improve the chondrogenic differentiation of MSCs through mechanisms involving both soluble factor secretion and cell–cell contact.224–226 As with monolayer cultures, combining these co-cultures with methods to mechanically stimulate the cells will provide new insights into their function and behavior. For example, combining tensile stimulation with co-cultures of MSCs and chondrocytes was reported to increase chondrogenic phenotypes and rapidity of cell expansion.227 Though few reports of 2D osteochondral co-cultures under tensile stimulation exist, the examination of fluid shear on cell–cell communication is a promising area of research, particularly through microfluidics. Microfluidic systems can be used to create complex multicompartment co-cultures with precise control of fluid flow and physical parameters, while the integration of sensors allows direct read-outs of cell responses.228 These microfluidic approaches have been used to show crosstalk between bone cells in response to fluid flow. For example, osteocyte-like MLO-Y4 cells were cultured with osteoclasts (RAW264.7) and exposed to 0.5 Pa shear stress simulation, which resulted in a decrease in RANKL expression in osteocytes, which suggests a reduced osteoclastogenic phenotype.229

Though these co-cultures are useful for examining cell–cell interactions, particularly in response to stimuli, the limitations of 2D cultures still apply, not least in terms of altered cell morphologies, a lack of physiological ECM, and cell–ECM interactions.192 The use of 2D models in OA research has been hugely beneficial and has provided many of the major steps forward in this field of research as well as establishing fundamental tenets of disease. Nonetheless, some aspects of disease, which are now recognized as being particularly important, cannot easily be captured by these methods.

Three-dimensional (3D) model systems to study cartilage and bone mechanobiology

Three-dimensional culture systems can recapitulate in vivo environments for cartilage and bone cells, where they are surrounded by and interact with ECM. In vitro 3D culture models have been developed using a number of biomaterial formats and from a range of naturally and synthetically derived polymers. The most prevalent material formats include lyophilized polymer scaffolds and hydrogels.230 These have been produced from natural polymers such as collagen (including gelatin), hyaluronic acid, chondroitin sulfate and alginate, or synthetic polymers, including polyethylene glycol, poly-D,L-lactic acid (PDLLA), and poly(N-isopropylacrylamide (PNIPAM). These polymers confer mechanical and biological properties to materials fabricated from them, which can be tuned to provide cues that can control specific cell responses or mimic the properties of the natural ECM. Natural, ECM-derived materials may also have the added advantage of containing cell attachment motifs as well as an inherent bioactivity that can be cell-instructive. Though alone materials fabricated from these natural polymers may not have sufficient mechanical properties (or range of mechanical properties achievable) for models of the load-bearing joint tissues, they can be easily modified chemically to achieve this. One example of this is gelatin methacryloyl (GelMA), which has been shown to be biocompatible, bioactive, and non-immunogenic.231 Other native osteochondral ECM constituents such as hyaluronic acid and chondroitin sulfate are commonly incorporated into materials for both their mechanical and biochemical properties and can be similarly modified chemically to further control their properties.232 Though numerous material formats are available, hydrogel models, in particular, allow tight control of 3D cell environments in vitro facilitating the investigation of the effects of altering intrinsic matrix microenvironment cues (e.g., mechanical and viscoelastic properties, cell adhesion motif density and type, and degradation) and extrinsic mechanical forces (e.g., compression and tension). Through such investigation, more accurate tissue mimics can be produced to create bone cartilage and osteochondral models.

Changes in ECM properties, including stiffness/strength, significantly impact OA pathogenesis and progression. Within cartilage, gradients of stiffness exist in healthy tissue, from the softest superficial zones to the stiffest deep calcified zones. To investigate the impact of ECM stiffness on chondrocyte behavior and function and on the chondrogenesis of MSCs, previous work has tested chondrocyte and chondrogenic MSC responses to 3D hydrogels over a large range of stiffnesses, with wide variation in results. Differences in material composition, cell source, and mechanical testing methods make definitive comparisons difficult. However, in general, the cartilage ECM produced by MSCs and chondrocytes in hydrogels with stiffnesses between approximately 7.5–40 kPa have been reported to be most similar to the hyaline cartilage observed at joint surfaces.233–239 Encapsulation in stiffer materials leads to the formation of hypertrophic chondrocyte phenotypes and initiation of osteogenic processes.237,240,241 The differential effects of stiffness on chondrogenic and osteogenic differentiation are of particular importance in designing models of the osteochondral interface. For example, recently hydrogels with tunable stiffness gradients were developed using gelatin-PNIPAM hydrogels containing both beta-sheet and amorphous silk nanofiber solutions. Through a combination of cross-linking and electric field alignment, a gradient of stiffness mimicking that of the ECM stiffness from the superficial to the deep cartilage/subchondral bone was produced.237 Using this system, MSC chondrogenesis was enhanced in softer regions of the hydrogel, while osteogenic differentiation was favored in the stiffer regions. This and other similar approaches can be used to mimic natural tissue stiffness gradients and produce multiple differentiated cell types from a single seeded population.242–244 Until recently, these investigations into intrinsic hydrogel mechanical cues have typically used elastic materials with varied stiffness or disregarded a material's viscous component. Both bone and cartilage ECM are highly viscoelastic, meaning that they display time-dependent deformation in response to an applied force and corresponding recovery time (relaxation time) for the material to return to its original form. The generation of hydrogels that de-couple stiffness and relaxation times allows the investigation of this effect on cell behavior. For example, alginate-poly(ethylene glycol) (PEG) hydrogels with controllable stiffness (∼3 kPa) and variable relaxation times were used to demonstrate that in faster relaxing gels, bovine chondrocytes significantly increased the volume and interconnectivity of the ECM they produced, while slower relaxation times promoted catabolic processes.245 This result highlights the importance of considering viscoelasticity when designing materials for osteochondral engineering and also the utility of these 3D culture systems in understanding the fundamental mechanobiology of the cells in the joint.

Aside from the intrinsic material/ECM properties of 3D models, the application of extrinsic forces can be investigated. The type of force, magnitude, and frequency of extrinsically applied mechanical forces have been determined to drive differential responses in bone and cartilage cells through 3D in vitro studies, with even nanoscale displacements able to control MSC osteogenic differentiation.246,247 These in vitro 3D mechanically stimulated assays are valuable tools as they allow precise control of loading type, magnitude, duration, and biochemical conditions, to understand thresholds for anabolic and catabolic cell responses and differences in the cell signaling. These can be taken into account in the development of treatment strategies.

In cartilage model cultures, compression is most commonly applied to simulate joint loading. Through these studies, compressive loading has been shown to modulate chondrocyte phenotypes, ECM biosynthesis, and inflammatory responses. For example, when stimulated with dynamic compression at physiological magnitudes, hydrogel embedded chondrocytes show reduced inflammatory response to exogenous IL-1β stimulation, increased MAPK and TGF-β pathway activities, and increase their proliferative capacity.186,190 Similarly, dynamic compression of chondrocytes cultured in PEG hydrogels increased cartilage-specific ECM deposition.248 Aside from compression, these hydrogel systems also allow the testing of other modes of mechanical stimulation, such as cyclical shear. Using different modes of mechanical stimulation has potential to be used to produce cartilage zone specific phenotypic responses or ECM organization in tissue engineered constructs.249

Co-culture and organ-on-a-chip model systems

The next generation of advanced 3D culture systems have the capacity to recreate more complex, joint mimicking environments by supporting multiple cell types or, for example, by directing tissue-specific differentiation MSCs or induced pluripotent stem cells (iPSCs) via controlled physical and biochemical environments250–252 (Fig. 4). Creating realistic bone and cartilage environments facilitates the investigation of crosstalk between the two. 3D co-culture models combining osteoblast culture with alginate bead-embedded chondrocytes were shown to be effective at directing bilateral phenotypic thorough such paracrine interactions. Notably, co-culture increased chondrocyte hypertrophy and matrix mineralization. Similarly, a 3D co-culture model system replicating cell–cell interaction between osteoblasts and chondrocytes in the presence of pulsate cyclic tensile stress (15 kPa; 23% strain) reported bilateral phenotypic change with increased chondrocyte hypertrophy, and down regulation of type II collagen, aggrecan, cartilage oligomeric matrix protein precursor (COMP), and SOX-9 expression.250 Together, these studies suggest that perhaps through positioning osteoblast and chondrocyte cell populations correctly in models, tissue-like gradients can be created in vitro. In a separate study, osteocytes and osteoblasts were seeded in type I collagen hydrogels and stimulated with mechanical loading, which increased type I collagen and prostaglandin E2 (PGE2) expression, demonstrating osteocyte influence on osteoblast behavior in response to mechanical loading.252 A challenging but important aspect of these models is recreating realistic communication between joint compartments or cells, since this is a vital factor in both healthy and diseased joints. One approach involves the creation of microfluidic devices, such as “organ-on-chip,” that contain carefully designed channel and chamber systems to recreate tissue-level mass transport and fluid flow.253 For instance, a “multiorgan chip” has been developed to recapitulate the complexity of the human bone marrow niche. Results from experiments using this device show that it could be used to control colony formation of granulocytes, erythrocytes, macrophages, and megakaryocytes.254 Furthermore, it could be used to regulate the expression of osteopontin, VEGF, angiopoietin 1, and fibronectin in these cells with greater accuracy than in standard monolayer conditions. In a similar study, a novel microchip was used to apply dynamic hydraulic compression of 1 psi at 1 Hz to human bone marrow-derived MSCs, which controlled cell proliferation, differentiation, and increased osteogenic ECM production.255 These methodologies are likely to become ever more important as model systems are developed to incorporate more aspects of the osteochondral microenvironment from macroscale structure down to the level of mass transport and molecular diffusion. However, attaining that level of complexity will require significant research. In the interim, the use of explant (multitissue) systems is another tool in our research armamentarium, which can be used to replicate the complex interactions between osteochondral tissues, while still retaining the levels of control that are required in the laboratory setting.

FIG. 4.

FIG. 4.

Three-dimensional models to study mechanotransduction in osteochondral tissue. (a) Unconfined compression—axial strain. Compression can be applied to 3D models in culture in unconfined (a) and confined (b) systems. For unconfined compression, mechanical testing machines can be used to apply defined deformation with defined parameters, though often these are limited by single sample analysis. Confined dynamic compression can be applied as a uniaxial or biaxial deformation to a three-dimensional construct. These systems can be applied with standard culture ware to increase the throughput of analysis. For biaxial deformation, simultaneous movement of sliding compartments can be used to result in shear deformation. (c) Schematic of a three-dimensional bioreactor to study bone ex vivo explants or scaffold cultures. A hydrostatic pressure bioreactor chamber is used to enable cyclic or continuous mechanical loading through a pumped system to control fluid flow of medium. Z (d) and y represent the direction of the strain created through the trabecular explant.

EXPLANT MODEL SYSTEMS

An alternative to 3D co-culture model system, which as we have seen can be complex, is ex vivo or explant model system. Explants are a valuable tool as they allow investigation of cell responses to controlled stimuli or environments while maintaining some aspects of the in vivo tissue, in particular the native ECM microenvironment. As OA is now recognized to be a disease of the whole joint, the use of osteochondral explants allows for the controlled analysis of how changes in one tissue type might affect another. Traditionally, femoral head or osteochondral core samples, from small or large animals, respectively, have been used for this purpose. These are of particular utility where the highly specialized ECM of bone or cartilage plays key roles in regulating cell behavior in response to specific stages or aspects of disease. While a potential drawback of using explants is that excessive ECM can limit diffusion, both bone and cartilage cells are unique in that they are specifically adapted for low oxygen and nutrient environments, facilitating the effective use of osteochondral explant culture. Furthermore, explants provide increased ability to control the mechanical and biochemical environment compared with in vivo experiments. These factors, among others, have seen an increased interest in the use of the explant model system in orthopedic research as well as many other areas of medicine.

To study the effects of physiological and pathological loading on skeletal tissues, such explant systems can be mechanically stimulated using modified mechanical/material testing protocols. A range of loading modes are available including unconfined compression, indentation, tension, and osmotic and hydrostatic pressures. The parameters that are used with these systems can test tissues responses to a variety of different types of loading and frequencies. For example, simple compression of bovine cartilage explants promotes matrix biosynthesis while dynamic (cyclic and intermittent) loading can differentially stimulate chondrocyte metabolism.256 Similarly, differences in cell and tissue responses to physiological and pathological loads can be studied using modifications of these test setups. For example, impact loading of cartilage explants, at different timepoints, showed a delayed (but significant) biological response following low impact, whereas high impact caused early and strong degenerative changes. The use of high impact (defined as imparting energy levels of 2.8 J) also resulted in a decreased tissue stiffness and increased cell death, which corelated with those degenerative changes. Furthermore, these changes were maintained for 4 weeks, and tissue degradation was manifested as increased glycosaminoglycan release and decreased overall content. These data provide consistent and realistic comparisons with real cases of joint injury and disease and demonstrate the utility of explants in this scenario.257 From a practical perspective, the ease with which biochemical agents can be added or detected in these mechanically stimulated culture environments is an additional advantage. For example, compression of cartilage explants was shown to modulate the proinflammatory responses that are normally generated in response to IL-1β and IL-4 stimulations.258

Explant cultures also have utility in determining the precise loading conditions and regimes that are most relevant to the joint and also how that initial damage can progress to disease. Using these systems, it was determined that applying compressive loading to cartilage explants at low frequency but for long time periods can produce a greater damage response than the same loads at higher frequency over shorter periods.259 Furthermore, close control of the precise magnitude of loading on cartilage explants (compression: 4–25 MPa) demonstrated force-specific apoptotic responses in chondrocytes peaking, as expected at higher magnitudes.259 More recently, this apoptotic response to high magnitude mechanical loading has been linked to increased mitochondrial dysfunction, including decreased basal respiration and ATP turnover, through direct mechanosignaling pathways.260–262 Similar investigations using bone explants have explored loading thresholds for eliciting tissue damage responses and to compare the micro- and macro-architecture of ovine, bovine, and human subchondral bone.263–266 Despite these insights, there remains much to learn about cell responses to loading in the joint, not least the order of cell response events following injury, the extent and importance of cellular crosstalk, and the loading conditions that lead to tissue microdamage. The expanding portfolio of well characterized explant models in the field has the potential to address many of these outstanding questions and provides an important preclinical tool in furthering our understanding of the joint injury and disease.

SUMMARY AND FUTURE PERSPECTIVES

The complexity of the cellular, architectural, and mechanical environments of the joint means that a range of model systems are required to understand the processes underpinning its health and disease. Improving our understanding of these key processes is critical for the identification of potential therapeutic targets. For this, it is important that the correct experimental model is selected to allow interrogation of the relevant question. Research advances over the last decade, in diseases like OA, have made it clear that multiple interacting signaling pathways and cell types must be considered rather than isolated targeting of a single pathway or molecule. In particular, investigating the impact of mechanosignaling on OA-relevant signaling pathways is a rapidly expanding area in the field, but there remain technical challenges. As discussed above, different in vitro and ex vivo models have complementary advantages and can be leveraged to determine mechanisms behind the different physiological and pathological features of the system. In addition, microfluidic/joint-on-a-chip technologies and mechanically stimulated osteochondral explant models allow the examination of bone-cartilage crosstalk in tightly controlled culture environments, and these are likely to be key going forward. Central to the success of these technologies is their ability to accurately recreate in vivo interactions that occur in OA, which, in turn, will allow identification and refinement of drug targets within the joint. As use of advanced joint-on-a-chip and explant systems becomes more widespread, it is important that validation and comparison with model systems (particularly in vivo preclinical models) is carefully conducted. The increased throughput and precision of these osteochondral models will decrease the downstream failure rate of identified therapeutic targets and allow rapid and accurate screening of potential targets. In conclusion, a wide range of bone, cartilage, and osteochondral experimental models are now available to researchers in the field of musculoskeletal medicine. These approaches continue to be refined, and, their complexity increased, to incorporate more cell types, treatments, and stimuli. Recent advances in the development of mechanically stimulated higher-throughput models and explant cultures, along with biomaterials technology, are significantly impacting our understanding of OA pathology. In particular, the ability to create biomaterial environments with spatially controlled physical properties through additive manufacturing techniques, such as 3D printing, provides distinct opportunities for detailed investigation of cell responses and in vitro model fabrication. Furthermore, combining these advances with techniques such as biomaterial-mediated RNA interference will allow the controlled shutdown of signaling pathways involved in cell responses and identification of possible therapeutics. In the near future, this will lead to the development of more effective therapies and disease-modifying drugs for this complex disease.

ACKNOWLEDGMENTS

T.H. acknowledges a Marie Skłodowska-Curie Individual Fellowship from the European Commission through the H2020 project ChondroCONNECT (Project ID: 894837). F.J.O.B. acknowledges funding from the European Community's Horizon 2020 research and innovation programme under ERC Advanced Grant Agreement No. 788753 (ReCaP). I.N.A. is funded under a Science Foundation Ireland Career Development Award held by O.D.K. (17/CDA/4699).

AUTHOR DECLARATIONS

Conflict of Interest

The authors have no conflicts to disclose.

Ethics Approval

Ethics approval was not required.

Author Contributions

T.H. and I.N.A. wrote the manuscript. All authors researched data for the article, made a substantial contribution to discussion of content and reviewed or edited the manuscript before submission.

DATA AVAILABILITY

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

References

  • 1. Goldring S. R. and Goldring M. B., “ Changes in the osteochondral unit during osteoarthritis: Structure, function and cartilage–bone crosstalk,” Nat. Rev. Rheumatol. 12, 632 (2016). 10.1038/nrrheum.2016.148 [DOI] [PubMed] [Google Scholar]
  • 2. Sanchez-Adams J., Leddy H. A., McNulty A. L., O'Conor C. J., and Guilak F., “ The mechanobiology of articular cartilage: Bearing the burden of osteoarthritis,” Curr. Rheumatol. Rep. 16, 451 (2014). 10.1007/s11926-014-0451-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Loeser R. F., Goldring S. R., Scanzello C. R., and Goldring M. B., “ Osteoarthritis: A disease of the joint as an organ,” Arthritis Rheum. 64, 1697–1707 (2012). 10.1002/art.34453 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Aspden R. M. and Saunders F., “ Osteoarthritis as an organ disease: From the cradle to the grave,” Eur. Cells Mater. 37, 74–87 (2019). 10.22203/eCM.v037a06 [DOI] [PubMed] [Google Scholar]
  • 5. Arden N. and Nevitt M. C., “ Osteoarthritis: Epidemiology,” Best Pract. Res. Clin. Rheumatol. 20, 3–25 (2006). 10.1016/j.berh.2005.09.007 [DOI] [PubMed] [Google Scholar]
  • 6. Johnson C. I., Argyle D. J., and Clements D. N., “ In vitro models for the study of osteoarthritis,” Vet. J. 209, 40–49 (2016). 10.1016/j.tvjl.2015.07.011 [DOI] [PubMed] [Google Scholar]
  • 7. Malfait A.-M. and Little C. B., “ On the predictive utility of animal models of osteoarthritis,” Arthritis Res. Ther. 17, 1–14 (2015). 10.1186/s13075-015-0747-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Esch E. W., Bahinski A., and Huh D., “ Organs-on-chips at the frontiers of drug discovery,” Nat. Rev. Drug Discovery 14, 248–260 (2015). 10.1038/nrd4539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Kutzner I. et al. , “ Loading of the knee joint during activities of daily living measured in vivo in five subjects,” J. Biomech. 43, 2164–2173 (2010). 10.1016/j.jbiomech.2010.03.046 [DOI] [PubMed] [Google Scholar]
  • 10. Goldring S. R., “ Role of bone in osteoarthritis pathogenesis,” Med. Clin. North Am. 93, 25–35 (2009). 10.1016/j.mcna.2008.09.006 [DOI] [PubMed] [Google Scholar]
  • 11. Goldring M. B. and Goldring S. R., “ Articular cartilage and subchondral bone in the pathogenesis of osteoarthritis,” Ann. N. Y. Acad. Sci. 1192, 230–237 (2010). 10.1111/j.1749-6632.2009.05240.x [DOI] [PubMed] [Google Scholar]
  • 12. Burr D. B. and Gallant M. A., “ Bone remodelling in osteoarthritis,” Nat. Rev. Rheumatol. 8, 665 (2012). 10.1038/nrrheum.2012.130 [DOI] [PubMed] [Google Scholar]
  • 13. Frost H. M., “ From Wolff's law to the Utah paradigm: Insights about bone physiology and its clinical applications,” Anat. Rec. 262, 398–419 (2001). 10.1002/ar.1049 [DOI] [PubMed] [Google Scholar]
  • 14. Xiong J. et al. , “ Matrix-embedded cells control osteoclast formation,” Nat. Med. 17, 1235 (2011). 10.1038/nm.2448 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Nakashima T. et al. , “ Evidence for osteocyte regulation of bone homeostasis through RANKL expression,” Nat. Med. 17, 1231 (2011). 10.1038/nm.2452 [DOI] [PubMed] [Google Scholar]
  • 16. Dallas S. L., Prideaux M., and Bonewald L. F., “ The osteocyte: An endocrine cell … and more,” Endocr. Rev. 34, 658–690 (2013). 10.1210/er.2012-1026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Eriksen E. F., “ Cellular mechanisms of bone remodeling,” Rev. Endocr. Metab. Disord. 11, 219–227 (2010). 10.1007/s11154-010-9153-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Malone A. M. et al. , “ Primary cilia mediate mechanosensing in bone cells by a calcium-independent mechanism,” Proc. Natl. Acad. Sci. 104, 13325–13330 (2007). 10.1073/pnas.0700636104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Hoey D. A., Tormey S., Ramcharan S., O'Brien F. J., and Jacobs C. R., “ Primary cilia‐mediated mechanotransduction in human mesenchymal stem cells,” Stem Cells 30, 2561–2570 (2012). 10.1002/stem.1235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Litzenberger J. B., Kim J.-B., Tummala P., and Jacobs C. R., “ β1 integrins mediate mechanosensitive signaling pathways in osteocytes,” Calcif. Tissue Int. 86, 325–332 (2010). 10.1007/s00223-010-9343-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Rubin J., Rubin C., and Jacobs C. R., “ Molecular pathways mediating mechanical signaling in bone,” Gene 367, 1–16 (2006). 10.1016/j.gene.2005.10.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Ziambaras K., Lecanda F., Steinberg T. H., and Civitelli R., “ Cyclic stretch enhances gap junctional communication between osteoblastic cells,” J. Bone Miner. Res. 13, 218–228 (1998). 10.1359/jbmr.1998.13.2.218 [DOI] [PubMed] [Google Scholar]
  • 23. Buckwalter J. A., Mankin H. J., and Grodzinsky A. J., “ Articular cartilage and osteoarthritis,” Instruct. Course Lect.-Am. Acad. Orthop. Surg. 54, 465 (2005). [PubMed] [Google Scholar]
  • 24. Sophia Fox A. J., Bedi A., and Rodeo S. A., “ The basic science of articular cartilage: Structure, composition, and function,” Sports Health 1, 461–468 (2009). 10.1177/1941738109350438 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Mow V. C., Ratcliffe A., and Poole A. R., “ Cartilage and diarthrodial joints as paradigms for hierarchical materials and structures,” Biomaterials 13, 67–97 (1992). 10.1016/0142-9612(92)90001-5 [DOI] [PubMed] [Google Scholar]
  • 26. Musumeci G., “ The effect of mechanical loading on articular cartilage,” J. Funct. Morphol. Kinesiol. 1(2), 154–161 (2016). [Google Scholar]
  • 27. Zhao Z. et al. , “ Mechanotransduction pathways in the regulation of cartilage chondrocyte homoeostasis,” J. Cell. Mol. Med. 24, 5408–5419 (2020). 10.1111/jcmm.15204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Grodzinsky A. J., Levenston M. E., Jin M., and Frank E. H., “ Cartilage tissue remodeling in response to mechanical forces,” Annu. Rev. Biomed. Eng. 2, 691–713 (2000). 10.1146/annurev.bioeng.2.1.691 [DOI] [PubMed] [Google Scholar]
  • 29. Choi J. B. et al. , “ Zonal changes in the three-dimensional morphology of the chondron under compression: The relationship among cellular, pericellular, and extracellular deformation in articular cartilage,” J. Biomech. 40, 2596–2603 (2007). 10.1016/j.jbiomech.2007.01.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Burr D. B. et al. , “ In vivo measurement of human tibial strains during vigorous activity,” Bone 18, 405–410 (1996). 10.1016/8756-3282(96)00028-2 [DOI] [PubMed] [Google Scholar]
  • 31. Weyts F., Li Y. S., van Leeuwen J., Weinans H., and Chien S., “ ERK activation and αvβ3 integrin signaling through Shc recruitment in response to mechanical stimulation in human osteoblasts,” J. Cell. Biochem. 87, 85–92 (2002). 10.1002/jcb.10278 [DOI] [PubMed] [Google Scholar]
  • 32. Ishida T., Peterson T. E., Kovach N. L., and Berk B. C., “ MAP kinase activation by flow in endothelial cells: Role of β1 integrins and tyrosine kinases,” Circ. Res. 79, 310–316 (1996). 10.1161/01.RES.79.2.310 [DOI] [PubMed] [Google Scholar]
  • 33. Pommerenke H. et al. , “ Stimulation of integrin receptors using a magnetic drag force device induces an intracellular free calcium response,” Eur. J. Cell Biol. 70, 157–164 (1996). [PubMed] [Google Scholar]
  • 34. Tatsumi S. et al. , “ Targeted ablation of osteocytes induces osteoporosis with defective mechanotransduction,” Cell Metab. 5, 464–475 (2007). 10.1016/j.cmet.2007.05.001 [DOI] [PubMed] [Google Scholar]
  • 35. Grzesik W. J. and Robey P. G., “ Bone matrix RGD glycoproteins: Immunolocalization and interaction with human primary osteoblastic bone cells in vitro,” J. Bone Miner. Res. 9, 487–496 (1994). 10.1002/jbmr.5650090408 [DOI] [PubMed] [Google Scholar]
  • 36. Sinha R. and Tuan R., “ Regulation of human osteoblast integrin expression by orthopedic implant materials,” Bone 18, 451–457 (1996). 10.1016/8756-3282(96)00044-0 [DOI] [PubMed] [Google Scholar]
  • 37. Zimmerman D., Jin F., Leboy P., Hardy S., and Damsky C., “ Impaired bone formation in transgenic mice resulting from altered integrin function in osteoblasts,” Dev. Biol. 220, 2–15 (2000). 10.1006/dbio.2000.9633 [DOI] [PubMed] [Google Scholar]
  • 38. Litzenberger J. B., Tang W. J., Castillo A. B., and Jacobs C. R., “ Deletion of β1 integrins from cortical osteocytes reduces load-induced bone formation,” Cell. Mol. Bioeng. 2, 416–424 (2009). 10.1007/s12195-009-0068-4 [DOI] [Google Scholar]
  • 39. McNamara L., Majeska R., Weinbaum S., Friedrich V., and Schaffler M., “ Attachment of osteocyte cell processes to the bone matrix,” Anat. Rec. 292, 355–363 (2009). 10.1002/ar.20869 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Wang Y., McNamara L. M., Schaffler M. B., and Weinbaum S., “ A model for the role of integrins in flow induced mechanotransduction in osteocytes,” Proc. Natl. Acad. Sci. 104, 15941–15946 (2007). 10.1073/pnas.0707246104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Rodan G. A., Bourret L. A., Harvey A., and Mensi T., “ Cyclic AMP and cyclic GMP: Mediators of the mechanical effects on bone remodeling,” Science 189, 467–469 (1975). 10.1126/science.168639 [DOI] [PubMed] [Google Scholar]
  • 42. Klein‐Nulend J., Roelofsen J., Semeins C. M., Bronckers A. L., and Burger E. H., “ Mechanical stimulation of osteopontin mRNA expression and synthesis in bone cell cultures,” J. Cell. Physiol. 170, 174–181 (1997). [DOI] [PubMed] [Google Scholar]
  • 43. Saito S. et al. , “ Involvement of PGE synthesis in the effect of intermittent pressure and interleukin-1β on bone resorption,” J. Dent. Res. 70, 27–33 (1991). 10.1177/00220345910700010401 [DOI] [PubMed] [Google Scholar]
  • 44. Goulet G., Cooper D., Coombe D., and Zernicke R., “ Influence of cortical canal architecture on lacunocanalicular pore pressure and fluid flow,” Comput. Methods Biomech. Biomed. Eng. 11, 379–387 (2008). 10.1080/10255840701814105 [DOI] [PubMed] [Google Scholar]
  • 45. Yang C.-M. et al. , “ Mechanical strain induces collagenase-3 (MMP-13) expression in MC3T3-E1 osteoblastic cells,” J. Biol. Chem. 279, 22158–22165 (2004). 10.1074/jbc.M401343200 [DOI] [PubMed] [Google Scholar]
  • 46. Liu D. et al. , “ Activation of extracellular-signal regulated kinase (ERK1/2) by fluid shear is Ca2+- and ATP-dependent in MC3T3-E1 osteoblasts,” Bone 42, 644–652 (2008). 10.1016/j.bone.2007.09.058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Boutahar N., Guignandon A., Vico L., and Lafage-Proust M.-H., “ Mechanical strain on osteoblasts activates autophosphorylation of focal adhesion kinase and proline-rich tyrosine kinase 2 tyrosine sites involved in ERK activation,” J. Biol. Chem. 279, 30588–30599 (2004). 10.1074/jbc.M313244200 [DOI] [PubMed] [Google Scholar]
  • 48. D. F. Ward, Jr. et al. , “ Mechanical strain enhances extracellular matrix-induced gene focusing and promotes osteogenic differentiation of human mesenchymal stem cells through an extracellular-related kinase-dependent pathway,” Stem Cells Dev. 16, 467–480 (2007). 10.1089/scd.2007.0034 [DOI] [PubMed] [Google Scholar]
  • 49. You J. et al. , “ Substrate deformation levels associated with routine physical activity are less stimulatory to bone cells relative to loading-induced oscillatory fluid flow,” J. Biomech. Eng. 122, 387–393 (2000). 10.1115/1.1287161 [DOI] [PubMed] [Google Scholar]
  • 50. You J. et al. , “ Osteopontin gene regulation by oscillatory fluid flow via intracellular calcium mobilization and activation of mitogen-activated protein kinase in MC3T3–E1 osteoblasts,” J. Biol. Chem. 276, 13365–13371 (2001). 10.1074/jbc.M009846200 [DOI] [PubMed] [Google Scholar]
  • 51. You L., Cowin S. C., Schaffler M. B., and Weinbaum S., “ A model for strain amplification in the actin cytoskeleton of osteocytes due to fluid drag on pericellular matrix,” J. Biomech. 34, 1375–1386 (2001). 10.1016/S0021-9290(01)00107-5 [DOI] [PubMed] [Google Scholar]
  • 52. Jacobs C. et al. , “ Differential effect of steady versus oscillating flow on bone cells,” J. Biomech. 31, 969–976 (1998). 10.1016/S0021-9290(98)00114-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Malone A. M. et al. , “ The role of actin cytoskeleton in oscillatory fluid flow-induced signaling in MC3T3-E1 osteoblasts,” Am. J. Physiol.-Cell Physiol. 292, C1830–C1836 (2007). 10.1152/ajpcell.00352.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Myers K. A., Rattner J. B., Shrive N. G., and Hart D. A., “ Osteoblast-like cells and fluid flow: Cytoskeleton-dependent shear sensitivity,” Biochem. Biophys. Res. Commun. 364, 214–219 (2007). 10.1016/j.bbrc.2007.09.109 [DOI] [PubMed] [Google Scholar]
  • 55. Chen N. X. et al. , “ Ca2+ regulates fluid shear-induced cytoskeletal reorganization and gene expression in osteoblasts,” Am. J. Physiol.-Cell Physiol. 278, C989–C997 (2000). 10.1152/ajpcell.2000.278.5.C989 [DOI] [PubMed] [Google Scholar]
  • 56. Civitelli R., “ Cell–cell communication in the osteoblast/osteocyte lineage,” Arch. Biochem. Biophys. 473, 188–192 (2008). 10.1016/j.abb.2008.04.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Schirrmacher K. et al. , “ Characterization of gap junctions between osteoblast-like cells in culture,” Calcif. Tissue Int. 51, 285–290 (1992). 10.1007/BF00334489 [DOI] [PubMed] [Google Scholar]
  • 58. Vander Molen M. A., Rubin C. T., McLeod K. J., McCauley L. K., and Donahue H. J., “ Gap junctional intercellular communication contributes to hormonal responsiveness in osteoblastic networks,” J. Biol. Chem. 271, 12165–12171 (1996). 10.1074/jbc.271.21.12165 [DOI] [PubMed] [Google Scholar]
  • 59. Alford A., Jacobs C., and Donahue H., “ Oscillating fluid flow regulates gap junction communication in osteocytic MLO-Y4 cells by an ERK1/2 MAP kinase-dependent mechanism⋆,” Bone 33, 64–70 (2003). 10.1016/S8756-3282(03)00167-4 [DOI] [PubMed] [Google Scholar]
  • 60. Genetos D. C., Kephart C. J., Zhang Y., Yellowley C. E., and Donahue H. J., “ Oscillating fluid flow activation of gap junction hemichannels induces ATP release from MLO‐Y4 osteocytes,” J. Cell. Physiol. 212, 207–214 (2007). 10.1002/jcp.21021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Saunders M. et al. , “ Fluid flow-induced prostaglandin E2 response of osteoblastic ROS 17/2.8 cells is gap junction-mediated and independent of cytosolic calcium,” Bone 32, 350–356 (2003). 10.1016/S8756-3282(03)00025-5 [DOI] [PubMed] [Google Scholar]
  • 62. Jekir M. G. and Donahue H. J., “ Gap junctions and osteoblast-like cell gene expression in response to fluid flow,” J. Biomech. Eng. 131, 011005 (2009). 10.1115/1.3005201 [DOI] [PubMed] [Google Scholar]
  • 63. Poole C. A., “ Articular cartilage chondrons: Form, function and failure,” J. Anat. 191, 1–13 (1997). 10.1046/j.1469-7580.1997.19110001.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Youn I., Choi J., Cao L., Setton L., and Guilak F., “ Zonal variations in the three-dimensional morphology of the chondron measured in situ using confocal microscopy,” Osteoarthritis Cartilage 14, 889–897 (2006). 10.1016/j.joca.2006.02.017 [DOI] [PubMed] [Google Scholar]
  • 65. Melrose J., Hayes A. J., Whitelock J. M., and Little C. B., “ Perlecan, the ‘jack of all trades’ proteoglycan of cartilaginous weight‐bearing connective tissues,” Bioessays 30, 457–469 (2008). 10.1002/bies.20748 [DOI] [PubMed] [Google Scholar]
  • 66. Knudson W., Ishizuka S., Terabe K., Askew E. B., and Knudson C. B., “ The pericellular hyaluronan of articular chondrocytes,” Matrix Biol. 78, 32–46 (2019). 10.1016/j.matbio.2018.02.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Dürr J., Lammi P., Goodman S. L., Aigner T., and von der Mark K., “ Identification and immunolocalization of laminin in cartilage,” Exp. Cell Res. 222, 225–233 (1996). 10.1006/excr.1996.0028 [DOI] [PubMed] [Google Scholar]
  • 68. Chang J., Nakajima H., and Anthony P. C., “ Structural rganization n of type VI collagen and fibronectin in agarose cultured chondrocytes and isolated chondrons extracted from adult canine tibial cartilage,” J. Anat. 190, 523–532 (1997). 10.1046/j.1469-7580.1997.19040523.x [DOI] [Google Scholar]
  • 69. Poole C. A., Ayad S., and Schofield J. R., “ Chondrons from articular cartilage: I. Immunolocalization of type VI collagen in the pericellular capsule of isolated canine tibial chondrons,” J. Cell Sci. 90, 635–643 (1988). 10.1242/jcs.90.4.635 [DOI] [PubMed] [Google Scholar]
  • 70. Kavanagh E. and Ashhurst D. E., “ Development and aging of the articular cartilage of the rabbit knee joint: Distribution of biglycan, decorin, and matrilin-1,” J. Histochem. Cytochem. 47, 1603–1615 (1999). 10.1177/002215549904701212 [DOI] [PubMed] [Google Scholar]
  • 71. Poole C. A., Gilbert R. T., Herbage D., and Hartmann D. J., “ Immunolocalization of type IX collagen in normal and spontaneously osteoarthritic canine tibial cartilage and isolated chondrons,” Osteoarthritis Cartilage 5, 191–204 (1997). 10.1016/S1063-4584(97)80014-3 [DOI] [PubMed] [Google Scholar]
  • 72. Vincent T. L., “ Targeting mechanotransduction pathways in osteoarthritis: A focus on the pericellular matrix,” Curr. Opin. Pharmacol. 13, 449–454 (2013). 10.1016/j.coph.2013.01.010 [DOI] [PubMed] [Google Scholar]
  • 73. Guilak F., Nims R. J., Dicks A., Wu C.-L., and Meulenbelt I., “ Osteoarthritis as a disease of the cartilage pericellular matrix,” Matrix Biol. 71, 40–50 (2018). 10.1016/j.matbio.2018.05.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Guilak F., Jones W. R., Ting-Beall H. P., and Lee G. M., “ The deformation behavior and mechanical properties of chondrocytes in articular cartilage,” Osteoarthritis Cartilage 7, 59–70 (1999). 10.1053/joca.1998.0162 [DOI] [PubMed] [Google Scholar]
  • 75. Darling E., Zauscher S., and Guilak F., “ Viscoelastic properties of zonal articular chondrocytes measured by atomic force microscopy,” Osteoarthritis Cartilage 14, 571–579 (2006). 10.1016/j.joca.2005.12.003 [DOI] [PubMed] [Google Scholar]
  • 76. Schinagl R. M., Gurskis D., Chen A. C., and Sah R. L., “ Depth‐dependent confined compression modulus of full‐thickness bovine articular cartilage,” J. Orthop. Res. 15, 499–506 (1997). 10.1002/jor.1100150404 [DOI] [PubMed] [Google Scholar]
  • 77. Vincent T. L., Hermansson M. A., Hansen U. N., Amis A. A., and Saklatvala J., “ Basic fibroblast growth factor mediates transduction of mechanical signals when articular cartilage is loaded,” Arthritis Rheum. 50, 526–533 (2004). 10.1002/art.20047 [DOI] [PubMed] [Google Scholar]
  • 78. Vincent T., McLean C., Full L., Peston D., and Saklatvala J., “ FGF-2 is bound to perlecan in the pericellular matrix of articular cartilage, where it acts as a chondrocyte mechanotransducer,” Osteoarthritis Cartilage 15, 752–763 (2007). 10.1016/j.joca.2007.01.021 [DOI] [PubMed] [Google Scholar]
  • 79. Meyer C. J. et al. , “ Mechanical control of cyclic AMP signalling and gene transcription through integrins,” Nat. Cell Biol. 2, 666–668 (2000). 10.1038/35023621 [DOI] [PubMed] [Google Scholar]
  • 80. Kumar D. and Lassar A. B., “ The transcriptional activity of Sox9 in chondrocytes is regulated by RhoA signaling and actin polymerization,” Mol. Cell. Biol. 29, 4262–4273 (2009). 10.1128/MCB.01779-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Juhász T. et al. , “ Mechanical loading stimulates chondrogenesis via the PKA/CREB-Sox9 and PP2A pathways in chicken micromass cultures,” Cell. Signalling 26, 468–482 (2014). 10.1016/j.cellsig.2013.12.001 [DOI] [PubMed] [Google Scholar]
  • 82. Wang Y. et al. , “ Strain-induced differentiation of fetal type II epithelial cells is mediated via the integrin α6β1-ADAM17/tumor necrosis factor-α-converting enzyme (TACE) signaling pathway,” J. Biol. Chem. 288, 25646–25657 (2013). 10.1074/jbc.M113.473777 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Zhao L., Li G., and Zhou G. Q., “ SOX9 Directly binds CREB as a novel synergism with the PKA pathway in BMP‐2–induced osteochondrogenic differentiation,” J. Bone Miner. Res. 24, 826–836 (2009). 10.1359/jbmr.081236 [DOI] [PubMed] [Google Scholar]
  • 84. Liang W. et al. , “ Periodic mechanical stress stimulates the FAK mitogenic signal in rat chondrocytes through ERK1/2 activity,” Cell. Physiol. Biochem. 32, 915–930 (2013). 10.1159/000354495 [DOI] [PubMed] [Google Scholar]
  • 85. Lee W. et al. , “ Synergy between Piezo1 and Piezo2 channels confers high-strain mechanosensitivity to articular cartilage,” Proc. Natl. Acad. Sci. 111, E5114–E5122 (2014). 10.1073/pnas.1414298111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. O'Conor C. J., Leddy H. A., Benefield H. C., Liedtke W. B., and Guilak F., “ TRPV4-mediated mechanotransduction regulates the metabolic response of chondrocytes to dynamic loading,” Proc. Natl. Acad. Sci. 111, 1316–1321 (2014). 10.1073/pnas.1319569111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Phan M. N. et al. , “ Functional characterization of TRPV4 as an osmotically sensitive ion channel in porcine articular chondrocytes,” Arthritis Rheum. 60, 3028–3037 (2009). 10.1002/art.24799 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Akasaki Y. et al. , “ Dysregulated FOXO transcription factors in articular cartilage in aging and osteoarthritis,” Osteoarthritis Cartilage 22, 162–170 (2014). 10.1016/j.joca.2013.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Matsuzaki T. et al. , “ FoxO transcription factors modulate autophagy and proteoglycan 4 in cartilage homeostasis and osteoarthritis,” Sci. Transl. Med. 10, eaan0746 (2018). 10.1126/scitranslmed.aan0746 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Lee H.-H. et al. , “ Hypoxia enhances chondrogenesis and prevents terminal differentiation through PI3K/Akt/FoxO dependent anti-apoptotic effect,” Sci. Rep. 3, 2863 (2013). 10.1038/srep02683 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. He Z. et al. , “ Strain-induced mechanotransduction through primary cilia, extracellular ATP, purinergic calcium signaling, and ERK1/2 transactivates CITED2 and downregulates MMP-1 and MMP-13 gene expression in chondrocytes,” Osteoarthritis Cartilage 24, 892–901 (2016). 10.1016/j.joca.2015.11.015 [DOI] [PubMed] [Google Scholar]
  • 92. Xiang W. et al. , “ Primary cilia and autophagy interaction is involved in mechanical stress mediated cartilage development via ERK/mTOR axis,” Life Sci. 218, 308–313 (2019). 10.1016/j.lfs.2019.01.001 [DOI] [PubMed] [Google Scholar]
  • 93. Zhang J. et al. , “ Connexin43 hemichannels mediate small molecule exchange between chondrocytes and matrix in biomechanically-stimulated temporomandibular joint cartilage,” Osteoarthritis Cartilage 22, 822–830 (2014). 10.1016/j.joca.2014.03.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Garcia M. and Knight M. M., “ Cyclic loading opens hemichannels to release ATP as part of a chondrocyte mechanotransduction pathway,” J. Orthop. Res. 28, 510–515 (2010). 10.1002/jor.21025 [DOI] [PubMed] [Google Scholar]
  • 95. Chowdhury T. and Knight M., “ Purinergic pathway suppresses the release of ·NO and stimulates proteoglycan synthesis in chondrocyte/agarose constructs subjected to dynamic compression,” J. Cell. Physiol. 209, 845–853 (2006). 10.1002/jcp.20768 [DOI] [PubMed] [Google Scholar]
  • 96. Bachrach N. M., Mow V. C., and Guilak F., “ Incompressibility of the solid matrix of articular cartilage under high hydrostatic pressures,” J. Biomech. 31, 445–451 (1998). 10.1016/S0021-9290(98)00035-9 [DOI] [PubMed] [Google Scholar]
  • 97. Carter D. R. and Wong M., “ Modelling cartilage mechanobiology,” Philos. Trans. R. Soc. London, Ser. B 358, 1461–1471 (2003). 10.1098/rstb.2003.1346 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Soltz M. A. and Ateshian G. A., “ Interstitial fluid pressurization during confined compression cyclical loading of articular cartilage,” Ann. Biomed. Eng. 28, 150–159 (2000). 10.1114/1.239 [DOI] [PubMed] [Google Scholar]
  • 99. Steward A. et al. , “ Cell–matrix interactions regulate mesenchymal stem cell response to hydrostatic pressure,” Acta Biomater. 8, 2153–2159 (2012). 10.1016/j.actbio.2012.03.016 [DOI] [PubMed] [Google Scholar]
  • 100. Jablonski C. L., Ferguson S., Pozzi A., and Clark A. L., “ Integrin α1β1 participates in chondrocyte transduction of osmotic stress,” Biochem. Biophys. Res. Commun. 445, 184–190 (2014). 10.1016/j.bbrc.2014.01.157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Hall A. C., “ Differential effects of hydrostatic pressure on cation transport pathways of isolated articular chondrocytes,” J. Cell. Physiol. 178, 197–204 (1999). [DOI] [PubMed] [Google Scholar]
  • 102. Browning J., Walker R., Hall A., and Wilkins R., “ Modulation of Na+ × H+ exchange by hydrostatic pressure in isolated bovine articular chondrocytes,” Acta Physiol. Scand. 166, 39–45 (1999). 10.1046/j.1365-201x.1999.00534.x [DOI] [PubMed] [Google Scholar]
  • 103. Mizuno S., “ A novel method for assessing effects of hydrostatic fluid pressure on intracellular calcium: A study with bovine articular chondrocytes,” Am. J. Physiol. 288, C329–C337 (2005). 10.1152/ajpcell.00131.2004 [DOI] [PubMed] [Google Scholar]
  • 104. Knight M., McGlashan S., Garcia M., Jensen C., and Poole C., “ Articular chondrocytes express connexin 43 hemichannels and P2 receptors—A putative mechanoreceptor complex involving the primary cilium?,” J. Anat. 214, 275–283 (2009). 10.1111/j.1469-7580.2008.01021.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Steward A. J., Kelly D. J., and Wagner D. R., “ Purinergic signaling regulates the transforming growth factor-β3-induced chondrogenic response of mesenchymal stem cells to hydrostatic pressure,” Tissue Eng., Part A 22, 831–839 (2016). 10.1089/ten.tea.2015.0047 [DOI] [PubMed] [Google Scholar]
  • 106. Steward A., Kelly D., and Wagner D., “ The role of calcium signalling in the chondrogenic response of mesenchymal stem cells to hydrostatic pressure,” Eur. Cell Mater. 28, 358–371 (2014). 10.22203/eCM.v028a25 [DOI] [PubMed] [Google Scholar]
  • 107. Zhao Y.-H. et al. , “ Hydrostatic pressure promotes the proliferation and osteogenic/chondrogenic differentiation of mesenchymal stem cells: The roles of RhoA and Rac1,” Stem Cell Res. 14, 283–296 (2015). 10.1016/j.scr.2015.02.006 [DOI] [PubMed] [Google Scholar]
  • 108. Zhao Y. et al. , “ The distinct effects of estrogen and hydrostatic pressure on mesenchymal stem cells differentiation: Involvement of estrogen receptor signaling,” Ann. Biomed. Eng. 44, 2971–2983 (2016). 10.1007/s10439-016-1631-5 [DOI] [PubMed] [Google Scholar]
  • 109. Das P., Schurman D., and Smith R. L., “ Nitric oxide and G proteins mediate the response of bovine articular chondrocytes to fluid‐induced shear,” J. Orthop. Res. 15, 87–93 (1997). 10.1002/jor.1100150113 [DOI] [PubMed] [Google Scholar]
  • 110. Mohtai M. et al. , “ Expression of interleukin‐6 in osteoarthritic chondrocytes and effects of fluid‐induced shear on this expression in normal human chondrocytes in vitro,” J. Orthop. Res. 14, 67–73 (1996). 10.1002/jor.1100140112 [DOI] [PubMed] [Google Scholar]
  • 111. Smith R. L. et al. , “ Effects of fluid‐induced shear on articular chondrocyte morphology and metabolism in vitro,” J. Orthop. Res. 13, 824–831 (1995). 10.1002/jor.1100130604 [DOI] [PubMed] [Google Scholar]
  • 112. Suh J.-K. et al. , “ Intermittent sub-ambient interstitial hydrostatic pressure as a potential mechanical stimulator for chondrocyte metabolism,” Osteoarthritis Cartilage 7, 71–80 (1999). 10.1053/joca.1998.0163 [DOI] [PubMed] [Google Scholar]
  • 113. Yeh C.-C. et al. , “ Shear stress modulates macrophage-induced urokinase plasminogen activator expression in human chondrocytes,” Arthritis Res. Ther. 15, R53 (2013). 10.1186/ar4215 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Yeh C. C. et al. , “ Regulation of plasminogen activator inhibitor 1 expression in human osteoarthritic chondrocytes by fluid shear stress: Role of protein kinase Cα,” Arthritis Rheum. 60, 2350–2361 (2009). 10.1002/art.24680 [DOI] [PubMed] [Google Scholar]
  • 115. Yokota H., Goldring M. B., and Sun H. B., “ CITED2-mediated regulation of MMP-1 and MMP-13 in human chondrocytes under flow shear,” J. Biol. Chem. 278, 47275–47280 (2003). 10.1074/jbc.M304652200 [DOI] [PubMed] [Google Scholar]
  • 116. Wang P., Zhu F., Lee N. H., and Konstantopoulos K., “ Shear-induced interleukin-6 synthesis in chondrocytes roles of E prostanoid (EP) 2 and EP3 in cAMP/protein kinase A-and PI3-K/Akt-dependent NF-κB activation,” J. Biol. Chem. 285, 24793–24804 (2010). 10.1074/jbc.M110.110320 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Guan P.-P. et al. , “ The role of cyclooxygenase-2, interleukin-1β and fibroblast growth factor-2 in the activation of matrix metalloproteinase-1 in sheared-chondrocytes and articular cartilage,” Sci. Rep. 5, 10412 (2015). 10.1038/srep10412 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 118. Zhu F., Wang P., Lee N. H., Goldring M. B., and Konstantopoulos K., “ Prolonged application of high fluid shear to chondrocytes recapitulates gene expression profiles associated with osteoarthritis,” PloS One 5, 015174 (2010). 10.1371/journal.pone.0015174 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Berbari N. F., O'Connor A. K., Haycraft C. J., and Yoder B. K., “ The primary cilium as a complex signaling center,” Curr. Biol. 19, R526–R535 (2009). 10.1016/j.cub.2009.05.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. McGlashan S. R., Jensen C. G., and Poole C. A., “ Localization of extracellular matrix receptors on the chondrocyte primary cilium,” J. Histochem. Cytochem. 54, 1005–1014 (2006). 10.1369/jhc.5A6866.2006 [DOI] [PubMed] [Google Scholar]
  • 121. Ruhlen R. and Marberry K., “ The chondrocyte primary cilium,” Osteoarthritis Cartilage 22, 1071–1076 (2014). 10.1016/j.joca.2014.05.011 [DOI] [PubMed] [Google Scholar]
  • 122. Fitzgerald J. B. et al. , “ Shear-and compression-induced chondrocyte transcription requires MAPK activation in cartilage explants,” J. Biol. Chem. 283, 6735–6743 (2008). 10.1074/jbc.M708670200 [DOI] [PubMed] [Google Scholar]
  • 123. Wang T., Wen C.-Y., Yan C.-H., Lu W.-W., and Chiu K.-Y., “ Spatial and temporal changes of subchondral bone proceed to microscopic articular cartilage degeneration in guinea pigs with spontaneous osteoarthritis,” Osteoarthritis Cartilage 21, 574–581 (2013). 10.1016/j.joca.2013.01.002 [DOI] [PubMed] [Google Scholar]
  • 124. Hügle T. and Geurts J., “ What drives osteoarthritis?—Synovial versus subchondral bone pathology,” Rheumatology 56, 1461–1471 (2017). 10.1093/rheumatology/kew389 [DOI] [PubMed] [Google Scholar]
  • 125. Botter S. M. et al. , “ Osteoarthritis induction leads to early and temporal subchondral plate porosity in the tibial plateau of mice: An in vivo microfocal computed tomography study,” Arthritis Rheum. 63, 2690–2699 (2011). 10.1002/art.30307 [DOI] [PubMed] [Google Scholar]
  • 126. Aizah N., Chong P. P., and Kamarul T., “ Early alterations of subchondral bone in the rat anterior cruciate ligament transection model of osteoarthritis,” Cartilage 2019, 1947603519878479. 10.1177/1947603519878479 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Arden N., Griffiths G. O., Hart D., Doyle D., and Spector T., “ The association between osteoarthritis and osteoporotic fracture: The Chingford study,” Rheumatology 35, 1299–1304 (1996). 10.1093/rheumatology/35.12.1299 [DOI] [PubMed] [Google Scholar]
  • 128. Hannan M. T., Anderson J. J., Zhang Y., Levy D., and Felson D. T., “ Bone mineral density and knee osteoarthritis in elderly men and women. The Framingham study,” Arthritis Rheum. 36, 1671–1680 (1993). 10.1002/art.1780361205 [DOI] [PubMed] [Google Scholar]
  • 129. Radin E., Paul I., and Rose R., “ Role of mechanical factors in pathogenesis of primary osteoarthritis,” Lancet 299, 519–522 (1972). 10.1016/S0140-6736(72)90179-1 [DOI] [PubMed] [Google Scholar]
  • 130. Radin E. L. and Rose R. M., “ Role of subchondral bone in the initiation and progression of cartilage damage,” Clin. Orthop. Relat. Res. 213, 34–40 (1986). 10.1097/00003086-198612000-00005 [DOI] [PubMed] [Google Scholar]
  • 131. Duncan H., Jundt J., Riddle J., Pitchford W., and Christopherson T., “ The tibial subchondral plate. A scanning electron microscopic study,” J. Bone Jt. Surg. 69, 1212–1220 (1987). 10.2106/00004623-198769080-00015 [DOI] [PubMed] [Google Scholar]
  • 132. Clark J. and Huber J., “ The structure of the human subchondral plate,” J. Bone Jt. Surg. 72, 866–873 (1990). 10.1302/0301-620X.72B5.2211774 [DOI] [PubMed] [Google Scholar]
  • 133. Lyons T. J., McClure S. F., Stoddart R. W., and McClure J., “ The normal human chondro-osseous junctional region: Evidence for contact of uncalcified cartilage with subchondral bone and marrow spaces,” BMC Musculoskeletal Disord. 7, 52 (2006). 10.1186/1471-2474-7-52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Pan J. et al. , “ Elevated cross-talk between subchondral bone and cartilage in osteoarthritic joints,” Bone 51, 212–217 (2012). 10.1016/j.bone.2011.11.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Goldring S. R., “ Alterations in periarticular bone and cross talk between subchondral bone and articular cartilage in osteoarthritis,” Ther. Adv. Musculoskeletal Dis. 4, 249–258 (2012). 10.1177/1759720X12437353 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Lu J. et al. , “ Positive‐feedback regulation of subchondral H‐type vessel formation by chondrocyte promotes osteoarthritis development in mice,” J. Bone Miner. Res. 33, 909–920 (2018). 10.1002/jbmr.3388 [DOI] [PubMed] [Google Scholar]
  • 137. Bayar A. et al. , “ Regional bone density changes in anterior cruciate ligament deficient knees: A DEXA study,” Knee 15, 373–377 (2008). 10.1016/j.knee.2008.05.005 [DOI] [PubMed] [Google Scholar]
  • 138. Van Meer B. et al. , “ Bone mineral density changes in the knee following anterior cruciate ligament rupture,” Osteoarthritis Cartilage 22, 154–161 (2014). 10.1016/j.joca.2013.11.005 [DOI] [PubMed] [Google Scholar]
  • 139. Kroker A. et al. , “ Longitudinal effects of acute anterior cruciate ligament tears on peri‐articular bone in human knees within the first year of injury,” J. Orthop. Res. 37, 2325–2336 (2019). 10.1002/jor.24410 [DOI] [PubMed] [Google Scholar]
  • 140. Mansell J. P., Collins C., and Bailey A. J., “ Bone, not cartilage, should be the major focus in osteoarthritis,” Nat. Clin. Pract. Rheumatol. 3, 306–307 (2007). 10.1038/ncprheum0505 [DOI] [PubMed] [Google Scholar]
  • 141. Mori S., Harruff R., and Burr D., “ Microcracks in articular calcified cartilage of human femoral heads,” Arch. Pathol. Lab. Med. 117, 196–198 (1993). [PubMed] [Google Scholar]
  • 142. Sokoloff L., “ Microcracks in the calcified layer of articular cartilage,” Arch. Pathol. Lab. Med. 117, 191 (1993). [PubMed] [Google Scholar]
  • 143. Kennedy O. D. et al. , “ Activation of resorption in fatigue-loaded bone involves both apoptosis and active pro-osteoclastogenic signaling by distinct osteocyte populations,” Bone 50, 1115–1122 (2012). 10.1016/j.bone.2012.01.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Bellido M. et al. , “ Subchondral bone microstructural damage by increased remodelling aggravates experimental osteoarthritis preceded by osteoporosis,” Arthritis Res. Ther. 12, R152 (2010). 10.1186/ar3103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Couchourel D. et al. , “ Altered mineralization of human osteoarthritic osteoblasts is attributable to abnormal type I collagen production,” Arthritis Rheum. 60, 1438–1450 (2009). 10.1002/art.24489 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Chan T. F. et al. , “ Elevated Dickkopf‐2 levels contribute to the abnormal phenotype of human osteoarthritic osteoblasts,” J. Bone Miner. Res. 26, 1399–1410 (2011). 10.1002/jbmr.358 [DOI] [PubMed] [Google Scholar]
  • 147. Li X. et al. , “ Dkk2 has a role in terminal osteoblast differentiation and mineralized matrix formation,” Nat. Genet. 37, 945–952 (2005). 10.1038/ng1614 [DOI] [PubMed] [Google Scholar]
  • 148. Mansell J. P. and Bailey A. J., “ Abnormal cancellous bone collagen metabolism in osteoarthritis,” J. Clin. Invest. 101, 1596–1603 (1998). 10.1172/JCI867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Li B. and Aspden R. M., “ Composition and mechanical properties of cancellous bone from the femoral head of patients with osteoporosis or osteoarthritis,” J. Bone Miner. Res. 12, 641–651 (1997). 10.1359/jbmr.1997.12.4.641 [DOI] [PubMed] [Google Scholar]
  • 150. Coats A. M., Zioupos P., and Aspden R. M., “ Material properties of subchondral bone from patients with osteoporosis or osteoarthritis by microindentation testing and electron probe microanalysis,” Calcif. Tissue Int. 73, 66–71 (2003). 10.1007/s00223-002-2080-8 [DOI] [PubMed] [Google Scholar]
  • 151. Behets C., Williams J. M., Chappard D., Devogelaer J. P., and Manicourt D. H., “ Effects of calcitonin on subchondral trabecular bone changes and on osteoarthritic cartilage lesions after acute anterior cruciate ligament deficiency,” J. Bone Miner. Res. 19, 1821–1826 (2004). 10.1359/JBMR.040609 [DOI] [PubMed] [Google Scholar]
  • 152. Muehleman C. et al. , “ The effect of bone remodeling inhibition by zoledronic acid in an animal model of cartilage matrix damage,” Osteoarthritis Cartilage 10, 226–233 (2002). 10.1053/joca.2001.0506 [DOI] [PubMed] [Google Scholar]
  • 153. Hayami T. et al. , “ The role of subchondral bone remodeling in osteoarthritis: Reduction of cartilage degeneration and prevention of osteophyte formation by alendronate in the rat anterior cruciate ligament transection model,” Arthritis Rheum. 50, 1193–1206 (2004). 10.1002/art.20124 [DOI] [PubMed] [Google Scholar]
  • 154. Siebelt M. et al. , “ Inhibited osteoclastic bone resorption through alendronate treatment in rats reduces severe osteoarthritis progression,” Bone 66, 163–170 (2014). 10.1016/j.bone.2014.06.009 [DOI] [PubMed] [Google Scholar]
  • 155. Spector T. D. et al. , “ Effect of risedronate on joint structure and symptoms of knee osteoarthritis: Results of the BRISK randomized, controlled trial [ISRCTN01928173],” Arthritis Res. Ther. 7, R625 (2005). 10.1186/ar1716 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Bingham C. O. III et al. , “ Risedronate decreases biochemical markers of cartilage degradation but does not decrease symptoms or slow radiographic progression in patients with medial compartment osteoarthritis of the knee: Results of the two‐year multinational knee osteoarthritis structural arthritis study,” Arthritis Rheum. 54, 3494–3507 (2006). 10.1002/art.22160 [DOI] [PubMed] [Google Scholar]
  • 157. Russell R., Watts N., Ebetino F., and Rogers M., “ Mechanisms of action of bisphosphonates: Similarities and differences and their potential influence on clinical efficacy,” Osteoporosis Int. 19, 733–759 (2008). 10.1007/s00198-007-0540-8 [DOI] [PubMed] [Google Scholar]
  • 158. Wong S. et al. , “ The pathogenesis of osteoarthritis of the hip. Evidence for primary osteocyte death,” Clin. Orthop. Relat. Res. 214, 305–312 (1987). 10.1097/00003086-198701000-00042 [DOI] [PubMed] [Google Scholar]
  • 159. Jaiprakash A. et al. , “ Phenotypic characterization of osteoarthritic osteocytes from the sclerotic zones: A possible pathological role in subchondral bone sclerosis,” Int. J. Biol. Sci. 8, 406 (2012). 10.7150/ijbs.4221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. van Hove R. P. et al. , “ Osteocyte morphology in human tibiae of different bone pathologies with different bone mineral density—Is there a role for mechanosensing?,” Bone 45, 321–329 (2009). 10.1016/j.bone.2009.04.238 [DOI] [PubMed] [Google Scholar]
  • 161. Laugesen A., Højfeldt J. W., and Helin K., “ Molecular mechanisms directing PRC2 recruitment and H3K27 methylation,” Mol. Cell 74, 8–18 (2019). 10.1016/j.molcel.2019.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Tang S. Y., Herber R. P., Ho S. P., and Alliston T., “ Matrix metalloproteinase–13 is required for osteocytic perilacunar remodeling and maintains bone fracture resistance,” J. Bone Miner. Res. 27, 1936–1950 (2012). 10.1002/jbmr.1646 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Inoue K. et al. , “ A crucial role for matrix metalloproteinase 2 in osteocytic canalicular formation and bone metabolism,” J. Biol. Chem. 281, 33814–33824 (2006). 10.1074/jbc.M607290200 [DOI] [PubMed] [Google Scholar]
  • 164. Holmbeck K. et al. , “ The metalloproteinase MT1-MMP is required for normal development and maintenance of osteocyte processes in bone,” J. Cell Sci. 118, 147–156 (2005). 10.1242/jcs.01581 [DOI] [PubMed] [Google Scholar]
  • 165. Teti A. and Zallone A., “ Do osteocytes contribute to bone mineral homeostasis? Osteocytic osteolysis revisited,” Bone 44, 11–16 (2009). 10.1016/j.bone.2008.09.017 [DOI] [PubMed] [Google Scholar]
  • 166. Glyn-Jones S. et al. , “ Osteoarthritis,” Lancet 386, 376–387 (2015). 10.1016/S0140-6736(14)60802-3 [DOI] [PubMed] [Google Scholar]
  • 167. Wu J. et al. , “ Comparative proteomic characterization of articular cartilage tissue from normal donors and patients with osteoarthritis,” Arthritis Rheum. 56, 3675–3684 (2007). 10.1002/art.22876 [DOI] [PubMed] [Google Scholar]
  • 168. Chery D. R. et al. , “ Early changes in cartilage pericellular matrix micromechanobiology portend the onset of post-traumatic osteoarthritis,” Acta Biomater. 111, 267–278 (2020). 10.1016/j.actbio.2020.05.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Wilusz R. E., Zauscher S., and Guilak F., “ Micromechanical mapping of early osteoarthritic changes in the pericellular matrix of human articular cartilage,” Osteoarthritis Cartilage 21, 1895–1903 (2013). 10.1016/j.joca.2013.08.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Zelenski N. A. et al. , “ Type VI collagen regulates pericellular matrix properties, chondrocyte swelling, and mechanotransduction in mouse articular cartilage,” Arthritis Rheumatol. 67, 1286–1294 (2015). 10.1002/art.39034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Danalache M. et al. , “ Changes in stiffness and biochemical composition of the pericellular matrix as a function of spatial chondrocyte rganization in osteoarthritic cartilage,” Osteoarthritis Cartilage 27, 823–832 (2019). 10.1016/j.joca.2019.01.008 [DOI] [Google Scholar]
  • 172. Aicher W. K. and Rolauffs B., “ The spatial rganization of joint surface chondrocytes: Review of its potential roles in tissue functioning, disease and early, preclinical diagnosis of osteoarthritis,” Ann. Rheum. Dis. 73, 645–653 (2014). 10.1136/annrheumdis-2013-204308 [DOI] [Google Scholar]
  • 173. Rolauffs B. et al. , “ Proliferative remodeling of the spatial organization of human superficial chondrocytes distant from focal early osteoarthritis,” Arthritis Rheum. 62, 489–498 (2010). 10.1002/art.27217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Felka T. et al. , “ Loss of spatial organization and destruction of the pericellular matrix in early osteoarthritis in vivo and in a novel in vitro methodology,” Osteoarthritis Cartilage 24, 1200–1209 (2016). 10.1016/j.joca.2016.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Holloway I. et al. , “ Increased presence of cells with multiple elongated processes in osteoarthritic femoral head cartilage,” Osteoarthritis Cartilage 12, 17–24 (2004). 10.1016/j.joca.2003.09.001 [DOI] [PubMed] [Google Scholar]
  • 176. Murray D. H., Bush P. G., Brenkel I. J., and Hall A. C., “ Abnormal human chondrocyte morphology is related to increased levels of cell‐associated IL‐1β and disruption to pericellular collagen type VI,” J. Orthop. Res. 28, 1507–1514 (2010). 10.1002/jor.21155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Polur I., Lee P. L., Servais J. M., Xu L., and Li Y., “ Role of HTRA1, a serine protease, in the progression of articular cartilage degeneration,” Histol. Histopathol. 25, 599 (2010). 10.14670/HH-25.599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Wilusz R. E., Sanchez-Adams J., and Guilak F., “ The structure and function of the pericellular matrix of articular cartilage,” Matrix Biol. 39, 25–32 (2014). 10.1016/j.matbio.2014.08.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Xu L. et al. , “ Activation of the discoidin domain receptor 2 induces expression of matrix metalloproteinase 13 associated with osteoarthritis in mice,” J. Biol. Chem. 280, 548–555 (2005). 10.1074/jbc.M411036200 [DOI] [PubMed] [Google Scholar]
  • 180. Holt D. et al. , “ Osteoarthritis-like changes in the heterozygous sedc mouse associated with the HtrA1–Ddr2–Mmp-13 degradative pathway: A new model of osteoarthritis,” Osteoarthritis Cartilage 20, 430–439 (2012). 10.1016/j.joca.2011.11.008 [DOI] [PubMed] [Google Scholar]
  • 181. Du G. et al. , “ Roles of TRPV4 and piezo channels in stretch-evoked Ca2+ response in chondrocytes,” Exp. Biol. Med. 245, 180–189 (2020). 10.1177/1535370219892601 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Lee W., Guilak F., and Liedtke W., Current Topics in Membranes ( Elsevier, 2017), Vol. 79, pp. 263–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Xu B. et al. , “ Excessive mechanical stress induces chondrocyte apoptosis through TRPV4 in an anterior cruciate ligament-transected rat osteoarthritis model,” Life Sci. 228, 158–166 (2019). 10.1016/j.lfs.2019.05.003 [DOI] [PubMed] [Google Scholar]
  • 184. Ismail H. M. et al. , “ Interleukin‐1 acts via the JNK‐2 signaling pathway to induce aggrecan degradation by human chondrocytes,” Arthritis Rheumatol. 67, 1826–1836 (2015). 10.1002/art.39099 [DOI] [PubMed] [Google Scholar]
  • 185. Ismail H. M. et al. , “ Brief report: JNK‐2 controls aggrecan degradation in murine articular cartilage and the development of experimental osteoarthritis,” Arthritis Rheumatol. 68, 1165–1171 (2016). 10.1002/art.39547 [DOI] [PubMed] [Google Scholar]
  • 186. Lima E. G. et al. , “ The beneficial effect of delayed compressive loading on tissue-engineered cartilage constructs cultured with TGF-β3,” Osteoarthritis Cartilage 15, 1025–1033 (2007). 10.1016/j.joca.2007.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Mauck R. L., Wang C. C. B., Oswald E. S., Ateshian G. A., and Hung C. T., “ The role of cell seeding density and nutrient supply for articular cartilage tissue engineering with deformational loading,” Osteoarthritis Cartilage 11, 879–890 (2003). 10.1016/j.joca.2003.08.006 [DOI] [PubMed] [Google Scholar]
  • 188. Waldman S. D., Spiteri C. G., Grynpas M. D., Pilliar R. M., and Kandel R. A., “ Long-term intermittent shear deformation improves the quality of cartilaginous tissue formed in vitro,” J. Orthop. Res. 21, 590–596 (2003). 10.1016/S0736-0266(03)00009-3 [DOI] [PubMed] [Google Scholar]
  • 189. De Croos J. N. A., Dhaliwal S. S., Grynpas M. D., Pilliar R. M., and Kandel R. A., “ Cyclic compressive mechanical stimulation induces sequential catabolic and anabolic gene changes in chondrocytes resulting in increased extracellular matrix accumulation,” Matrix Biol. 25, 323–331 (2006). 10.1016/j.matbio.2006.03.005 [DOI] [PubMed] [Google Scholar]
  • 190. Nam J., Aguda B. D., Rath B., and Agarwal S., “ Biomechanical thresholds regulate inflammation through the NF-κB pathway: Experiments and modeling,” PloS One 4, e5262 (2009). 10.1371/journal.pone.0005262 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Sylvester J., El Mabrouk M., Ahmad R., Chaudry A., and Zafarullah M., “ Interleukin-1 induction of aggrecanase gene expression in human articular chondrocytes is mediated by mitogen-activated protein kinases,” Cell. Physiol. Biochem. 30, 563–574 (2012). 10.1159/000341438 [DOI] [PubMed] [Google Scholar]
  • 192. Novakofski K. D., Torre C. J., and Fortier L. A., “ Interleukin-1α, -6, and -8 decrease Cdc42 activity resulting in loss of articular chondrocyte phenotype,” J. Orthop. Res. 30, 246–251 (2012). 10.1002/jor.21515 [DOI] [PubMed] [Google Scholar]
  • 193. Nicholson I. P., Gault E. A., Foote C. G., Nasir L., and Bennett D., “ Human telomerase reverse transcriptase (hTERT) extends the lifespan of canine chondrocytes in vitro without inducing neoplastic transformation,” Vet. J. 174, 570–576 (2007). 10.1016/j.tvjl.2007.07.009 [DOI] [PubMed] [Google Scholar]
  • 194. Das R. H. J. et al. , “ In vitro expansion affects the response of chondrocytes to mechanical stimulation,” Osteoarthritis Cartilage 16, 385–391 (2008). 10.1016/j.joca.2007.07.014 [DOI] [PubMed] [Google Scholar]
  • 195. Huang J., Ballou L. R., and Hasty K. A., “ Cyclic equibiaxial tensile strain induces both anabolic and catabolic responses in articular chondrocytes,” Gene 404, 101–109 (2007). 10.1016/j.gene.2007.09.007 [DOI] [PubMed] [Google Scholar]
  • 196. Honda K. et al. , “ The effects of high magnitude cyclic tensile load on cartilage matrix metabolism in cultured chondrocytes,” Eur. J. Cell Biol. 79, 601–609 (2000). 10.1078/0171-9335-00089 [DOI] [PubMed] [Google Scholar]
  • 197. Chang S. H. et al. , “ Excessive mechanical loading promotes osteoarthritis through the gremlin-1–NF-κB pathway,” Nat. Commun. 10, 1442 (2019). 10.1038/s41467-019-09491-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Plotkin L. I. et al. , “ Mechanical stimulation prevents osteocyte apoptosis: Requirement of integrins, Src kinases, and ERKs,” Am. J. Physiol.-Cell Physiol. 289, C633–C643 (2005). 10.1152/ajpcell.00278.2004 [DOI] [PubMed] [Google Scholar]
  • 199. He Y.-B. et al. , “ Mechanical stretch promotes the osteogenic differentiation of bone mesenchymal stem cells induced by erythropoietin,” Stem Cells Int. 2019, 1839627. 10.1155/2019/1839627 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Zeng Z., Yin X., Zhang X., Jing D., and Feng X., “ Cyclic stretch enhances bone morphogenetic protein-2-induced osteoblastic differentiation through the inhibition of Hey1,” Int. J. Mol. Med. 36, 1273–1281 (2015). 10.3892/ijmm.2015.2354 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Wang D. et al. , “ The interactions between mTOR and NF‐κB: A novel mechanism mediating mechanical stretch‐stimulated osteoblast differentiation,” J. Cell. Physiol. 236, 4592 (2020). 10.1002/jcp.30184 [DOI] [PubMed] [Google Scholar]
  • 202. Kurata K. et al. , “ Mechanical strain effect on bone‐resorbing activity and messenger RNA expressions of marker enzymes in isolated osteoclast culture,” J. Bone Miner. Res. 16, 722–730 (2001). 10.1359/jbmr.2001.16.4.722 [DOI] [PubMed] [Google Scholar]
  • 203. Matsui T. S., Wu H., and Deguchi S., “ Deformable 96-well cell culture plate compatible with high-throughput screening platforms,” PloS One 13, e0203448 (2018). 10.1371/journal.pone.0203448 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Sherman S. A. et al. , “ Stretch injury of human induced pluripotent stem cell derived neurons in a 96 well format,” Sci. Rep. 6, 34097 (2016). 10.1038/srep34097 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Lee M. S. et al. , “ Effects of shear stress on nitric oxide and matrix protein gene expression in human osteoarthritic chondrocytes in vitro,” J. Orthop. Res. 20, 556–561 (2002). 10.1016/S0736-0266(01)00149-8 [DOI] [PubMed] [Google Scholar]
  • 206. Malaviya P. and Nerem R. M., “ Fluid-induced shear stress stimulates chondrocyte proliferation partially mediated via TGF-β1,” Tissue Eng. 8, 581–590 (2002). 10.1089/107632702760240508 [DOI] [PubMed] [Google Scholar]
  • 207. Wang P., Zhu F., Tong Z., and Konstantopoulos K., “ Response of chondrocytes to shear stress: Antagonistic effects of the binding partners Toll-like receptor 4 and caveolin-1,” FASEB J. 25, 3401–3415 (2011). 10.1096/fj.11-184861 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Jaasma M. J. and O'brien F. J., “ Mechanical stimulation of osteoblasts using steady and dynamic fluid flow,” Tissue Eng., Part A 14, 1213–1223 (2008). 10.1089/ten.tea.2007.0321 [DOI] [PubMed] [Google Scholar]
  • 209. McCoy R. J. and O'Brien F. J., “ Influence of shear stress in perfusion bioreactor cultures for the development of three-dimensional bone tissue constructs: A review,” Tissue Eng., Part B 16, 587–601 (2010). 10.1089/ten.teb.2010.0370 [DOI] [PubMed] [Google Scholar]
  • 210. Li Y. J. et al. , “ Oscillatory fluid flow affects human marrow stromal cell proliferation and differentiation,” J. Orthop. Res. 22, 1283–1289 (2004). 10.1016/j.orthres.2004.04.002 [DOI] [PubMed] [Google Scholar]
  • 211. Kim C. H., You L., Yellowley C. E., and Jacobs C. R., “ Oscillatory fluid flow-induced shear stress decreases osteoclastogenesis through RANKL and OPG signaling,” Bone 39, 1043–1047 (2006). 10.1016/j.bone.2006.05.017 [DOI] [PubMed] [Google Scholar]
  • 212. Li J., Rose E., Frances D., Sun Y., and You L., “ Effect of oscillating fluid flow stimulation on osteocyte mRNA expression,” J. Biomech. 45, 247–251 (2012). 10.1016/j.jbiomech.2011.10.037 [DOI] [PubMed] [Google Scholar]
  • 213. Govey P. M. et al. , “ Integrative transcriptomic and proteomic analysis of osteocytic cells exposed to fluid flow reveals novel mechano-sensitive signaling pathways,” J. Biomech. 47, 1838–1845 (2014). 10.1016/j.jbiomech.2014.03.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Lu X. L., Huo B., Park M., and Guo X. E., “ Calcium response in osteocytic networks under steady and oscillatory fluid flow,” Bone 51, 466–473 (2012). 10.1016/j.bone.2012.05.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Ponik S. M., Triplett J. W., and Pavalko F. M., “ Osteoblasts and osteocytes respond differently to oscillatory and unidirectional fluid flow profiles,” J. Cell. Biochem. 100, 794–807 (2007). 10.1002/jcb.21089 [DOI] [PubMed] [Google Scholar]
  • 216. Ikenoue T. et al. , “ Mechanoregulation of human articular chondrocyte aggrecan and type II collagen expression by intermittent hydrostatic pressure in vitro,” J. Orthop. Res. 21, 110–116 (2003). 10.1016/S0736-0266(02)00091-8 [DOI] [PubMed] [Google Scholar]
  • 217. Smith R. L. et al. , “ Effects of intermittent hydrostatic pressure and BMP-2 on osteoarthritic human chondrocyte metabolism in vitro,” J. Orthop. Res. 29, 361–368 (2011). 10.1002/jor.21250 [DOI] [PubMed] [Google Scholar]
  • 218. Inoue H. et al. , “ Hydrostatic pressure influences HIF-2 alpha expression in chondrocytes,” Int. J. Mol. Sci. 16, 1043–1050 (2015). 10.3390/ijms16011043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Le L. et al. , “ The role of microRNA-3085 in chondrocyte function,” Sci. Rep. 10, 21923 (2020). 10.1038/s41598-020-78606-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Cheleschi S. et al. , “ Hydrostatic pressure regulates microRNA expression levels in osteoarthritic chondrocyte cultures via the Wnt/β-catenin pathway,” Int. J. Mol. Sci. 18, 133 (2017). 10.3390/ijms18010133 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221. Stavenschi E., Corrigan M. A., Johnson G. P., Riffault M., and Hoey D. A., “ Physiological cyclic hydrostatic pressure induces osteogenic lineage commitment of human bone marrow stem cells: A systematic study,” Stem Cell Res. Ther. 9, 276–276 (2018). 10.1186/s13287-018-1025-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Gardinier J. D., Majumdar S., Duncan R. L., and Wang L., “ Cyclic hydraulic pressure and fluid flow differentially modulate cytoskeleton re-organization in MC3T3 osteoblasts,” Cell. Mol. Bioeng. 2, 133–143 (2009). 10.1007/s12195-008-0038-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Little C. B. and Hunter D. J., “ Post-traumatic osteoarthritis: From mouse models to clinical trials,” Nat. Rev. Rheumatol. 9, 485–497 (2013). 10.1038/nrrheum.2013.72 [DOI] [PubMed] [Google Scholar]
  • 224. Aung A., Gupta G., Majid G., and Varghese S., “ Osteoarthritic chondrocyte–secreted morphogens induce chondrogenic differentiation of human mesenchymal stem cells,” Arthritis Rheum. 63, 148–158 (2011). 10.1002/art.30086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Tang X., Sheng L., Xie F., and Zhang Q., “ Differentiation of bone marrow-derived mesenchymal stem cells into chondrocytes using chondrocyte extract,” Mol. Med. Rep. 6, 745–749 (2012). 10.3892/mmr.2012.996 [DOI] [PubMed] [Google Scholar]
  • 226. de Windt T. S. et al. , “ Direct cell–cell contact with chondrocytes is a key mechanism in multipotent mesenchymal stromal cell-mediated chondrogenesis,” Tissue Eng., Part A 21, 2536–2547 (2015). 10.1089/ten.tea.2014.0673 [DOI] [PubMed] [Google Scholar]
  • 227. Ouyang X., Xie Y., and Wang G., “ Mechanical stimulation promotes the proliferation and the cartilage phenotype of mesenchymal stem cells and chondrocytes co-cultured in vitro,” Biomed. Pharmacother. 117, 109146 (2019). 10.1016/j.biopha.2019.109146 [DOI] [PubMed] [Google Scholar]
  • 228. Carvalho M. R., Reis R. L., and Oliveira J. M., “ Mimicking the 3D biology of osteochondral tissue with microfluidic-based solutions: Breakthroughs towards boosting drug testing and discovery,” Drug Discovery Today 23, 711–718 (2018). 10.1016/j.drudis.2018.01.008 [DOI] [PubMed] [Google Scholar]
  • 229. Middleton K., Al-Dujaili S., Mei X., Günther A., and You L., “ Microfluidic co-culture platform for investigating osteocyte-osteoclast signalling during fluid shear stress mechanostimulation,” J. Biomech. 59, 35–42 (2017). 10.1016/j.jbiomech.2017.05.012 [DOI] [PubMed] [Google Scholar]
  • 230. Filardo G. et al. , “ Scaffolds for knee chondral and osteochondral defects: Indications for different clinical scenarios. A consensus statement,” Cartilage 2020, 1947603519894729. 10.1177/1947603519894729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Meinert C., Schrobback K., Hutmacher D. W., and Klein T. J., “ A novel bioreactor system for biaxial mechanical loading enhances the properties of tissue-engineered human cartilage,” Sci. Rep. 7, 16997 (2017). 10.1038/s41598-017-16523-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Levett P. A. et al. , “ A biomimetic extracellular matrix for cartilage tissue engineering centered on photocurable gelatin, hyaluronic acid and chondroitin sulfate,” Acta Biomater. 10, 214–223 (2014). 10.1016/j.actbio.2013.10.005 [DOI] [PubMed] [Google Scholar]
  • 233. Liu E., Zhu D., Diaz E. G., Tong X., and Yang F., “ Gradient hydrogels for optimizing niche cues to enhance cell-based cartilage regeneration,” Tissue Eng., Part A 27, 929 (2020). 10.1089/ten.tea.2020.0158 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Zhu D., Tong X., Trinh P., and Yang F., “ Mimicking cartilage tissue zonal organization by engineering tissue-scale gradient hydrogels as 3D cell niche,” Tissue Eng., Part A 24, 1–10 (2018). 10.1089/ten.tea.2016.0453 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Wang T. and Yang F., “ A comparative study of chondroitin sulfate and heparan sulfate for directing three-dimensional chondrogenesis of mesenchymal stem cells,” Stem Cell Res. Ther. 8, —284 (2017). 10.1186/s13287-017-0728-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Lai J. H., Kajiyama G., Smith R. L., Maloney W., and Yang F., “ Stem cells catalyze cartilage formation by neonatal articular chondrocytes in 3D biomimetic hydrogels,” Sci. Rep. 3, 3553 (2013). 10.1038/srep03553 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Xu G. et al. , “ Electric field-driven building blocks for introducing multiple gradients to hydrogels,” Protein Cell 11, 267—285 (2020). 10.1007/s13238-020-00692-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Engler A. J., Sen S., Sweeney H. L., and Discher D. E., “ Matrix elasticity directs stem cell lineage specification,” Cell 126, 677–689 (2006). 10.1016/j.cell.2006.06.044 [DOI] [PubMed] [Google Scholar]
  • 239. Lin H. et al. , “ Optimization of photocrosslinked gelatin/hyaluronic acid hybrid scaffold for the repair of cartilage defect,” J. Tissue Eng. Regener. Med. 13, 1418–1429 (2019). 10.1002/term.2883 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Ding Z. et al. , “ Nanoscale silk–hydroxyapatite hydrogels for injectable bone biomaterials,” ACS Appl. Mater. Interfaces 9, 16913–16921 (2017). 10.1021/acsami.7b03932 [DOI] [PubMed] [Google Scholar]
  • 241. Moeinzadeh S., Shariati S. R. P., and Jabbari E., “ Comparative effect of physicomechanical and biomolecular cues on zone-specific chondrogenic differentiation of mesenchymal stem cells,” Biomaterials 92, 57–70 (2016). 10.1016/j.biomaterials.2016.03.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Stüdle C. et al. , “ Spatially confined induction of endochondral ossification by functionalized hydrogels for ectopic engineering of osteochondral tissues,” Biomaterials 171, 219–229 (2018). 10.1016/j.biomaterials.2018.04.025 [DOI] [PubMed] [Google Scholar]
  • 243. Nguyen L. H., Kudva A. K., Saxena N. S., and Roy K., “ Engineering articular cartilage with spatially-varying matrix composition and mechanical properties from a single stem cell population using a multi-layered hydrogel,” Biomaterials 32, 6946–6952 (2011). 10.1016/j.biomaterials.2011.06.014 [DOI] [PubMed] [Google Scholar]
  • 244. Zhang B., Huang J., and Narayan R. J., “ Gradient scaffolds for osteochondral tissue engineering and regeneration,” J. Mater. Chem. B 8, 8149–8170 (2020). 10.1039/D0TB00688B [DOI] [PubMed] [Google Scholar]
  • 245. Lee H.-P., Gu L., Mooney D. J., Levenston M. E., and Chaudhuri O., “ Mechanical confinement regulates cartilage matrix formation by chondrocytes,” Nat. Mater. 16, 1243–1251 (2017). 10.1038/nmat4993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Tsimbouri P. M. et al. , “ Stimulation of 3D osteogenesis by mesenchymal stem cells using a nanovibrational bioreactor,” Nat. Biomed. Eng. 1, 758–770 (2017). 10.1038/s41551-017-0127-4 [DOI] [PubMed] [Google Scholar]
  • 247. Hodgkinson T. et al. , “ The use of nanovibration to discover specific and potent bioactive metabolites that stimulate osteogenic differentiation in mesenchymal stem cells,” Sci. Adv. 7, eabb7921 (2021). 10.1126/sciadv.abb7921 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Nicodemus G. D. and Bryant S. J., “ Mechanical loading regimes affect the anabolic and catabolic activities by chondrocytes encapsulated in PEG hydrogels,” Osteoarthritis Cartilage 18, 126–137 (2010). 10.1016/j.joca.2009.08.005 [DOI] [PubMed] [Google Scholar]
  • 249. Park I.-S. et al. , “ Effect of joint mimicking loading system on zonal organization into tissue-engineered cartilage,” PloS One 13, e0202834 (2018). 10.1371/journal.pone.0202834 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250. Lin Y.-Y. et al. , “ Applying an excessive mechanical stress alters the effect of subchondral osteoblasts on chondrocytes in a co-culture system,” Eur. J. Oral Sci. 118, 151–158 (2010). 10.1111/j.1600-0722.2010.00710.x [DOI] [PubMed] [Google Scholar]
  • 251. Beekhuizen M. et al. , “ Osteoarthritic synovial tissue inhibition of proteoglycan production in human osteoarthritic knee cartilage: Establishment and characterization of a long-term cartilage–synovium coculture,” Arthritis Rheum. 63, 1918–1927 (2011). 10.1002/art.30364 [DOI] [PubMed] [Google Scholar]
  • 252. Vazquez M. et al. , “ A new method to investigate how mechanical loading of osteocytes controls osteoblasts,” Front. Endocrinol. 5, 208 (2014). 10.3389/fendo.2014.00208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Bhatia S. N. and Ingber D. E., “ Microfluidic organs-on-chips,” Nat. Biotechnol. 32, 760–772 (2014). 10.1038/nbt.2989 [DOI] [PubMed] [Google Scholar]
  • 254. Sieber S. et al. , “ Bone marrow-on-a-chip: Long-term culture of human haematopoietic stem cells in a three-dimensional microfluidic environment,” J. Tissue Eng. Regener. Med. 12, 479–489 (2018). 10.1002/term.2507 [DOI] [PubMed] [Google Scholar]
  • 255. Park S.-H. et al. , “ Chip-based comparison of the osteogenesis of human bone marrow- and adipose tissue-derived mesenchymal stem cells under mechanical stimulation,” PloS One 7, e46689 (2012). 10.1371/journal.pone.0046689 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256. Szczesny S. E., “ Ex vivo models of musculoskeletal tissues,” Connect Tissue Res. 61, 245–247 (2020). 10.1080/03008207.2020.1742418 [DOI] [PubMed] [Google Scholar]
  • 257. Natoli R. M., Scott C. C., and Athanasiou K. A., “ Temporal effects of impact on articular cartilage cell death, gene expression, matrix biochemistry, and biomechanics,” Ann. Biomed. Eng. 36, 780–792 (2008). 10.1007/s10439-008-9472-5 [DOI] [PubMed] [Google Scholar]
  • 258. Dolzani P. et al. , “ Ex vivo physiological compression of human osteoarthritis cartilage modulates cellular and matrix components,” PloS One 14, e0222947 (2019). 10.1371/journal.pone.0222947 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259. Loening A. M. et al. , “ Injurious mechanical compression of bovine articular cartilage induces chondrocyte apoptosis,” Arch. Biochem. Biophys. 381, 205–212 (2000). 10.1006/abbi.2000.1988 [DOI] [PubMed] [Google Scholar]
  • 260. Delco M. L., Bonnevie E. D., Bonassar L. J., and Fortier L. A., “ Mitochondrial dysfunction is an acute response of articular chondrocytes to mechanical injury,” J. Orthop. Res. 36, 739–750 (2018). 10.1002/jor.23651 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Cillero-Pastor B., Rego-Pérez I., Oreiro N., Fernandez-Lopez C., and Blanco F. J., “ Mitochondrial respiratory chain dysfunction modulates metalloproteases -1, -3 and -13 in human normal chondrocytes in culture,” BMC Musculoskeletal Disord. 14, 235 (2013). 10.1186/1471-2474-14-235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Coleman M. C., Ramakrishnan P. S., Brouillette M. J., and Martin J. A., “ Injurious loading of articular cartilage compromises chondrocyte respiratory function,” Arthritis Rheumatol. 68, 662–671 (2016). 10.1002/art.39460 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263. Endres S., Kratz M., Wunsch S., and Jones D. B., “ Zetos: A culture loading system for trabecular bone. Investigation of different loading signal intensities on bovine bone cylinders,” J Musculoskeletal Neuronal Interact. 9, 173–183 (2009). [PubMed] [Google Scholar]
  • 264. Davies C. M. et al. , “ Mechanically loaded ex vivo bone culture system ‘Zetos’: Systems and culture preparation,” Eur. Cell Mater. 11, 57–75 (2006). 10.22203/eCM.v011a07 [DOI] [PubMed] [Google Scholar]
  • 265. Vivanco J. et al. , “ Apparent elastic modulus of ex vivo trabecular bovine bone increases with dynamic loading,” Proc. Inst. Mech. Eng., Part H 227, 904–912 (2013). 10.1177/0954411913486855 [DOI] [PubMed] [Google Scholar]
  • 266. Mann V., Huber C., Kogianni G., Jones D., and Noble B., “ The influence of mechanical stimulation on osteocyte apoptosis and bone viability in human trabecular bone,” J. Musculoskeletal Neuronal Interact. 6, 408–417 (2006). [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.


Articles from APL Bioengineering are provided here courtesy of American Institute of Physics

RESOURCES