Introduction

Osteoarthritis (OA) is the most common degenerative joint disease, affecting more than 25% of the population over 18 years-old. Pathological changes seen in OA joints include progressive loss and destruction of articular cartilage, thickening of the subchondral bone, formation of osteophytes, variable degrees of inflammation of the synovium, degeneration of ligaments and menisci of the knee and hypertrophy of the joint capsule.1 The etiology of OA is multi-factorial and includes joint injury, obesity, aging, and heredity.15 Because the molecular mechanisms involved in OA initiation and progression remain poorly understood, there are no current interventions to restore degraded cartilage or decelerate disease progression. Studies using genetic mouse models suggest that growth factors, including transforming growth factor-β (TGF-β), Wnt3a and Indian hedgehog, and signaling molecules, such as Smad3, β-catenin and HIF-2α,610 are involved in OA development. One feature common to several OA animal models is the upregulation of Runx2.7,8,1113 Runx2 is a key transcription factor directly regulating the transcription of genes encoding matrix degradation enzymes in articular chondrocytes.1417 In this review article, we will discuss the etiology of OA, the available mouse models for OA research and current techniques used in OA studies. In addition, we will also summarize the recent progress on elucidating the molecular mechanisms of OA pain. Our goal is to provide readers a comprehensive coverage on OA research approaches and the most up-to-date progress on understanding the molecular mechanism of OA development.

Etiology

OA is the most prevalent joint disease associated with pain and disability. It has been forecast that 25% of the adult population, or more than 50 million people in the US, will be affected by this disease by the year 2020 and that OA will be a major cause of morbidity and physical limitation among individuals over the age of 40.18,19 Major clinical symptoms include chronic pain, joint instability, stiffness and radiographic joint space narrowing.20 Although OA primarily affects the elderly, sports-related traumatic injuries at all ages can lead to post-traumatic OA. Currently, apart from pain management and end stage surgical intervention, there are no effective therapeutic treatments for OA. Thus, there is an unmet clinical need for studies of the etiology and alternative treatments for OA. In recent years, studies using the surgically induced destabilization of the medial meniscus (DMM) model and tissue or cells from human patients demonstrated that genetic, mechanical, and environmental factors are associated with the development of OA. At the cellular and molecular level, OA is characterized by the alteration of the healthy homeostatic state toward a catabolic state.

Aging

One of the most common risk factors for OA is age. A majority of people over the age of 65 were diagnosed with radiographic changes in one or more joints.2125 In addition to cartilage, aging affects other joint tissues, including synovium, subchondral bone and muscle, which is thought to contribute to changes in joint loading. Studies using articular chondrocytes and other cells suggest that aging cells show elevated oxidative stress that promotes cell senescence and alters mitochondrial function.2629 In a rare form of OA, Kashin-Back disease, disease progression was associated with mitochondrial dysfunction and cell death.30 Another hallmark of aging chondrocytes is reduced repair response, partially due to alteration of the receptor expression pattern. In chondrocytes from aged and OA cartilage, the ratio of TGF-β receptor ALK1 to ALK5 was increased, leading to down-regulation of the TGF-β pathway and shift from matrix synthesis activity to catabolic matrix metalloproteinase (MMP) expression.31,32 Recent studies also indicate that methylation of the entire genomic DNA displayed a different signature pattern in aging cells.33,34 Genome-wide sequencing of OA patients also confirmed that this epigenetic alteration occurred in OA chondrocytes,3537 partially due to changes in expression of Dnmts (methylation) and Tets (de-methylation) enzymes.3840

Obesity

In recent years, obesity has become a worldwide epidemic characterized by an increased body composition of adipose tissue. The association between obesity and OA has long been recognized.41,42 Patients with obesity develop OA earlier and have more severe symptoms, higher risk for infection and more technical difficulties for total joint replacement surgery. In addition to increased biomechanical loading on the knee joint, obesity is thought to contribute to low-grade systemic inflammation through secretion of adipose tissue-derived cytokines, called adipokines.4345 Specifically, levels of pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6, IL-8, and tumor necrosis factor alpha (TNF-α) were elevated4650 in high-fat diet-induced mouse obesity models5154 and in obese patients.5557 These inflammatory factors may trigger the nuclear factor-κB (NF-κB) signaling pathway to stimulate an articular chondrocyte catabolic process and lead to extracellular matrix (ECM) degradation through the upregulation of MMPs.5860

Sport injury

Knee injury is the major cause of OA in young adults, increasing the risk for OA more than four times. Recent clinical reports showed that 41%–51% of participants with previous knee injuries have radiographic signs of knee OA in later years.61 Cartilage tissue tear, joint dislocation and ligament strains and tears are the most common injuries seen clinically that may lead to OA. Trauma-related sport injuries can cause bone, cartilage, ligament, and meniscus damage, all of which can negatively affect joint stabilization.6266 Signs of inflammation observed in both patients with traumatic knee OA and in mouse injury models include increased cytokine and chemokine production, synovial tissue expansion, inflammatory cell infiltration, and NF-κB pathway activation.67

Inflammation

It has been established that the chronic low-grade inflammation found in OA contributes to disease development and progression. During OA progression, the entire synovial joint, including cartilage, subchondral bone, and synovium, are involved in the inflammation process.68 In aging and diabetic patients, conventional inflammatory factors, such as IL-1β and TNF-α, as well as chemokines, were reported to contribute to the systemic inflammation that leads to activation of NF-κB signaling in both synovial cells and chondrocytes. Innate inflammatory signals were also involved in OA pathogenesis, including damage associated molecular patterns (DAMPs), alarmins (S100A8 and S100A9) and complement.6971 DAMPs and alarmins were reported to be abundant in OA joints, signaling through either toll-like receptors (TLR) or the canonical NF-κB pathway to modulate the expression of MMPs and a disintegrin and metalloprotease with thrombospondin motif (ADAMTS) in chondrocytes.7276 Complement can be activated in OA chondrocytes and synovial cells by DAMPs, ECM fragments and dead-cell debris.77,78 Recent studies further clarified that systemic inflammation can re-program chondrocytes through inflammatory mediators toward hypertrophic differentiation and catabolic responses through the NF-κB pathway,9,10,79 the ZIP8/Zn+/MTF1 axis,80 and autophagy mechanisms.8185 Indeed, the recent Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of OA and control samples provide evidence that inflammation signals contribute to OA pathogenesis through cytokine-induced mitogen-activated protein (MAP) kinases, NF-κB activation, and oxidative phosphorylation.149 and post-traumatic150152 OA, mechanics of individual chondrocytes,151,153 and quality evaluation of engineered neo-tissues.154156

Notably, AFM-nanoindentation has made it possible to study the mechanical properties of murine cartilage. Previously, the ~100 μm thickness of murine cartilage prevented such attempts. Because in vivo OA studies are largely dependent on murine models,157 nanoindentation provides a critical bridge across two crucial fields of OA research: biology and biomechanics. The benefit of nanoindentation for murine model studies has been demonstrated by a number of recent studies. For example, cartilage in mice lacking collagen IX (Col9a1−/−)148 showed abnormally higher moduli, while those lacking lubricin (Prg4−/−)158 or chondroadherin (Chad−/−)159 showed lower moduli. Col9a1−/− and Prg4−/− mice also developed macroscopic signs of OA,148,158 underscoring the high correlation between abnormalities in cartilage biomechanics and OA. Li et al. also recently demonstrated the applicability of nanoindentation to the murine meniscus.160 Further applications of nanoindentation to clinically relevant OA models, such as the DMM model,110 hold the potential of assessing OA as an entire joint disease through biomechanical symptoms in multiple murine synovial tissues.

Two other recent technological advances provide paths to further in-depth studies. First, Wilusz et al.161 stained cartilage cryosections with immunofluorescence antibodies of the pericellular matrix signature molecules, type VI collagen and perlecan.162 Using immunofluorescence guidance, nanoindentation was used to delineate the mechanical behavior of cartilage pericellular matrix and ECM,161163 and to reveal the role of type VI collagen in each matrix by employing Col6−/− mice.164 Therefore, it is now possible to directly examine the relationships across micro-domains between biochemical content and biomechanical properties of cartilage,161 meniscus165 or other synovial tissues in situ. Second, Nia et al.166 converted the AFM to a high-bandwidth nanorheometer. This tool enabled separation of the fluid flow-driven poroelasticity and macromolecular frictional intrinsic viscoelasticity that govern cartilage energy-dissipative mechanics.166168 Hydraulic permeability, the property that regulates poroelasticity, was found to be mainly determined by aggrecan rather than collagen169 and to change more drastically than modulus upon depletion of aggrecan.166,170 This new tool provides a comprehensive approach beyond the scope of elastic modulus for assessing cartilage functional changes in OA.

Molecules mediating OA pain

The perception of OA pain is a complex and dynamic process involving structural and biochemical alterations at the joint as well as in the peripheral and central nervous systems. While there have been extensive studies of mediators of OA joint degeneration, only recently have studies begun to characterize biochemical influences on and in the peripheral and central nervous systems in OA. In this regard, OA appears to show similarities and differences with other conditions causing pain.171,172 There are a wide variety of signaling pathways linked to joint destruction and/or pain. In this section we will discuss three emerging and highly relevant pathways that provide insight into the mechanisms underlying OA pain.

Chemotactic cytokine ligand 2/chemokine (C–C motif) receptor 2

Chemotactic cytokine ligand 2 (CCL2), also known as monocyte chemoattractant protein 1 (MCP-1), is well-known to mediate the migration and infiltration of monocytes and macrophages by signaling through chemokine (C–C motif) receptor 2 (CCR2).173 In arthritis, CCL2 promotes inflammation of the joint.174 Evidence also suggests that CCL2 is an important mediator of neuroinflammation.175,176 In neuropathic pain, CCL2 expression is increased in microglia and in sensory neurons in the dorsal root ganglia (DRGs), where CCL2 can be further transported and released into central spinal nerve terminals. Increased CCL2/CCR2 signaling has been correlated with direct excitability of nociceptive neurons and microglial activation, leading to persistent hyperalgesia and allodynia.177,178

In a DMM mouse OA model, CCL2 and CCR2 levels were elevated in DRGs at 8 weeks post surgery, correlating with increased OA-associated pain behaviors. Increased CCL2 and CCR2 levels in the DRG were thought to mediate pro-nociceptive effects both by increasing sensory neuron excitability through CCL2/CCR2 signaling directly in DRG sensory neurons and through CCL2/CCR2-mediated recruitment of macrophages in the DRG. Compared with wild-type mice, Ccr2-null mice showed reduced pain behaviors following DMM with similar levels of joint damage.179 Although CCR2 antagonists are currently being assessed in clinical studies, no clinical studies have targeted CCL2 or CCR2 in OA pain.180

Nerve growth factor/tropomyosin receptor kinase A

In both clinical and animal studies, the targeted inhibition of nerve growth factor (NGF) and inhibition of its cognate receptor, tropomyosin receptor kinase A (TrkA), reduced OA pain. Clinically, the systemic administration of NGF caused persistent whole-body muscle hyperalgesia in healthy human subjects,174,177 while anti-NGF antibody, tanezumab, therapy significantly reduced OA pain.181184 There are a number of potential mechanisms through which NGF mediates pain. Over-expressed NGF in peripheral tissues can bind directly to TrkA at sensory neuron nerve terminals and be retrogradely transported to the DRG. There it stimulates sensory neurons to activate mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling.185 The activation of the NGF-MAPK/ERK axis upregulates the expression of pain-related molecules, including transient receptor potential cation channel subfamily V member I (TRPV1), substance P, calcitonin gene-related peptide (CGRP), brain-derived neurotrophic factor (BDNF), and nociceptor-specific ion channels, such as Cav 3.2, 3.3, and Nav1.8.186188

In addition to direct signaling of sensory neurons, NGF promotes algesic effects by targeting other cell types. For example, NGF/TrkA signaling occurs in mast cells, triggering release of pro-inflammatory and pain mediators, including histamine and prostaglandins, in addition to NGF.186,189 NGF signaling is upregulated by pro-inflammatory mediators, and NGF promotes leukocyte chemotaxis and vascular permeability, further stimulating inflammation.190192 NGF/TrkA signaling further promotes angiogenesis and nerve growth. The process of angiogenesis is not only inflammatory, but also serves as a track for nerve growth into the joint.193

Given the high efficacy of targeting NGF in a clinical study on reducing OA pain, it is of great interest to further define NGF/TrkA pain signaling mechanisms and to find additional therapeutic targets in this pathway. Recent evidence indicates that loss of PKCδ signaling significantly increases both NGF and TrkA in the DRG and synovium, is associated with increased MAPK/ERK signaling at the innervating DRGs, and is associated with OA hyperalgesia.194 However, in recent clinical studies, a small population of patients treated with systemic anti-NGF therapy exhibited rapid progression of OA and were more prone to bone fractures.195 Considering the analgesic effects by anti-NGF therapy on OA-associated pain, understanding of the precise roles of the NGF/TrkA pathway in different joint tissues in OA and OA-associated pain is of great interest.

ADAMTS5

The use of Adamts5 KO mice and therapeutic treatment with anti-ADAMTS5 antibody in wild-type mice produce inhibition of ADAMTS5 signaling/expression in the DMM model, resulting in reduction of both joint degeneration and pain.98,196,197 ADAMTS5 is a major aggrecanase, and because aggrecan is a major component of the proteoglycans in cartilage that provides compressive resistance, ADAMTS5 is thought to be a critical mediator of cartilage degeneration during the development of OA.198 Although variations in pain signaling can be independent from the degree of joint degeneration, the use of Adamts5 KO mice and direct inhibition of joint degeneration with anti-ADAMTS5 antibody may provide insight into how joint degeneration produces OA pain. For example, hyalectan fragments generated by ADAMTS5 have been suggested to directly stimulate nociceptive neurons as well as glial activation, promoting increased pain perception.196,199 Furthermore, inhibition of ADAMTS5 following DMM resulted in reduced levels of CCL2 in DRG neurons, thus suggesting a role for CCL2 in OA-specific pain.197

Pain-related behavior tests

Pain is the most common reason patients seek medical treatment and is a major indication for joint replacement surgery.200,201 Therefore, evaluating pain in pre-clinical animal models is of critical importance to better understand mechanisms of and to develop treatments for OA pain. The evaluation of OA pain in animals involves indirect and direct measures.

Recognizing pain as a clinical sign and quantitatively assessing pain intensity are essential in research for effective OA pain management. Rodent animal models are routinely used for basic and pre-clinical studies because of the relatively low cost of animal maintenance, the abundance of historical data for comparison, and smaller amounts of drugs required for experimental studies. For pain measurements, rodents have advantages over other small animal models, such as rabbits, which present challenges to obtain a pain response and are immobile if startled by an unfamiliar observer. Mice are usually used for the development of genetically engineered strains to enable molecular understanding of OA progression and pain in vivo.202 Larger animals, including dogs, sheep, goats, and horses are also sometimes used for modeling OA pain.202,203

A wide range of direct and indirect measures of pain are used in small animal models of OA. Indirect and/or direct measures of pain include static or dynamic weight bearing, foot posture, gait analysis, spontaneous activity, as well as sensitivity to mechanical allodynia, mechanical hyperalgesia, and thermal, and cold stimuli.202,203 Among indirect tests involving pain-evoked behaviors, mechanical stimuli may be the most correlated with OA pain. A commonly used measure of indirect pain is the von Frey test for mechanical allodynia using filaments to assess referred pain.186,194,196,202,204 Direct mechanical hyperalgesia is performed using an analgesymeter for paw pressure pain threshold. Additional direct measures of OA pain include the hind limb withdrawal test, vocalization evoked by knee compression on the affected knee, the struggle reaction to knee extension, and ambulation and rearing spontaneous movements.194,202,203 Weight-bearing and gait analyses may have important translational relevance for assessing OA pain because these tests are also used to assess clinical OA pain.203 However, obtaining clear pain responses from weight bearing or gait is challenging when using the unilateral DMM mouse model because the nature of OA pain is a dull pain unlike that of, for example, sharp inflammatory pain.

In large animals, pain behavior testing is more challenging and there is no consensus for the best method of evaluating pain.202 However, dogs, the most commonly used large animal, have been suggested to provide the best predictive modeling for OA pain translated into the clinical setting.205 Methods used for assessing pain in large animals are restricted to assessing degree of lameness, gait analysis, and subjective rating scales, which assess descriptors of pain similar to those of humans.

Overall, there is a wide range of pain-behavior tests for small and large animal models. Although no animal model or pain behavior test perfectly translates to OA-associated pain in patients, these tests yield a valuable understanding of the mechanisms of OA pain and allow assessment of treatments for relief from OA-associated pain. Rodents will continue to be widely used for basic OA pain research, but large animals continue to be important because of their greater potential for modeling clinical OA pain.

Future perspective

Although significant progress has been made in OA research in recent years, very little is yet known about the molecular mechanisms of OA initiation and progression. OA is a heterogeneous disease caused by multiple factors. One important potential factor for OA development is Runx2, which is upregulated in several OA mouse models and in cartilage samples derived from patients with OA disease.7,8,11,13,91 Key questions that need to be addressed are: (1) Is Runx2 a central molecule mediating OA development in joint tissue?; and (2) Could manipulation of Runx2 expression be used to treat OA disease? OA is a disease affecting the entire joint, including articular cartilage, subchondral bone, synovial tissues and menisci. In which of these joint tissues OA damage first occurs during disease initiation is currently unknown; this is important because it is directly related to OA treatment. In addition, the interplaying mechanisms among different OA symptoms, such as articular cartilage degradation, osteophyte formation, subchondral sclerosis and synovial hyperplasia, await clarification. The understanding of the molecular mechanisms underlying these issues will accelerate the development of novel therapeutic strategies for OA.