Abstract
This study aims to determine the molecular mechanisms and analgesic effects of transient receptor potential vanilloid 1 (TRPV1) in the treatments of osteoarthritis (OA) and rheumatoid arthritis (RA). We summarize and analyse current studies regarding the biological functions and mechanisms of TRPV1 in arthritis. We search and analyse the related literature in Google Scholar, Web of Science and PubMed databases from inception to September 2023 through the multi‐combination of keywords like ‘TRPV1’, ‘ion channel’, ‘osteoarthritis’, ‘rheumatoid arthritis’ and ‘pain’. TRPV1 plays a crucial role in regulating downstream gene expression and maintaining cellular function and homeostasis, especially in chondrocytes, synovial fibroblasts, macrophages and osteoclasts. In addition, TRPV1 is located in sensory nerve endings and plays an important role in nerve sensitization, defunctionalization or central sensitization. TRPV1 is a non‐selective cation channel protein. Extensive evidence in recent years has established the significant involvement of TRPV1 in the development of arthritis pain and inflammation, positioning it as a promising therapeutic target for arthritis. TRPV1 likely represents a feasible therapeutic target for the treatment of OA and RA.
TRPV1, a member of the TRP family, is widely expressed in primary sensory neurons and various cells and participates in pain signal transduction of thermal, mechanical and chemical stimulation. Recently, functional changes in TRPV1 channels have been considered as risk factors for OA and RA. Abnormal expression and function of TRPV1 channels are associated with chondrocyte ferroptosis, synovial inflammatory response, macrophage polarization and bone homeostasis. In addition, TRPV1 channels play important roles in neural sensitization, defunctionalization or central sensitization and are involved in the occurrence of pain in OA and RA. Based on clinical and preclinical evidence, TRPV1‐targeting drugs and regulation of its expression exhibit prominent curative effects in OA and RA. TRPV1 may be a novel potential therapeutic target for OA and RA treatment.

1. INTRODUCTION
Arthritis is a chronic disease that refers to the joint's inflammation involving progressive degenerative or inflammatory pathological changes in articular cartilage, synovium, subchondral bone and other tissues that eventually lead to joint disability. Arthritis is categorized into two major forms known as osteoarthritis (OA) and rheumatoid arthritis (RA) which remain the core reasons for disability and pain in the elderly. The stiffness and pain caused by arthritis limit an individual's joint function and affect their quality of life. Symptoms worsen gradually with age and deteriorate the joints as the immune system attacks the joints. 1 In addition to pain, individuals with arthritis also experience other clinical symptoms, such as joint stiffness and dysfunction. 2 The currently available treatment for arthritis mainly includes symptomatic treatment including non‐pharmacological rehabilitation (e.g., exercise and diet), pharmacological therapy and surgical intervention. 3 , 4 Drug therapy is generally used to reduce the symptoms of arthritis. Such drugs include non‐steroidal anti‐inflammatory drugs (NSAIDs), opioids, corticosteroids and even immunosuppressive agents; however, these drugs are found to be associated with adverse side effects. 5 , 6 , 7 While TNF inhibitors, anti‐IL‐17/23 antibodies and Janus Kinase inhibitors have shown their efficacy in the management of RA, some of them are still associated with adverse side effects, including headache, hypertension, injection site reactions and allergies and a higher rate of upper respiratory tract infections. 8 , 9 , 10 In addition, the side effects of drugs can even lead to the complete deterioration of joints. 11 Therefore, there is an urgent need to identify the potential target molecules and develop new drugs for local or systemic applications for the treatment of arthritis.
Transient receptor potential (TRP) channels act as ‘molecular receptors’ of cells, composed of six subunits containing transmembrane domains, which were discovered in Drosophila. 12 TRP channels respond to a variety of stimuli, including light, temperature, touch, osmotic pressure, taste, sound and pain. 13 The TRP channel family is divided into six subfamilies and harbours 28 members in mammals, including TRP canonical (TRPC), TRP vanilloid (TRPV), TRP melastatin (TRPM), TRP polycystin (TRPP), TRP mucolipin (TRPML) and TRP ankyrin (TRPA) subfamilies. 14 TRPV1, a member of the TRPV subfamily, is a non‐selective cation channel protein that can be activated by capsaicin, high temperature (>43°C), low PH (<6) and other harmful stimuli. 15 After activation of TRPV1, calcium and sodium ions are allowed to flow into the cells, eventually leading to the release of molecules involved in pain transmission, such as calcitonin gene‐related peptide (CGRP), substance P, 16 somatostatin, 17 glutamate 18 and bradykinin. 19 TRP channel activation exacerbates local inflammation by releasing pro‐inflammatory mediators while also exerting anti‐inflammatory effects. Somatostatin employs systemic anti‐inflammatory and analgesic effects in Complete Freund's adjuvant (CFA)‐induced arthritis models 17 and reduces matrix metallopeptidase (MMP) 1 production by synoviocytes from RA patients. 20 It has been reported that TRPV1 is expressed in various cells, such as chondrocytes, synovial fibroblasts, osteoclasts and osteoblasts. 21 , 22 , 23 , 24 , 25 , 26 , 27 TRPV1 is known to play an analgesic role in various diseases, including OA, 28 RA, 29 postherpetic neuralgia, 30 diabetic neuropathy 31 and cancer. 32 TRPV1 not only serves as a source of anti‐ferroptosis in chondrocytes but also is imperative for inflammation and pain progression in arthritis. 33 , 34 Therefore, based on such characteristics and involvement in arthritis pain, TRPV1 is considered a potential target for the treatment of arthritis.
Here, we reviewed the current treatment status of OA and RA. We have discussed the expression and functional correlation of TRPV1 in different tissues under arthritis condition, as well as the role of TRPV1 in arthritis pain. Additionally, we briefly summarized the clinical application of TRPV1 agonists and antagonists in arthritis diseases.
2. CURRENT TREATMENT STATUS OF OA AND RA
Osteoarthritis (OA), the most common form of arthritis, is a chronic disease characterized by synovial inflammation, cartilage degradation, changes in subchondral bone mass and osteophyte formation. 35 , 36 , 37 OA mainly affects the knee, hip, hands and spinal joints. 35 , 38 , 39 The risk factors for OA include genetics, age, sex, congenital anatomical abnormalities and metabolic disorders. 40 In addition, joint injury and mechanical damage are also responsible for the occurrence of OA. 41 The number of OA patients is increasing yearly, especially with the aging population is significantly expanding in recent years, representing a substantial burden on patients' social and economic well‐being. 42 , 43 However, there are currently no effective treatment strategies available for OA. The purpose of all existing treatments is to relieve symptoms and prevent the complications and progression of the disease to improve the quality of life. Pain is one of the main symptoms of OA and can be relieved by taking NSAIDs and opioid drugs; nevertheless, their efficacy decreases over time. The long‐term use of oral NSAIDs increases the risk of gastrointestinal and cardiovascular side effects. 6 In clinical practice, it is crucial to enhance personalized medication and monitoring for high‐risk and elderly individuals. Furthermore, opioids present significant challenges in terms of tolerance and safety risks, such as abuse potential and rapid tolerance. 7 The use of opioids is linked to the progression of structural degeneration in knee joint. 44 When individuals do not respond to oral or topical analgesics, intra‐articular corticosteroid injections are often administered, 45 providing short‐term pain relief and improved function. However, prolonged usage of this drug fails to alleviate symptoms and may even worsen cartilage degeneration. 46 , 47 Total joint replacement is a viable treatment option for individuals with severe functional impairment, but it has certain limitations due to the longevity of the prosthesis and potential inflammatory reactions. 48 , 49 Total knee arthroplasty is not ideal for individuals with higher preoperative knee pain scores at rest owing to their lower pain threshold, increasing the risk of persistent pain after surgery. 50 Considering these unsatisfactory outcomes, there is an urgent need to discover novel pharmacological approaches for the treatment of OA.
Rheumatoid Arthritis (RA) is an inflammatory autoimmune disease characterized by chronic inflammation of the synovium, leading to the destruction of cartilage and bone tissue in the joints. This progressive condition manifests through symptoms like joint pain, swelling and stiffness. 51 Inflammatory cytokines produced by T cells, B cells, macrophages and synovial fibroblasts in RA, such as tumour necrosis factor‐alpha (TNF‐α), interleukin (IL)‐1 and IL‐17 aggravate the inflammatory response of the synovium and bone destruction. 52 Early and active treatment with disease‐modifying antirheumatic drugs (DMARDs) has proven effective in slowing down the progression of RA and alleviating clinical symptoms. 53 Currently, the main drugs used for the treatment of RA are NSAIDs, glucocorticoids, DMARDs and surgical therapy. DMARDs are divided into conventional synthetic (cs) DMARDs, biological (b) DMARDs and targeted synthetic (ts) DMARDs. Among them, methotrexate (MTX) is a widely used csDMARD and is considered a promising drug for RA treatment. 54 In individuals with an inadequate methotrexate response, bDMARDs combined with methotrexate have shown superiority over bDMARDs monotherapy. 55 , 56 However, considering the potential long‐term immunosuppressive effects and increased risk of infection associated with MTX use, bDMARDs or tsDMARDs should be considered as alternative options for individuals with contraindications to MTX. 56 Glucocorticoids remain common and essential treatment for RA, although their prolonged low‐dose usage raises the risk of cardiovascular disease, osteoporosis and fracture. 57 , 58 , 59 With the rapid advancement of RA treatment drugs, the multifaceted selection and combination of drugs provide more treatment possibilities for the personalized treatment of individuals with RA. In cases of severe RA with joint dysfunction, deformity or insufficient relief from conservative treatment, surgical interventions are often considered the primary approach to alleviate disease progression. 60
In the context of OA and RA, drug therapy plays a crucial role in managing symptoms and slowing disease progression. However, the effectiveness of many drugs for OA or RA treatment is limited due to individual resistance and contraindications, making the development of novel drugs with improved efficacy and fewer side effects a pressing need. In this regard, the TRPV1 channels, which belong to the ion channel family called TRP channels, act as a promising therapeutic target for the development of new drugs with enhanced efficacy and safety profiles.
3. ROLE OF TRPV1 IN ARTICULAR CARTILAGE
Chondrocytes are the sole cell type in articular cartilage, playing a crucial role in maintaining the normal physiological function of articular cartilage. 61 In the pathophysiology of OA, inflammatory molecules like IL‐1β, TNF and IL‐6 act as critical mediators. Specifically, IL‐1β and TNF hinder chondrocyte anabolism and promote the release of proteolytic enzymes, thereby accelerating cartilage degeneration. 62 , 63 Valdes et al. demonstrated that TRPV1 gene expression in chondrocytes was increased in response to pro‐inflammatory cytokines IL‐1 and TNF‐α, suggesting that changes in TRPV1 expression in chondrocytes appear to play an important role in the structural damage in OA. 64 However, only a few studies have evaluated the role of TRPV1 in cartilage function. Furthermore, various types of chondrocyte death, including cell necrosis, apoptosis, autophage 61 and ferroptosis 65 are involved in OA pathogenesis. Many previous studies have demonstrated that inhibition of TRPV1 channels can reduce chondrocyte degeneration, but the role of TRPV1 channel activity in arthritic chondrocytes is controversial. Interestingly, recent research in mice has indicated that the TRPV1 agonist capsaicin activates TRPV1 channels in OA chondrocytes, promoting the expression of the antioxidant enzyme glutathione peroxidase 4 (GPX4). GPX4 protects chondrocytes from ferroptosis and reduces cartilage degradation. 34 , 66 , 67 GPX4 is a crucial regulator of ferroptosis, and the reduction in its expression leads to increased sensitivity of chondrocytes to oxidative stress and extracellular matrix degradation. These findings suggest a complex role for TRPV1 in maintaining cartilage homeostasis, necessitating further investigation into its mechanism and impact on chondrocyte function in OA.
Chondrocytes are known to respond to mechanical stimuli such as compression forces and shear stress. Excessive mechanical loading contributes to OA pathogenesis, leading to catabolic reactions and chondrocyte death. 68 In vitro stimulation of chondrocytes with pro‐inflammatory cytokines has been utilized to mimic changes in cartilage degradation associated with OA. 62 , 69 , 70 Inflammatory cytokines induce extracellular calcium ion (Ca2+) influx and nuclear translocation of nuclear factor‐kappa B (NF‐κB) in chondrocytes, exacerbating cartilage degradation. 71 Mechanical strain promotes the efflux of intracellular Ca2+ through the TRPV1 channel, and the intracellular Ca2+ concentration decreases to inhibit NF‐κB activity, leading to a decrease in the expression of a disintegrin and metalloproteinase with thrombospondin motifs 9 (ADAMTS9) induced by inflammatory cytokines. Inhibition of TRPV1 activity using a mechanically gated channel inhibitor, such as capsazepine, abolishes the regulation of ADAMTS9 by mechanical strain, while inhibition of TRPV2 and TRPV4 has no such effect. 71 Therefore, mechanical strain‐induced attenuation of ADAMTS9 expression in chondrocytes may be mainly achieved by TRPV1‐associated channels. It is commonly known the fact that cell migration is influenced by changes in intracellular Ca2+ concentration. TRPV1 is a non‐selective Ca2+ channel implicated in cell migration. 72 Studies on high‐density micromass cell cultures in chickens and mice have demonstrated that mesenchymal cells expressing TRPV1 migrate and proliferate, contributing to cartilage formation, which implies that TRPV1 activity may affect chondrocyte proliferation and extracellular matrix production. 73
4. ROLE OF TRPV1 IN SYNOVIUM
Cartilage degeneration and the production of cartilage fragments in arthritis promote the inflammatory response of the synovium and are often accompanied by infiltration of macrophages and T lymphocytes. 74 These cells secrete cytokines and chemokines, exacerbating cartilage catabolism. Inflammatory factors can also promote angiogenesis. 75 Simultaneously, a reduction of synovial fluid leads to increased friction between articular cartilages, forming a vicious circle that eventually leads to progressive joint degeneration. 76 , 77 Clinically, individuals with OA have a 50% chance to develop synovitis, which causes joint pain, hyperthermia and swelling. 78 The severity of synovitis is strongly correlated with the progression of cartilage destruction in OA. 79 Synovial fibroblasts are important cells of the joint synovium and play a crucial role in the inflammatory response of arthritis. 80 In chronic arthritis induced by Complete Freund's Adjuvant, mice with TRPV1 receptor deletion showed significantly reduced joint swelling and cartilage destruction. 81 , 82 The expression of TRPV1 mRNA and protein in synovial tissue is increased in human and rat arthritis models. 22 , 23 , 24 TRPV1 activation in synovial fibroblasts can promote the expression of pro‐inflammatory cytokines and participate in the partial production of reactive oxygen species (ROS). 22 , 83 , 84 , 85 Conversely, the expression of TRPV1 can also be upregulated by pro‐inflammatory cytokines and ROS. 83 It has been shown that subcutaneous injection of the peptide APHC3 (a TRPV1 antagonist) extracted from sea anemones reduced joint swelling, thermal and mechanical hypersensitivity of pain and improved grip strength in complete Freund's Adjuvant (CFA‐) and monosodium iodoacetate (MIA)‐induced OA. 86 The destruction of articular cartilage was significantly reduced from day 8 to day 15, and the concentration of IL‐1β in synovial fluid was significantly reduced at day 15. 86 The TRPV1 antagonist can reduce chondrocyte degeneration by inhibiting the joint inflammatory response, suggesting that targeting the TRPV1 channel is a potentially valid approach for the treatment of joint synovial inflammation.
Synovial macrophages are necessary for maintaining homeostasis within the joint. The abnormal aggregation of synovial macrophages in joints has been implicated in the pathogenesis of OA and RA, 87 and the inhibition of macrophage generation and aggregation can reduce structural damage. Recent studies showed that the expression of macrophages and TRPV1 was increased in synovial tissues of the knee joint of individuals with RA and OA, and most TRPV1 is highly localized to CD68‐positive macrophages. 23 Subsequently, another study showed that TRPV1 is always expressed in M1 macrophages, 24 suggesting that TRPV1 may be related to the function of M1 macrophages. Intra‐articular injection of a TRPV1 agonist capsaicin activates TRPV1 and inhibits M1 macrophage polarization because the activation of TRPV1 induces large amounts of Ca2+ influx through TRPV1 channels. This increase in Ca2+ influx leads to the phosphorylation of intracellular calmodulin‐dependent protein kinase II (CaMKII), promotes the nuclear translocation of nuclear factor erythroid‐2 related factor 2 (Nrf2) and ultimately results in the reduction of macrophages polarization. 24 Compared with the control group, the infiltration of M1 macrophages in the synovium of OA mice after capsaicin injection was reduced, the cartilage surface was smoother, and there was a reduction observed in both the surface fibrosis area and the loss of cartilage extracellular matrix. 24 These findings suggest that, through activating the Ca2+/CaMKII/Nrf2 pathway, TRPV1 agonist has the ability to reduce M1 macrophage infiltration in the synovial tissue inflammation.
In addition to the identification of TRPV1 downstream signalling molecules, the upstream regulatory mechanisms of TRPV1 have also been reported by a recent study. It has been reported that TRPV1 is regulated by hyaluronic acid (HA). 88 HA is continuously secreted by synovial lining cells into the joint fluid and is widely distributed in the extracellular matrix in mammals. 89 HA affects the viscoelasticity of synovial fluid and plays an important role in the lubrication and cushioning of joints. 90 Compared with normal joint synovial fluid, HA in synovial fluid usually depolymerizes from high to low molecular weight in OA, resulting in a decrease in the viscoelasticity of synovial fluid, 91 which eventually leads to joint damage. Interestingly, Caires and colleagues found that administration of HA inhibited the opening of TRPV1 channels in nociceptive terminals, reduced the response to noxious stimuli and reduced joint pain. 88 A reasonable explanation is that HA may normally maintain the TRPV1 channel of nociceptive terminals in a closed state. When arthritis occurs, HA elastoviscous filtering capacity decreases, reducing the regulation of the TRPV1 channel and increasing the excitability of peripheral nociceptive neurons.
5. ROLE OF TRPV1 IN SUBCHONDRAL BONE
Normal bone homeostasis depends not only on extracellular Ca2+ concentration but also on the regulation of multiple bone cells and their intracellular Ca2+ signalling. 92 The dynamic balance between osteoblast‐mediated bone formation and osteoclast‐mediated bone resorption ensures the relative stability of the mechanical strength of trabecular bone, allowing joints to withstand sufficient stress. 93 In OA, decreased osteoprotegerin secretion by osteocytes leads to enhanced osteoclast‐mediated bone resorption. 94 With the increase in mechanical loading, the decrease in sclerostin secreted by osteocytes leads to the enhancement of the wingless‐type MMTV integration site (Wnt) signalling pathway, ultimately resulting in the enhancement of osteoblast‐mediated bone formation. 39 In the late stage of OA, abnormal bone remodelling caused by joint biomechanical disturbances can lead to insufficient mineralization of subchondral bone, making it more vulnerable to destruction. 95 The receptor activator of nuclear factor kappa‐B ligand (RANKL) plays a crucial role in bone loss in OA and RA. Osteoclasts can participate in the regulation of bone resorption through the RANKL‐RANK signalling pathway. 96 , 97 , 98 Several studies have shown that TRPV1 is expressed in osteoblasts and osteoclasts and is involved in balancing bone resorption and formation. 26 , 27 , 99 TRPV1 antagonists inhibited the osteoclast differentiation in vitro and reduced bone resorption and protected ovariectomy‐induced bone loss in mice. 27 Increased bone volume fraction, trabecular thickness and bone mineral density were observed in TRPV1 knockout (KO) mice. 100 This may be due to the fact that absence of TRPV1 inhibits extracellular Ca2+ influx into osteoclasts, leading to the down‐regulation of nuclear factor of activated T cells c1 (NFATc1), a downstream transcription factor for Ca2+ signalling and ultimately leads to the reduction of osteoclast formation. 100 NFATc1 is a key downstream target of the RANKL‐RANK signalling pathway involved in osteoclastogenesis and plays a crucial role in regulating bone homeostasis. 101 Therefore, TRPV1 channels participate in the regulation of bone remodelling by affecting NFATc1 activity during OA pathology. Pretreatment of rats with TRPV1 agonists, such as resiniferatoxin (RTX) causes desensitization of nerve endings expressing TRPV1 receptors without affecting the non‐neural TRPV1 channels. 102 Pre‐treatment with capsaicin for 2 weeks could effectively inhibit the reduction of bone volume and trabecular number in MIA‐induced OA, 103 indicating that capsaicin pre‐treatment reduces bone damage in early OA, such as the destruction of trabecular bone, but whether it is effective for end‐stage OA remains unclear. Furthermore, RTX pre‐treatment resulted in increased bone mass and thickness of the trabecular while reducing average trabecular separation in the collagen‐antibody‐induced RA mouse model, suggesting that defunctionalization of capsaicin‐sensitive afferents leads to decreased bone resorption during joint inflammation. 104
Tissue acidosis is an essential pathological feature of arthritis, where inflammation of synovial tissue during arthritis progression leads to localized acidification, which may lead to changes in ion channel activity. With the increase in hydrogen ion concentration in RA synovial fluid, extracellular acidification can induce chondrocyte pyroptosis by activating acid‐sensing ion channel 1a (ASIC1a), leading to irreversible cartilage degradation. 105 , 106 Moderate extracellular acidification (pH >6) does not directly activate TRPV1 channels 107 but it inhibits induced cell death by modulating Ca2+ and NF‐κB translocation and ROS production in synovial cells. 108 Particularly, local acidosis promotes osteoclastogenesis and SB‐366791 (TRPV1 antagonist) inhibits osteoclast activation. 109 Studies have also shown that bone loss can be reduced by simultaneously stimulating cannabinoid receptor 2 and blocking TRPV1 channels. 110 These findings reveal TRPV1 as a potential therapeutic target for preventing bone loss and regulating bone mass in arthritis. Future studies may be aiming at investigation how TRPV1 expression in osteoclasts can be exploited to restore the balance between osteoclast and osteoblast bone homeostasis under pathological conditions of arthritis.
6. ROLE OF TRPV1 IN ARTHRITIS PAIN
TRPV1 is a critical ion channel involved in pain perception and is considered a potential target for the treatment of OA pain. The Ile585Val genetic variant in the coding region of the TRPV1 gene is associated with pain in individuals with symptomatic OA. 64 Early studies suggested that compared with wild‐type counterparts, TRPV1 receptor KO (Trpv1 −/− ) mice showed little response to inflammatory heat pain but a normal response to noisier mechanical stimuli. 111 , 112 Some recent studies have also reported that TRPV1 receptor gene‐deleted (Trpv1 −/− ) mice reduce mechanical hyperalgesia. 81 , 82 , 113 Similarly, TRPV1 antagonists were effective in suppressing arthritic mechanical allodynia. 114 These studies suggest that the TRPV1 receptor is essential for maintaining mechanical hyperalgesia and joint inflammatory pain.
The sensitization of peripheral afferents and central sensitization are key factors in the development of arthritis pain. According to different neurochemical characteristics, primary afferent nociceptors can be divided into peptidergic and non‐peptidergic neurons, and most TRPV1‐positive afferent nerves are peptidergic. 115 TRPV1 is mainly located in C‐fibre and some Aδ‐fibre primary afferents, 116 involved in the conduction of neurogenic and mechanical pain that innervates the knee and ankle. 117 TRPV1 is highly enriched in nociceptors containing neuropeptides, such as afferents containing CGRP and substance P. 115 , 118 The co‐localization of TRPV1 and CGRP in afferent nerves is increased in OA and is thought to play a role in the perception of joint pain. 119 CGRP is related to the pain of peripheral and central sensitization of arthritis and can promote the production of TNF‐α, IL‐1β and IL‐6 in monocytes, thereby increasing the inflammatory response of tissues and cartilage degradation. 120 , 121 , 122 , 123 In arthritis, nociceptor nerve endings can promote the release of neuropeptides (CGRP, substance P, glutamate and somatostatin) from C‐fibre neurons into the periphery by activating TRPV1. 17 , 124 , 125 Subsequent nociceptor depolarization generates action potentials along the L3‐L5 dorsal root ganglion (DRG), causing the release of neurotransmitters in layers I and II of the dorsal horn of the spinal cord, 126 , 127 leading to central sensitization 128 (Figure 1). Somatostatin exerts both anti‐inflammatory and analgesic effects in arthritis, 17 suggesting that the analgesic effect induced by TRPV1 agonists may be partially attributed to somatostatin released from primary afferent nerves. Furthermore, local activation of metabotropic glutamate receptor 5 (mGluR5) in the infralimbic cortex (IL) is associated with thermal hyperalgesia in rodents. 129 Spinal TRPV1 also participates in the descending pronociceptive effect induced by 5mGluR5 activation in mono‐arthritis (ARTH). 130
FIGURE 1.

TRPV1 channels play an essential role in generating OA pain. (A) Nociceptive signals from nociceptors are transmitted to the spinal cord via unmyelinated C or myelinated A fibres. Nociceptive signals are transmitted from the dorsal horn of the spinal cord to higher centres and then to the cerebral cortex to produce pain sensation. (B) In response to inflammatory factors such as nerve growth factor (NGF), tumour necrosis factor‐alpha (TNF‐α) and interleukin (IL)‐1β, TRPV1 channels are activated to transmit pain signals. Protein kinase C (PKC) promotes TRPV1 phosphorylation, increasing sensitivity. Activation of TRPV1 channels leads to increased neurotransmitter release and extracellular regulated protein kinases (ERK) phosphorylation, leading to peripheral nerve sensitization. In addition, joint oedema, vasodilatation and inflammatory response occur under the action of inflammatory factors. (C) Nociceptive signals are transmitted along the dorsal root ganglion (DRG) and central synapses in the spinal cord release various neuropeptides (e.g., calcitonin‐gene‐related peptide (CGRP), glutamate or substance P) resulting in central sensitization.
The extracellular signal‐regulated kinase (ERK) is a widely expressed cell signalling molecule. ERK is phosphorylated in sensory neurons, thought to underlie the mechanism of thermal hyperalgesia. 131 Studies have shown that arthritis pain is associated with the activation of the ERK pathway, and inhibition of this pathway reduces nociceptive behaviour and inflammatory response. 132 Activation of TRPV1 channels can promote the release of CGRP and glutamate, thus increasing ERK phosphorylation in neurons, suggesting that TRPV1 may mediate the activation of ERK in the traumatic primary afferent nerves. 133 During arthritis pain, multiple inflammatory mediators directly act on auto‐receptors and can enhance TRPV1 channel protein phosphorylation by activating various protein kinases. These mediators function as endogenous ligands of TRPV1 channels. 134 , 135 , 136 Protein kinase Cε (PKCε) mediates the phosphorylation and dephosphorylation of TRPV1 at Ser502 and Ser800, playing an indispensable role in inflammation and neuropathic pain progression. 137 In MIA‐induced arthritis rats, PKCɛ activates TRPV1 by inducing TRPV1 phosphorylation at Ser800, resulting in inflammatory hyperalgesia in the rats. However, TRPV1 at Ser502 is not affected, possibly due to the different functions of PKA and PKC in regulating TRPV1 in various neurons or diseases. 138 Increased expression of inflammatory factors, such as nerve growth factor (NGF) in OA promotes TRPV1 phosphorylation and functional enhancement, leading to obvious hyperalgesia. 139 Activation of the NGF receptor tyrosine kinase A (TrkA) at Y760 triggers the activation of Src kinase by phosphoinositide 3‐kinase, resulting in increased TRPV1 phosphorylation. This facilitates the translocation and insertion of TRPV1 into the cell membrane surface and ultimately induces hyperalgesia. 140 In addition, several endogenous ligands, such as cannabinoids, linoleic acid metabolites and lysophosphatidic acid directly activate the TRPV1 channel to promote the release of neuropeptides from nerve endings. This results in inflammatory responses and the production of pain. 141 This therapeutic approach may preserve drug efficacy while avoiding drug‐induced hyperthermia by mediating endogenous ligands, thereby inhibiting TRPV1 channel activity rather than directly inhibiting TRPV1 activity.
TRPV1 antagonists and activators serve as agents that contribute to the analgesic effects in arthritis (Table 1). TRPV1 antagonists such as A‐995662, A‐889425, JNJ‐17203212 or APHC3 reduce the excitability of peripheral afferent fibres (C and Aδ fibres) and inhibit the release of neuropeptides in the spinal cord, effectively reducing changes in pain‐related behaviour. These changes include thermal and mechanical hyperalgesia, asymmetric weight‐bearing of extremities and decreased hindlimb withdrawal threshold in OA and RA models. 23 , 86 , 127 , 144 TRPV1 agonists, such as capsaicin and RTX, can reduce pain in arthritis after pre‐treatment, 103 , 126 , 149 , 150 which is associated with the specific desensitization, 151 defunctionalization 152 and structural ablation 153 of nerve endings (Table 2). Long‐term use or high doses of the agonists capsaicin or RTX can act on the terminals of nociceptive fibres expressing TRPV1, activate TRPV1 and mediate a large influx of Ca2+. This leads to long‐term defunctionalization of its nerve terminals without responding to further deleterious stimuli and resulting in long‐lasting analgesic effects. 149 , 164 However, the role of TRPV1 in arthritis pain is also controversial. Some studies have reported that systemic administration of the TRPV1 inhibitor AZD1386 has no effect on pain management in individuals with knee OA compared to administering the placebo. 165 In contrast, another study reported that AZD1386 failed to relieve pain in a rat model of OA. 166 Furthermore, Okun et al. found that the TRPV1 antagonist AMG981 did not affect the persistent pain caused by advanced MIA. 145 It is possible that TRPV1 expression in the DRG is increased mainly in the early stages of MIA‐induced OA, whereas TRPV1 has no effect on mechanical nociception or does not play a dominant role in persistent pain at later time points. 167 The discrepancies in these studies may reflect the drug effects on different animal models, on different types of pain, such as acute and chronic pain or on different stages of pain, such as early and late stages of pain.
TABLE 1.
Effects of TRPV1 channel drugs in preclinical models.
| Type of models | Exp. animals | Drugs | Treatments | Effects | Refs. |
|---|---|---|---|---|---|
| Carrageenan‐induced knee‐joint RA | Male Sprague–Dawley rats | RTX | 30 μL (0.0003% or 0.003%) for 8 days; single intra‐articular administration. | Pain score↓; Limp and weight distribution ratio↓; Joint swelling↓ | 142 |
| MIA‐induced OA | Male Sprague–Dawley rats | A784168; A795614 | A784168 (3, 10 or 30 μmol/kg) or A795614 (30, 100 or 300 μmol/kg); oral administration; A784168 or A795614 (10, 30,1 00 nmol); intrathecal administration. | Weight‐bearing differences↓; mechanical allodynia↓; thermal hyperalgesia↓ | 114 |
| MIA‐induced OA | Male Sprague–Dawley rats | ABT‐102; A993610 | ABT‐102 (3 μmol/kg or 10 μmol/kg) or A993610 (12 μmol/kg) twice daily for 12 days; oral administration. | Inflammatory pain↓; weight‐bearing differences↓; grip force↑ | 143 |
| MIA‐induced OA | Sprague–Dawley rats | Capsaicin | Pretreatment with 100 μL 0.5% capsaicin for 14 days; intra‐articular administration. | Mechanical hyperalgesia↓; weight‐bearing defect of the hind paw↓; paw ambulation scores↑; subchondral bone erosions and destruction in knee joints↓ | 103 |
| MIA‐induced OA | Male Sprague–Dawley rats | A995662 | Single administration of 3, 10, 30 or 100 μmol/kg; oral administration; Repeated administration of 15 μmol/kg (twice per day) for 12 days; oral administration. | Pain↓; hind limb grip force↑; basal spinal release of CGRP and glutamate ↓ | 127 |
| MIA‐induced OA | Male Sprague–Dawley rats | A889425 | 10, 30, 100 or 300 μmol/kg; oral administration; 10 and 30 μmol/kg, intravenous administration. | Behavioural grip force↑; the evoked firing of nociceptive specific neurons in OA↓ | 144 |
| MIA‐induced OA | Male Sprague–Dawley rats | AMG9810 | 30 mg/kg, intraperitoneal administration. | Thermal hyperalgesia↓; the weight‐bearing shift induced by a 3 mg dose of MIA↓ | 145 |
| MIA‐induced OA | Male Sprague–Dawley rats | JNJ‐17203212 | 1 mg/50 μL; intra‐articular injection; 1 mg/300 μL, i.p. injection; 0.075 or 0.15 mg/100 μL; intra‐arterial injection; 18.75, 37.5 or 75 μg/50 μL; topical spinal injection. | Weight‐bearing asymmetry↓; mechanically evoked responses of knee joint afferents and dorsal horn wide dynamic range neurons ↓ | 23 |
| MIA‐induced OA | Male Sprague–Dawley rats | RTX | 50 μL (0.0003%, 0.003% or 0.03%) for 21 days; intra‐articular injection. | Weight‐bearing↑; mechanical allodynia↓; thermal hyperalgesia↓; expression of CGRP in DRG↓ | 126 |
| Carrageenan‐induced RA | Male C57BL/6 mice | CapsaicinRTX | Pretreatment with 0.01% Capsaicin, 0.001% RTX or 0.0003% RTX for 7 days; intra‐articular injection. | Evoked pain scores↓; antinociception↑; dynamic weight bearing↑ | 146 |
| OA | Dogs | RTX | Single intra‐articular injection of 10 μg/100 μL. | Pain scores↓; gait and weight‐bearing↑ | 147 |
| Collagen‐antibody‐induced RA | Male C57BL/6 mice | RTX | Pretreatment of 30, 70 or 100 μg/kg for 3 consecutive days; subcutaneous injection. | Mechanical hyperalgesia↓; cold allodynia↑; joint oedema and arthritis severity↓; bone density and trabecular bone thickness↑ | 104 |
| CFA‐induced RA; MIA‐induced OA | Male Sprague–Dawley rats | APHC | 0.01, 0.05, 0.1 or 1 mg/kg; subcutaneous administration. | Joint swelling↓; pain‐induced behaviour↓; articular cartilage destruction↓; IL‐1β concentration in synovial fluid↓ | 86 |
| OA | Male and female C57BL/6 mice | Capsaicin | 0.5 and 1.0 mg/kg five times per week for 12 weeks; i.p. injection. | DMM‐induced pain in male mice↓; TRPV1 expression in DRG of male mice↓; joint cartilage degeneration has not changed | 148 |
Abbreviations: ↑, increased; ↓, decreased; CFA, Complete Freund's adjuvant; CGRP, calcitonin gene‐related peptide; DMM, destabilized medial meniscus; DRG, dorsal root ganglion; IL‐1β, interleukin‐1β; MIA, monosodium iodoacetate; OA, osteoarthritis; RA, rheumatoid arthritis; RTX, resiniferatoxin.
TABLE 2.
The Functional mechanism of TRPV1 channel drugs in different treatment patterns or drug concentrations.
| Drugs | Treatment patterns/drug concentrations | Functional mechanism | Refs. |
|---|---|---|---|
| TRPV1 agonists | A single intrathecal injection | Depletion of substance P from primary sensory neurons. | 16, 154, 155, 156 |
| Concentration of treatment | TRPV1 activation induces Ca2+ influx and inactivation of voltage‐dependent sodium channels, voltage‐dependent calcium channels and Piezo2. Inactivation of these channels causes a decrease in their ability to generate and propagate action potentials and leads to transient analgesia. | 157, 158 | |
| TRPV1 activation induces Ca2+ influx, leading to receptor desensitization of TRPV1 ion channels. Desensitization of the TRPV1 receptor reduces the response of TRPV1 to endogenous ligands or inflammatory mediators, which in turn reduces TRPV1‐related hyperalgesia. | |||
| Single high‐concentration or multiple low‐concentration | TRPV1 activation and influx of large amounts of Ca2+, leading to the activation of Ca2+‐dependent protease calpain. Ablation of nerve terminals by calpain activation results in long‐term defunctionalization of TRPV1 expressing afferents and mediates long‐lasting analgesia for chronic pain. | 157, 159, 160 | |
| TRPV1 antagonists | Concentration of treatment | Blocking channel pores and inhibiting heat‐ and proton‐induced Ca2+ influx in sensory neurons, thereby reducing inward current generation or neuropeptide release. In addition to TRPV1, capsazepine also blocks voltage‐sensitive calcium channels and nicotinic acetylcholine receptors, ultimately exerting analgesic effects. | 23, 161, 162, 163 |
Systemic administration of TRPV1 antagonists induces hyperthermia in both humans and animals. 168 , 169 Under normal conditions, neurons expressing TRPV1 in internal organs become overactivated to inhibit skin vasoconstriction. However, systemic administration of TRPV1 antagonists leads to the desensitization of these neurons resulting in cutaneous vasoconstriction and subsequent hyperthermia. 170 , 171 Notably, TRPV1 KO mice did not exhibit severe dysregulation in thermo‐regulation, 172 suggesting that other mechanisms also play a role in maintaining a relatively stable body temperature. Furthermore, in an OA pain model, local administration of TRPV1 antagonist JNJ‐17203212 produced the same analgesic effect as systemic administration and did not affect core body temperature. 23 Concerning the route of administration, topical capsaicin creams or high‐dose capsaicin‐containing patches have been approved as effective adjuvant therapy for the insistent pain of post‐herpetic neuralgia and in the treatment of OA, but their use has been limited by the relatively weak efficacy of capsaicin creams, the need for multiple applications and skin irritation. 173 Topical capsaicin creams can be used in combination with other analgesics, providing a low‐risk choice for patients who cannot achieve pain control using other regimens. Oral or intraperitoneal administration has been widely introduced in preclinical trials but is not an ideal option, given the adverse effects of capsaicin on blood pressure, respiration and other reflex pathways and the potential for increased body temperature and cardiovascular complications associated with TRPV1 antagonists. 174 , 175 In addition, TRPV1 antagonists can exert potent analgesic effects by blocking the centrally located TRPV1 receptors. Some drugs (e.g., A‐425619 and A‐795614) have low central nervous system penetration, so intrathecal analgesia is better than oral administration. 103 , 114 The intrathecal injection can achieve the same analgesic effect as an intra‐articular injection, and intrathecal injection can also alleviate the pain caused by local inflammatory factors within the joint and surrounding soft tissues. 176 Intra‐articular therapeutic strategies appear well suited for the management of OA, in particular, the knee OA. Intra‐articular RTX suppressed the carrageenan‐induced oedema by at least one‐third, and its smaller therapeutic dose was approximately 1000 times lower than the dose producing analgesia following systemic administration of RTX in rats. 142 , 177 Additionally, intra‐articular administration of an analgesic dose of TRPV1 agonist or antagonist could reduce many adverse effects, such as hyperthermia and the unnecessary loss of C‐fibre in the non‐target tissues.
There is growing evidence of sex differences in TRPV1 sensitivity. Clinical studies have shown gender disparities in TRPV1‐mediated pain responses in various body regions, including the back of the hand, ankle, dental pulp and forehead. 178 , 179 , 180 The results of Bai et al. suggest that testosterone may regulate TRPV1 expression and affect pain responses in the CFA‐induced chronic pain model. 181 In another study related to OA, a high‐dose TRPV1 antagonist, capsazepine (30 mg/kg), was found to reverse mechanical pain abnormalities more significantly in male OA mice than in female mice. Additionally, down‐regulation of TRPV1 expression was observed in the DRG tissues of male after treatment, but not in female mice. These findings indicate the presence of sex differences in the regulation of TRPV1 expression by capsazepine. 148 Most current studies on the role and mechanisms of TRPV1 channels in arthritis pain have primarily involved male rodents. Therefore, it is essential to identify sex‐specific genes related to TRPV1 and elucidate the underlying mechanisms of gender differences in the treatment of arthritis pain.
7. CLINICAL APPLICATION OF TRPV1‐TARGETING DRUGS IN ARTHRITIS
TRPV1 agonists and antagonists are currently used in the treatment of arthritis pain. TRPV1 antagonists act directly on TRPV1 receptors to reduce pain‐related signal transduction and neuropeptide release, thereby inhibiting the generation of pain. The analgesic effect of TRPV1 agonists (such as capsaicin) can be maintained for weeks or even months through desensitization, neuropeptide depletion and nociceptor terminal ablation. However, the pain is restored due to the regeneration of nociceptor nerves after the application of capsaicin. 182 Since TRPV1 and TRPA1 are co‐expressed in neurons, capsaicin‐induced ablation of nociceptor terminals may affect TRPA1‐mediated pain and inflammation. 183 , 184 Therefore, capsaicin treatment can block all functions of TRPV1 expressing nociceptors, providing individuals with a more effective therapeutic effect. Here, we summarize the current TRPV1 channel drugs mainly used for arthritis treatment (Table 3).
TABLE 3.
TRPV1 channel drugs for the treatment of arthritis pain.
| Type of arthritis | Number of patients | Drugs | Treatments | Effects | Refs. |
|---|---|---|---|---|---|
| RA, OA | 70 (OA patients) and 31 (RA) patients | Capsaicin cream | 0.025% capsaicin cream or placebo four times daily; 4 weeks | RA and OA patients demonstrated 57% and 33% reductions in pain, respectively. | 29 |
| OA | 167 | Capsaicin glyceryl trinitrate | placebo, 0.025% capsaicin, 1.33% glyceryl trinitrate or 0.025% capsaicin and 1.33% glyceryl trinitrate four times daily; 6 weeks |
Glyceryl trinitrate enhanced the analgesic effect of capsaicin; A combination of capsaicin and glyceryl trinitrate reduced the burning sensation. |
185 |
| OA | 611 | Civamide cream | civamide cream 0.075% group applied on average 2.4 g/day and civamide cream 0.01% (control) group applied on average 2.6 g/day, three times daily; 12 weeks and extension | The treatment effect of the civamide cream 0.075% group was better than that of the civamide cream 0.01% (control) group. | 186 |
| OA | 191 | AZD1386 | 30 mg or 90 mg AZD1386 or placebo; 4 weeks |
Elevated alanine aminotransferase and aspartate aminotransferase; No treatment differences in WOMAC pain |
165 |
| OA | 33 | Mavatrep (JNJ39439335) | A single dose of mavatrep (50 mg), naproxen (500 mg) or placebo was administered twice daily | Pain, stiffness and physical function were significantly reduced. | 187 |
| OA | 24 | Mavatrep (JNJ39439335) | 10, 25 or 50 mg mavatrep or placebo, once a day | The pain was significantly reduced after stair climbing. | 188 |
| OA | 54 | NEO6860 | NEO6860 (500 mg), naproxen (500 mg) or placebo twice daily; 1 day | The NEO6860 treatment group had a trend of analgesia, but the difference was not statistically significant compared with the placebo group. | 189 |
| OA | 172 | CNTX‐4975 | Single intra‐articular injection of a placebo, CNTX −4975 0.5 mg or CNTX‐4975 1.0 mg; 24 weeks | OA–associated pain decreased at week 12 in the 0.5‐mg group and at week 24 in the 1.0‐mg group. | 190 |
Abbreviations: OA, osteoarthritis; RA, rheumatoid arthritis; WOMAC, Western Ontario and McMaster Universities Arthritis Index.
8. TRPV1 AGONISTS
In a 4‐week, double‐blind, randomized, multi‐centre study, the use of topical capsaicin cream showed a reduction in pain among patients with OA when compared to a placebo. However, it did not provide pain relief for individuals with RA. It should be noted that the treatment site experienced a transient burning sensation as a side effect of the capsaicin cream. 29 , 191 The analgesic effect of combined topical capsaicin and nitroglycerin was increased compared with capsaicin alone. 185 There was also evidence that topical capsaicin was superior to placebo in terms of pain relief and that topical NSAIDs and topical capsaicin provided comparable pain relief efficacy. 192 Civamide is another TRPV1 receptor agonist with a mechanism of action similar to capsaicin. The continuous application of 0.075% civamide showed positive effects on pain reduction and Subject Global Evaluation (SGE) in individuals with OA for a duration of 1 year. Moreover, the use of civamide cream does not result in systemic absorption and is considered safe based on its profile. 186
CNTX‐4975 is a highly purified trans‐capsaicin. The efficacy and safety of a single intra‐articular injection of different doses (0.5 mg or 1.0 mg) of CNTX‐4975 were recently evaluated in a randomized, double‐blind, placebo‐controlled, dose‐ranging, Phase IIb study involving 175 patients with knee OA who were clinically assessed over a period of 24 weeks. 190 Significant improvements were observed in individuals treated with 1 mg of CNTX‐4975 across various parameters, including the Western Ontario and McMaster Universities (WOMAC) Osteoarthritis Index, pain while walking scores, joint stiffness and joint function when compared to the placebo group. These positive treatment effects were sustained for up to 24 weeks. This study found no significant difference in the incidence of treatment‐emergent adverse events between the placebo and CNTX‐4975 groups, indicating that the use of CNTX‐4975 does not result in serious safety concerns. 190
9. TRPV1 ANTAGONISTS
JNJ‐39439335 also known as mavatrep, functions as an antagonist for the TRPV1 receptor. A Phase 1b study was conducted in which individuals with OA were administered a single dose of 50 mg mavatrep, 500 mg naproxen twice daily or a placebo. The results demonstrated that the individuals treated with mavatrep showed significant pain reduction, as measured by pain after climbing stairs (PASC), Numerical Rating Scale (NRS) score and WOMAC score. However, some adverse events emerged in the mavatrep group, including cold sensation, decreased heat sensation, dysgeusia, paresthesia and heat sensation. 187 Another subsequent 10‐week clinical study observed that repeated administration of mavatrep did not lead to tolerance. 188
Another TRPV1 receptor antagonist, NEO6860, was studied in a Phase II trial involving individuals with OA. The individuals received a one‐day treatment (two doses) of NEO6860 (500 mg twice daily), naproxen (500 mg twice daily) or a placebo. The assessment of knee pain intensity revealed that individuals treated with NEO6860 and naproxen experienced less pain at 3 and 24 h after the staircase pain test. However, statistically significant differences between NEO6860 and placebo treatment were only observed at the 24 h time point. Adverse events were also reported in the NEO6860 treatment group, including headache, nausea, dizziness, fatigue, increased blood pressure/hypertension and hypoesthesia, which raised safety concerns. 189
While several TRPV1 agonists or antagonists have demonstrated improved efficacy in pre‐clinical and clinical trials for arthritis pain, attention needs to be given to adverse reactions, such as drug‐induced increases in core body temperature. TRPV1 channels in nociceptive sensory neurons can undergo conformational changes triggered by proton, thermal and chemical ligands. TRPV1 antagonists that have a minimal impact on proton‐induced gating can effectively avoid the adverse effects of hyperthermia. 193 , 194 Consequently, the multi‐steric gating feature of TRPV1 channels presents a new approach for developing potent TRPV1 antagonists without the risk of hyperthermia. 141 Additionally, exploring other therapeutic strategies, such as targeting both TRPA1 and TRPV1 or the fatty acid amide hydrolase and TRPV1, may yield more effective analgesic effects compared to TRPV1 antagonists alone. 167 , 195
10. CONCLUSION AND FUTURE DIRECTIONS
The TRPV1 channels play a crucial role in nociception during arthritis, and numerous studies have observed an upregulation of this channel on sensory nerves in relation to arthritis pain. TRPV1 channels are widely expressed in histiocytes, nerves and brain regions that are relevant to arthritis pathophysiology. They are involved in regulating chondrocyte ferroptosis, synovial inflammatory response, macrophage polarization and bone homeostasis (Figure 2). Additionally, TRPV1 channels play a vital role in nerve sensitization, defunctionalization or central sensitization, and they can release various neuropeptides that contribute to arthritis development. Both pre‐clinical and clinical data have indicated that TRPV1 agonists, when administered as a pre‐treatment, may have the potential to prevent or slow down the progression of OA. Consequently, targeting the TRPV1 channels holds significant promise as a therapeutic approach for arthritis treatment and the management of arthritic pain.
FIGURE 2.

The role of TRPV1 on different cell types of the joint during the onset and progression of OA. (A) The anti‐ferroptosis effect of capsaicin through the TRPV1‐Glutathione peroxidase 4 (GPX4) pathway in OA chondrocytes. Mechanical stress reduces intracellular Ca2+ concentration through TRPV1 channels leading to a cytokine‐induced decrease in A Disintegrin and Metalloproteinase with Thrombospondin Motif (ADAMTS) 9 expression. In addition, the activation of TRPV1 on chondrocytes can potentially induce an upregulation of TRPV1 gene expression. Similarly, the activation of TRPV1 on synovial cells can trigger the release of inflammatory factors, the production of reactive oxygen species (ROS) and an increase in TRPV1 gene expression. Conversely, inflammatory factors, ROS and neuropeptides promote TRPV1 activation. (B) Hyaluronic acid (HA) in synovial fluid inhibits the opening of TRPV1 channels and reduces Ca2+ influx into cells. (C) Capsaicin inhibits the polarization of M1 macrophages through TRPV1 channels, reduces the release of pro‐inflammatory factors and thus alleviates the progression of OA. (D) Inactivation of TRPV1 channels inhibits Ca2+ influx and the expression of its downstream transcription factor nuclear factor of activated T cells c1 (NFATc1), leading to reduced osteoclastogenesis, reducing bone loss in early OA.
Although great progress has been made in the research of TRPV1 channels in OA and RA, there are still some limitations. In order to make better use of TRPV1 channels for the treatment of arthritis, the following problems need to be addressed. First, the contribution of TRPV1 to angiogenesis and lymphangiogenesis in arthritis pathogenesis should be assessed. Synovial angiogenesis in RA and OA promotes the invasion of inflammatory cells and increases the distribution of joint pain receptors, accelerating the degeneration of articular cartilage and the occurrence of pain. 76 Some clinical drugs, such as simvastatin and erythropoietin, induce endothelial nitric oxide synthase phosphorylation and increase NO synthesis in endothelial cells by activating TRPV1‐mediated Ca2+ influx, ultimately promoting angiogenesis. 196 , 197 Lymphatic endothelial cells (LECs) express various acid‐sensing receptors, acidic conditions (pH = 6.4) induce IL‐8 expression through activation of TRPV1 in LECs, treatment with the TRPV1 antagonist I‐RTX inhibited the expression of IL‐8. 198 Serum IL‐8 plays a role in knee OA, and its expression levels are positively correlated with increased knee symptoms, infrapatellar fat pad signal intensity alteration, and serum levels of bone or cartilage biomarkers. 199 In addition, it has been reported that capsazepine could prevent the hypertonic environment‐induced decrease of contraction rate and lymph flow by blocking the TRPV1 channel, indicating that TRPV1 is particularly important in regulating the rate of spontaneous contraction of lymphatic vessels and lymph flow. 200 It is possible to alleviate arthritis pain by inhibiting angiogenesis and lymphangiogenesis through the inhibition of TRPV1 activity (Figure 3).
FIGURE 3.

The relationship between angiogenesis, lymphangiogenesis, innervation, pain and joint destruction. The schematic diagram of the hypothetical molecular mechanism of the therapeutic potential of TRPV1 agonists and antagonists in the treatment of OA and RA.
Consequently, unravelling the link between TRPV1 and angiogenesis or lymphangiogenesis in arthritis holds significant implications for comprehending the pharmacological mechanisms of TRPV1 and identifying novel therapeutic targets for arthritis. Additionally, TRPV1 plays multiple roles in arthritis. Activation of TRPV1 in cartilage protects chondrocytes from degeneration, while inhibition of TRPV1 in synovial tissue of joints inhibits synovial inflammatory responses. Therefore, targeted TRPV1 therapy should be carefully and pragmatically considered as the balance between the beneficial and the adverse effects, and the choice of treatment should be tailored to each patient's specific condition, disease severity and treatment goals. Selectively targeting specific tissue or spinal cord nerve endings using drugs may avoid some systemic adverse effects, such as targeting the expression of the TRPV1 channel using genome‐editing tools, which could block specific cell or certain populations of TRPV1+ neurons and thus be much better tolerated. MiR‐375 and miR‐455 were identified to repress TRPV1 expression via targeting the 3’‐UTR of TRPV1 mRNA, which has been shown to be downregulated in patients with low back pain with neuropathic pain, 201 so they may also function as therapeutic targets to indirectly modulate TRPV1 activity in arthritis. Furthermore, employing TRPV1‐RNAi as a potential therapeutic approach could effectively target the TRPV1 receptor to address pathological alterations in arthritis and preserve essential physiological defence mechanisms. Small interfering RNA (siRNA) therapy can not only target mRNA to silence the expression of catabolic genes to suppress the inflammatory response of arthritis but also silence the expression of genes that inhibit tissue regeneration by improving cartilage and bone tissue regeneration. 202 Several studies have shown that siRNA targeting TRPV1 can be used to treat dry eye disease, hyperalgesia induced by Complete Freund's Adjuvant and bone cancer pain. 203 , 204 , 205 Targeting TRPV1 using TRPV1 siRNA to reduce protein expression by inhibiting mRNA expression has become a method for evaluating gene‐specific activity. 206 Ultimately, the involvement of TRPV1 in arthritic conditions remains a subject of debate. It is crucial to exercise caution in the study design and result analysis when investigating TRPV1 channels, particularly in terms of agonist or antagonist dosage, treatment duration, TRPV1 expression distribution, potential splicing variants and functional implications. Taking these factors into account will contribute to a more comprehensive understanding of the role of TRPV1 in arthritic diseases.
In summary, the involvement of TRPV1 in the development of arthritis highlights its significance in the disease's pathogenesis. Further research and drug development efforts are essential to ensure the safety and effectiveness of TRPV1 as a therapeutic target. Anticipated future studies will shed more light on the precise role and regulatory mechanisms of TRPV1 in arthritis. This knowledge will establish a solid foundation for advancing targeted TRPV1 therapies and their clinical applications.
AUTHOR CONTRIBUTIONS
Di Chen, Liping Tong and Yan Chen contributed conception and design of the study; Zhidong Liao wrote the first draft of the manuscript and prepared the figures and tables; Muhammad Umar and Xingyun Huang wrote sections of the manuscript; Ling Qin, Guozhi Xiao, Liping Tong and Di Chen provided the critical revisions. All authors read and approved the final manuscript.
FUNDING INFORMATION
This work was supported by the National Key Research and Development Program of China (2021YFB3800800) to Liping Tong and Di Chen. This project was also supported by the National Natural Science Foundation of China (NSFC) grants 82030067 and 82250710174 to Di Chen. This project was also supported by the NSFC grant 82161160342 to Di Chen and Ling Qin.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
Liao Z, Umar M, Huang X, et al. Transient receptor potential vanilloid 1: A potential therapeutic target for the treatment of osteoarthritis and rheumatoid arthritis. Cell Prolif. 2024;57(3):e13569. doi: 10.1111/cpr.13569
[Correction added on 05 January 2024, after first online publication: An affiliation to First Affiliated Hospital of Guangxi Medical University, Guangxi, China has been added for Yan Chen and Zhidong Liao].
Contributor Information
Yan Chen, Email: cy003@connect.hku.hk.
Liping Tong, Email: lp.tong@siat.ac.cn.
Di Chen, Email: di.chen@siat.ac.cn.
REFERENCES
- 1. Palazzo C, Nguyen C, Lefevre‐Colau MM, et al. Risk factors and burden of osteoarthritis. Ann Phys Rehabil Med. 2016;59:134‐138. [DOI] [PubMed] [Google Scholar]
- 2. Huang WH, Ong TY, Fu SC, Yung SH. Prevalence of patellofemoral joint osteoarthritis after anterior cruciate ligament injury and associated risk factors: a systematic review. J Orthop Transl. 2020;22:14‐25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Sharma L. Osteoarthritis of the knee. N Engl J Med. 2021;384:51‐59. [DOI] [PubMed] [Google Scholar]
- 4. Smolen JS, Aletaha D, Barton A, et al. Rheumatoid arthritis. Nat Rev Dis Primers. 2018;4:18001. [DOI] [PubMed] [Google Scholar]
- 5. Pelletier JP, Martel‐Pelletier J, Rannou F, Cooper C. Efficacy and safety of oral NSAIDs and analgesics in the management of osteoarthritis: evidence from real‐life setting trials and surveys. Semin Arthritis Rheum. 2016;45:S22‐S27. [DOI] [PubMed] [Google Scholar]
- 6. Zeng C, Wei J, Persson MSM, et al. Relative efficacy and safety of topical non‐steroidal anti‐inflammatory drugs for osteoarthritis: a systematic review and network meta‐analysis of randomised controlled trials and observational studies. Br J Sports Med. 2018;52:642‐650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Salsitz EA. Chronic pain, chronic opioid addiction: a complex nexus. J Med Toxicol. 2016;12:54‐57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Ben Mrid R, Bouchmaa N, Ainani H, et al. Anti‐rheumatoid drugs advancements: new insights into the molecular treatment of rheumatoid arthritis. Biomed Pharmacother. 2022;151:113126. [DOI] [PubMed] [Google Scholar]
- 9. Wei M, Duan DM. Efficacy and safety of monoclonal antibodies targeting interleukin‐17 pathway for inflammatory arthritis: a meta‐analysis of randomized controlled clinical trials. Drug Des Dev Ther. 2016;10:2771‐2777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Yuan N, Yu GM, Liv D, et al. An emerging role of interleukin‐23 in rheumatoid arthritis. Immunopharm Immunot. 2019;41:185‐191. [DOI] [PubMed] [Google Scholar]
- 11. Thakur S, Riyaz B, Patil A, et al. Novel drug delivery systems for NSAIDs in management of rheumatoid arthritis: an overview. Biomed Pharmacother. 2018;106:1011‐1023. [DOI] [PubMed] [Google Scholar]
- 12. Montell C, Rubin GM. Molecular characterization of the drosophila trp locus: a putative integral membrane protein required for phototransduction. Neuron. 1989;2:1313‐1323. [DOI] [PubMed] [Google Scholar]
- 13. Clapham DE. TRP channels as cellular sensors. Nature. 2003;426:517‐524. [DOI] [PubMed] [Google Scholar]
- 14. Venkatachalam K, Montell C. TRP channels. Annu Rev Biochem. 2007;76:387‐417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Caterina MJ, Schumacher MA, Tominaga M, et al. The capsaicin receptor: a heat‐activated ion channel in the pain pathway. Nature. 1997;389:816‐824. [DOI] [PubMed] [Google Scholar]
- 16. Premkumar LS, Sikand P. TRPV1: a target for next generation analgesics. Curr Neuropharmacol. 2008;6:151‐163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Helyes Z, Szabo A, Nemeth J, et al. Antiinflammatory and analgesic effects of somatostatin released from capsaicin‐sensitive sensory nerve terminals in a Freund's adjuvant‐induced chronic arthritis model in the rat. Arthritis Rheum. 2004;50:1677‐1685. [DOI] [PubMed] [Google Scholar]
- 18. Pan YZ, Pan HL. Primary afferent stimulation differentially potentiates excitatory and inhibitory inputs to spinal lamina II outer and inner neurons. J Neurophysiol. 2004;91:2413‐2421. [DOI] [PubMed] [Google Scholar]
- 19. Mizumura K, Sugiura T, Katanosaka K, et al. Excitation and sensitization of nociceptors by bradykinin: what do we know? Exp Brain Res. 2009;196:53‐65. [DOI] [PubMed] [Google Scholar]
- 20. Takeba Y, Suzuki N, Takeno M, et al. Modulation of synovial cell function by somatostatin in patients with rheumatoid arthritis. Arthritis Rheum. 1997;40:2128‐2138. [DOI] [PubMed] [Google Scholar]
- 21. Gavenis K, Schumacher C, Schneider U, et al. Expression of ion channels of the TRP family in articular chondrocytes from osteoarthritic patients: changes between native and in vitro propagated chondrocytes. Mol Cell Biochem. 2009;321:135‐143. [DOI] [PubMed] [Google Scholar]
- 22. Engler A, Aeschlimann A, Simmen BR, et al. Expression of transient receptor potential vanilloid 1 (TRPV1) in synovial fibroblasts from patients with osteoarthritis and rheumatoid arthritis. Biochem Biophys Res Commun. 2007;359:884‐888. [DOI] [PubMed] [Google Scholar]
- 23. Kelly S, Chapman RJ, Woodhams S, et al. Increased function of pronociceptive TRPV1 at the level of the joint in a rat model of osteoarthritis pain. Ann Rheum Dis. 2015;74:252‐259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Lv Z, Xu X, Sun Z, et al. TRPV1 alleviates osteoarthritis by inhibiting M1 macrophage polarization via Ca(2+)/CaMKII/Nrf2 signaling pathway. Cell Death Dis. 2021;12:504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Menigoz A, Boudes M. The expression pattern of TRPV1 in brain. J Neurosci. 2011;31:13025‐13027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Rossi F, Siniscalco D, Luongo L, et al. The endovanilloid/endocannabinoid system in human osteoclasts: possible involvement in bone formation and resorption. Bone. 2009;44:476‐484. [DOI] [PubMed] [Google Scholar]
- 27. Idris AI, Landao‐Bassonga E, Ralston SH. The TRPV1 ion channel antagonist capsazepine inhibits osteoclast and osteoblast differentiation in vitro and ovariectomy induced bone loss in vivo. Bone. 2010;46:1089‐1099. [DOI] [PubMed] [Google Scholar]
- 28. Honore P, Wismer CT, Mikusa J, et al. A‐425619 [1‐isoquinolin‐5‐yl‐3‐(4‐trifluoromethyl‐benzyl)‐urea], a novel transient receptor potential type V1 receptor antagonist, relieves pathophysiological pain associated with inflammation and tissue injury in rats. J Pharmacol Exp Ther. 2005;314:410‐421. [DOI] [PubMed] [Google Scholar]
- 29. Deal CL, Schnitzer TJ, Lipstein E, et al. Treatment of arthritis with topical capsaicin: a double‐blind trial. Clin Ther. 1991;13:383‐395. [PubMed] [Google Scholar]
- 30. Anand P, Bley K. Topical capsaicin for pain management: therapeutic potential and mechanisms of action of the new high‐concentration capsaicin 8% patch. Br J Anaesth. 2011;107:490‐502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Webster LR, Peppin JF, Murphy FT, et al. Efficacy, safety, and tolerability of NGX‐4010, capsaicin 8% patch, in an open‐label study of patients with peripheral neuropathic pain. Diabetes Res Clin Pract. 2011;93:187‐197. [DOI] [PubMed] [Google Scholar]
- 32. Ghilardi JR, Rohrich H, Lindsay TH, et al. Selective blockade of the capsaicin receptor TRPV1 attenuates bone cancer pain. J Neurosci. 2005;25:3126‐3131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Keeble J, Russell F, Curtis B, et al. Involvement of transient receptor potential vanilloid 1 in the vascular and hyperalgesic components of joint inflammation. Arthritis Rheum. 2005;52:3248‐3256. [DOI] [PubMed] [Google Scholar]
- 34. Lv Z, Han J, Li J, et al. Single cell RNA‐seq analysis identifies ferroptotic chondrocyte cluster and reveals TRPV1 as an anti‐ferroptotic target in osteoarthritis. EBioMedicine. 2022;84:104258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Chen D. Osteoarthritis: A complicated joint disease requiring extensive studies with multiple approaches comment. J Orthop Transl. 2022;32:130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Teunissen M, Meij BP, Snel L, et al. The catabolic‐to‐anabolic shift seen in the canine osteoarthritic cartilage treated with knee joint distraction occurs after the distraction period. J Orthop Transl. 2023;38:44‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Yan WQ, Maimaitimin M, Zhao FY, et al. The transplantation of particulated juvenile allograft cartilage and synovium for the repair of meniscal defect in a lapine model. J Orthop Transl. 2022;33:72‐89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Zhang Y, Jordan JM. Epidemiology of osteoarthritis. Clin Geriatr Med. 2010;26:355‐369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Martel‐Pelletier J, Barr AJ, Cicuttini FM, et al. Osteoarthritis. Nat Rev Dis Primers. 2016;2:16072. [DOI] [PubMed] [Google Scholar]
- 40. Sellam J, Berenbaum F. Is osteoarthritis a metabolic disease? Jt Bone Spine. 2013;80:568‐573. [DOI] [PubMed] [Google Scholar]
- 41. Huang Z, Chen J, Ma J, et al. Effectiveness of low‐level laser therapy in patients with knee osteoarthritis: a systematic review and meta‐analysis. Osteoarth Cartil. 2015;23:1437‐1444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Piscitelli P, Iolascon G, di Tanna G, et al. Socioeconomic burden of total joint arthroplasty for symptomatic hip and knee osteoarthritis in the Italian population: a 5‐year analysis based on hospitalization records. Arthritis Care Res (Hoboken). 2012;64:1320‐1327. [DOI] [PubMed] [Google Scholar]
- 43. Huang Z, Ding C, Li T, Yu SP. Current status and future prospects for disease modification in osteoarthritis. Rheumatology (Oxford). 2018;57:iv108‐iv123. [DOI] [PubMed] [Google Scholar]
- 44. Bodden J, Joseph GB, Schiro S, et al. Opioid users show worse baseline knee osteoarthritis and faster progression of degenerative changes: a retrospective case‐control study based on data from the osteoarthritis initiative (OAI). Arthritis Res Ther. 2021;23:146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Nelson AE, Allen KD, Golightly YM, et al. A systematic review of recommendations and guidelines for the management of osteoarthritis: the chronic osteoarthritis management initiative of the U.S. bone and joint initiative. Semin Arthritis Rheum. 2014;43:701‐712. [DOI] [PubMed] [Google Scholar]
- 46. Liu SH, Dube CE, Eaton CB, et al. Longterm effectiveness of intraarticular injections on patient‐reported symptoms in knee osteoarthritis. J Rheumatol. 2018;45:1316‐1324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Wernecke C, Braun HJ, Dragoo JL. The effect of intra‐articular corticosteroids on articular cartilage: a systematic review. Orthop J Sports Med. 2015;3:2325967115581163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Skou ST, Roos EM, Laursen MB, et al. A randomized, controlled trial of total knee replacement. N Engl J Med. 2015;373:1597‐1606. [DOI] [PubMed] [Google Scholar]
- 49. Fibel KH, Hillstrom HJ, Halpern BC. State‐of‐the‐art management of knee osteoarthritis. World J Clin Cases. 2015;3:89‐101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Lundblad H, Kreicbergs A, Jansson KA. Prediction of persistent pain after total knee replacement for osteoarthritis. J Bone Joint Surg Br. 2008;90:166‐171. [DOI] [PubMed] [Google Scholar]
- 51. Scott DL, Wolfe F, Huizinga TW. Rheumatoid arthritis. Lancet. 2010;376:1094‐1108. [DOI] [PubMed] [Google Scholar]
- 52. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011;365:2205‐2219. [DOI] [PubMed] [Google Scholar]
- 53. Smolen JS, Aletaha D, Koeller M, et al. New therapies for treatment of rheumatoid arthritis. Lancet. 2007;370:1861‐1874. [DOI] [PubMed] [Google Scholar]
- 54. Weinblatt ME. Efficacy of methotrexate in rheumatoid arthritis. Br J Rheumatol. 1995;34(2):43‐48. [PubMed] [Google Scholar]
- 55. Nishimoto N, Miyasaka N, Yamamoto K, et al. Study of active controlled tocilizumab monotherapy for rheumatoid arthritis patients with an inadequate response to methotrexate (SATORI): significant reduction in disease activity and serum vascular endothelial growth factor by IL‐6 receptor inhibition therapy. Mod Rheumatol. 2009;19:12‐19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Smolen JS, Landewe R, Bijlsma J, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease‐modifying antirheumatic drugs: 2016 update. Ann Rheum Dis. 2017;76:960‐977. [DOI] [PubMed] [Google Scholar]
- 57. Spivey CA, Griffith J, Kaplan C, et al. A retrospective analysis of corticosteroid utilization before initiation of biologic DMARDs among patients with rheumatoid arthritis in the United States. Rheumatol Ther. 2018;5:255‐270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Liou YS, Lin TK, Chen HY, Jong GP. Medications associated with fracture risk in patients with rheumatoid arthritis. Ann Rheum Dis. 2021;80:e140. [DOI] [PubMed] [Google Scholar]
- 59. Wang Y, Zhao R, Gu Z, et al. Effects of glucocorticoids on osteoporosis in rheumatoid arthritis: a systematic review and meta‐analysis. Osteoporos Int. 2020;31:1401‐1409. [DOI] [PubMed] [Google Scholar]
- 60. Dunbar RP, Alexiades MM. Decision making in rheumatoid arthritis. Determining surgical priorities. Rheum Dis Clin North Am. 1998;24:35‐54. [DOI] [PubMed] [Google Scholar]
- 61. Charlier E, Relic B, Deroyer C, et al. Insights on molecular mechanisms of chondrocytes death in osteoarthritis. Int J Mol Sci. 2016;17:2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Kobayashi M, Squires GR, Mousa A, et al. Role of interleukin‐1 and tumor necrosis factor alpha in matrix degradation of human osteoarthritic cartilage. Arthritis Rheum. 2005;52:128‐135. [DOI] [PubMed] [Google Scholar]
- 63. Lefebvre V, Peeters‐Joris C, Vaes G. Modulation by interleukin 1 and tumor necrosis factor alpha of production of collagenase, tissue inhibitor of metalloproteinases and collagen types in differentiated and dedifferentiated articular chondrocytes. Biochim Biophys Acta. 1990;1052:366‐378. [DOI] [PubMed] [Google Scholar]
- 64. Valdes AM, De Wilde G, Doherty SA, et al. The Ile585Val TRPV1 variant is involved in risk of painful knee osteoarthritis. Ann Rheum Dis. 2011;70:1556‐1561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron‐dependent form of nonapoptotic cell death. Cell. 2012;149:1060‐1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Yao X, Sun K, Yu S, et al. Chondrocyte ferroptosis contribute to the progression of osteoarthritis. J Orthop Translat. 2021;27:33‐43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Miao Y, Chen Y, Xue F, et al. Contribution of ferroptosis and GPX4's dual functions to osteoarthritis progression. EBioMedicine. 2022;76:103847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Argote PF, Kaplan JT, Poon A, et al. Chondrocyte viability is lost during high‐rate impact loading by transfer of amplified strain, but not stress, to pericellular and cellular regions. Osteoarth Cartil. 2019;27:1822‐1830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Stove J, Huch K, Gunther KP, Scharf HP. Interleukin‐1beta induces different gene expression of stromelysin, aggrecan and tumor‐necrosis‐factor‐stimulated gene 6 in human osteoarthritic chondrocytes in vitro. Pathobiology. 2000;68:144‐149. [DOI] [PubMed] [Google Scholar]
- 70. Hsueh YH, Buddhakosai W, Le PN, et al. Therapeutic effect of induced pluripotent stem cell‐derived extracellular vesicles in an in vitro and in vivo osteoarthritis model. J Orthop Translat. 2023;38:141‐155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Ohtsuki T, Shinaoka A, Kumagishi‐Shinaoka K, et al. Mechanical strain attenuates cytokine‐induced ADAMTS9 expression via transient receptor potential vanilloid type 1. Exp Cell Res. 2019;383:111556. [DOI] [PubMed] [Google Scholar]
- 72. Waning J, Vriens J, Owsianik G, et al. A novel function of capsaicin‐sensitive TRPV1 channels: involvement in cell migration. Cell Calcium. 2007;42:17‐25. [DOI] [PubMed] [Google Scholar]
- 73. Somogyi CS, Matta C, Foldvari Z, et al. Polymodal transient receptor potential Vanilloid (TRPV) ion channels in chondrogenic cells. Int J Mol Sci. 2015;16:18412‐18438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Philp AM, Davis ET, Jones SW. Developing anti‐inflammatory therapeutics for patients with osteoarthritis. Rheumatology (Oxford). 2017;56:869‐881. [DOI] [PubMed] [Google Scholar]
- 75. Motta F, Barone E, Sica A, Selmi C. Inflammaging and osteoarthritis. Clin Rev Allergy Immunol. 2023;64:222‐238. [DOI] [PubMed] [Google Scholar]
- 76. MacDonald IJ, Liu SC, Su CM, et al. Implications of angiogenesis involvement in arthritis. Int J Mol Sci. 2018;19:2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Sanchez‐Lopez E, Coras R, Torres A, et al. Synovial inflammation in osteoarthritis progression. Nat Rev Rheumatol. 2022;18:258‐275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Guccione AA, Felson DT, Anderson JJ, et al. The effects of specific medical conditions on the functional limitations of elders in the Framingham study. Am J Public Health. 1994;84:351‐358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Ayral X, Pickering EH, Woodworth TG, et al. Synovitis: a potential predictive factor of structural progression of medial tibiofemoral knee osteoarthritis—results of a 1 year longitudinal arthroscopic study in 422 patients. Osteoarthritis Cartilage. 2005;13:361‐367. [DOI] [PubMed] [Google Scholar]
- 80. Ritchlin C. Fibroblast biology. Effector signals released by the synovial fibroblast in arthritis. Arthritis Res. 2000;2:356‐360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Szabo A, Helyes Z, Sandor K, et al. Role of transient receptor potential vanilloid 1 receptors in adjuvant‐induced chronic arthritis: in vivo study using gene‐deficient mice. J Pharmacol Exp Ther. 2005;314:111‐119. [DOI] [PubMed] [Google Scholar]
- 82. Hsieh WS, Kung CC, Huang SL, et al. TDAG8, TRPV1, and ASIC3 involved in establishing hyperalgesic priming in experimental rheumatoid arthritis. Sci Rep. 2017;7:8870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Westlund KN, Kochukov MY, Lu Y, McNearney TA. Impact of central and peripheral TRPV1 and ROS levels on proinflammatory mediators and nociceptive behavior. Mol Pain. 2010;6:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Terenzi R, Romano E, Manetti M, et al. Neuropeptides activate TRPV1 in rheumatoid arthritis fibroblast‐like synoviocytes and foster IL‐6 and IL‐8 production. Ann Rheum Dis. 2013;72:1107‐1109. [DOI] [PubMed] [Google Scholar]
- 85. Hu F, Sun WW, Zhao XT, et al. TRPV1 mediates cell death in rat synovial fibroblasts through calcium entry‐dependent ROS production and mitochondrial depolarization. Biochem Biophys Res Commun. 2008;369:989‐993. [DOI] [PubMed] [Google Scholar]
- 86. Logashina YA, Palikova YA, Palikov VA, et al. Anti‐inflammatory and analgesic effects of TRPV1 polypeptide modulator APHC3 in models of osteo‐ and rheumatoid arthritis. Mar Drugs. 2021;19:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Daghestani HN, Pieper CF, Kraus VB. Soluble macrophage biomarkers indicate inflammatory phenotypes in patients with knee osteoarthritis. Arthritis Rheumatol. 2015;67:956‐965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Caires R, Luis E, Taberner FJ, et al. Hyaluronan modulates TRPV1 channel opening, reducing peripheral nociceptor activity and pain. Nat Commun. 2015;6:8095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Gomis A, Pawlak M, Balazs EA, et al. Effects of different molecular weight elastoviscous hyaluronan solutions on articular nociceptive afferents. Arthritis Rheum. 2004;50:314‐326. [DOI] [PubMed] [Google Scholar]
- 90. Evanko SP, Tammi MI, Tammi RH, Wight TN. Hyaluronan‐dependent pericellular matrix. Adv Drug Deliv Rev. 2007;59:1351‐1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Balazs EA, Denlinger JL. Clinical uses of hyaluronan. Ciba Found Symp. 1989;143:265‐275. discussion 275–280, 281–265. [DOI] [PubMed] [Google Scholar]
- 92. Lieben L, Carmeliet G. The involvement of TRP channels in bone homeostasis. Front Endocrinol (Lausanne). 2012;3:99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Henriksen K, Neutzsky‐Wulff AV, Bonewald LF, Karsdal MA. Local communication on and within bone controls bone remodeling. Bone. 2009;44:1026‐1033. [DOI] [PubMed] [Google Scholar]
- 94. Plotkin LI, Gortazar AR, Davis HM, et al. Inhibition of osteocyte apoptosis prevents the increase in osteocytic receptor activator of nuclear factor kappaB ligand (RANKL) but does not stop bone resorption or the loss of bone induced by unloading. J Biol Chem. 2015;290:18934‐18942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Burr DB. Anatomy and physiology of the mineralized tissues: role in the pathogenesis of osteoarthrosis. Osteoarth Cartil. 2004;12:S20‐S30. [DOI] [PubMed] [Google Scholar]
- 96. Kovacs B, Vajda E, Nagy EE. Regulatory effects and interactions of the Wnt and OPG‐RANKL‐RANK signaling at the bone‐cartilage interface in osteoarthritis. Int J Mol Sci. 2019;20:4653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Takeuchi T, Tanaka Y, Soen S, et al. Effects of the anti‐RANKL antibody denosumab on joint structural damage in patients with rheumatoid arthritis treated with conventional synthetic disease‐modifying antirheumatic drugs (DESIRABLE study): a randomised, double‐blind, placebo‐controlled phase 3 trial. Ann Rheum Dis. 2019;78:899‐907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Lu K, Ma F, Yi D, et al. Molecular signaling in temporomandibular joint osteoarthritis. J Orthop Transl. 2022;32:21‐27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Rossi F, Bellini G, Torella M, et al. The genetic ablation or pharmacological inhibition of TRPV1 signalling is beneficial for the restoration of quiescent osteoclast activity in ovariectomized mice. Br J Pharmacol. 2014;171:2621‐2630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. He LH, Liu M, He Y, et al. TRPV1 deletion impaired fracture healing and inhibited osteoclast and osteoblast differentiation. Sci Rep. 2017;7:42385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Kuroda Y, Hisatsune C, Nakamura T, et al. Osteoblasts induce Ca2+ oscillation‐independent NFATc1 activation during osteoclastogenesis. Proc Natl Acad Sci U S A. 2008;105:8643‐8648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Kun J, Helyes Z, Perkecz A, et al. Effect of surgical and chemical sensory denervation on non‐neural expression of the transient receptor potential vanilloid 1 (TRPV1) receptors in the rat. J Mol Neurosci. 2012;48:795‐803. [DOI] [PubMed] [Google Scholar]
- 103. Kalff KM, El Mouedden M, van Egmond J, et al. Pre‐treatment with capsaicin in a rat osteoarthritis model reduces the symptoms of pain and bone damage induced by monosodium iodoacetate. Eur J Pharmacol. 2010;641:108‐113. [DOI] [PubMed] [Google Scholar]
- 104. Borbely E, Kiss T, Szabadfi K, et al. Complex role of capsaicin‐sensitive afferents in the collagen antibody‐induced autoimmune arthritis of the mouse. Sci Rep. 2018;8:15916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Farr M, Garvey K, Bold AM, et al. Significance of the hydrogen ion concentration in synovial fluid in rheumatoid arthritis. Clin Exp Rheumatol. 1985;3:99‐104. [PubMed] [Google Scholar]
- 106. Zu SQ, Feng YB, Zhu CJ, et al. Acid‐sensing ion channel 1a mediates acid‐induced pyroptosis through calpain‐2/calcineurin pathway in rat articular chondrocytes. Cell Biol Int. 2020;44:2140‐2152. [DOI] [PubMed] [Google Scholar]
- 107. Tominaga M, Tominaga T. Structure and function of TRPV1. Pflugers Arch. 2005;451:143‐150. [DOI] [PubMed] [Google Scholar]
- 108. Hu F, Yang S, Zhao D, et al. Moderate extracellular acidification inhibits capsaicin‐induced cell death through regulating calcium mobilization, NF‐kappaB translocation and ROS production in synoviocytes. Biochem Biophys Res Commun. 2012;424:196‐200. [DOI] [PubMed] [Google Scholar]
- 109. Reni C, Mangialardi G, Meloni M, Madeddu P. Diabetes stimulates osteoclastogenesis by acidosis‐induced activation of transient receptor potential cation channels. Sci Rep. 2016;6:30639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Rossi F, Bellini G, Luongo L, et al. The endovanilloid/endocannabinoid system: a new potential target for osteoporosis therapy. Bone. 2011;48:997‐1007. [DOI] [PubMed] [Google Scholar]
- 111. Caterina MJ, Leffler A, Malmberg AB, et al. Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science. 2000;288:306‐313. [DOI] [PubMed] [Google Scholar]
- 112. Davis JB, Gray J, Gunthorpe MJ, et al. Vanilloid receptor‐1 is essential for inflammatory thermal hyperalgesia. Nature. 2000;405:183‐187. [DOI] [PubMed] [Google Scholar]
- 113. Barton NJ, McQueen DS, Thomson D, et al. Attenuation of experimental arthritis in TRPV1R knockout mice. Exp Mol Pathol. 2006;81:166‐170. [DOI] [PubMed] [Google Scholar]
- 114. Cui M, Honore P, Zhong C, et al. TRPV1 receptors in the CNS play a key role in broad‐spectrum analgesia of TRPV1 antagonists. J Neurosci. 2006;26:9385‐9393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Tominaga M, Caterina MJ, Malmberg AB, et al. The cloned capsaicin receptor integrates multiple pain‐producing stimuli. Neuron. 1998;21:531‐543. [DOI] [PubMed] [Google Scholar]
- 116. Morgan M, Nencini S, Thai J, Ivanusic JJ. TRPV1 activation alters the function of Adelta and C fiber sensory neurons that innervate bone. Bone. 2019;123:168‐175. [DOI] [PubMed] [Google Scholar]
- 117. Cho WG, Valtschanoff JG. Vanilloid receptor TRPV1‐positive sensory afferents in the mouse ankle and knee joints. Brain Res. 2008;1219:59‐65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Ioi H, Kido MA, Zhang JQ, et al. Capsaicin receptor expression in the rat temporomandibular joint. Cell Tissue Res. 2006;325:47‐54. [DOI] [PubMed] [Google Scholar]
- 119. Fernihough J, Gentry C, Bevan S, Winter J. Regulation of calcitonin gene‐related peptide and TRPV1 in a rat model of osteoarthritis. Neurosci Lett. 2005;388:75‐80. [DOI] [PubMed] [Google Scholar]
- 120. Lotz M, Vaughan JH, Carson DA. Effect of neuropeptides on production of inflammatory cytokines by human monocytes. Science. 1988;241:1218‐1221. [DOI] [PubMed] [Google Scholar]
- 121. McDougall JJ, Karimian SM, Ferrell WR. Prolonged alteration of vasoconstrictor and vasodilator responses in rat knee joints by adjuvant monoarthritis. Exp Physiol. 1995;80:349‐357. [DOI] [PubMed] [Google Scholar]
- 122. Zhang L, Hoff AO, Wimalawansa SJ, et al. Arthritic calcitonin/alpha calcitonin gene‐related peptide knockout mice have reduced nociceptive hypersensitivity. Pain. 2001;89:265‐273. [DOI] [PubMed] [Google Scholar]
- 123. Schaible HG, Ebersberger A, von Banchet GS. Mechanisms of pain in arthritis. Ann N Y Acad Sci. 2002;966:343‐354. [DOI] [PubMed] [Google Scholar]
- 124. Holzer P. Local effector functions of capsaicin‐sensitive sensory nerve endings: involvement of tachykinins, calcitonin gene‐related peptide and other neuropeptides. Neuroscience. 1988;24:739‐768. [DOI] [PubMed] [Google Scholar]
- 125. Meng J, Ovsepian SV, Wang J, et al. Activation of TRPV1 mediates calcitonin gene‐related peptide release, which excites trigeminal sensory neurons and is attenuated by a retargeted botulinum toxin with anti‐nociceptive potential. J Neurosci. 2009;29:4981‐4992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Kim Y, Kim EH, Lee KS, et al. The effects of intra‐articular resiniferatoxin on monosodium iodoacetate‐induced osteoarthritic pain in rats. Korean J Physiol Pharmacol. 2016;20:129‐136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Puttfarcken PS, Han P, Joshi SK, et al. A‐995662 [(R)‐8‐(4‐methyl‐5‐(4‐(trifluoromethyl)phenyl)oxazol‐2‐ylamino)‐1,2,3,4‐tetrahydronaphthalen‐2‐ol], a novel, selective TRPV1 receptor antagonist, reduces spinal release of glutamate and CGRP in a rat knee joint pain model. Pain. 2010;150:319‐326. [DOI] [PubMed] [Google Scholar]
- 128. Latremoliere A, Woolf CJ. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J Pain. 2009;10:895‐926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. David‐Pereira A, Puga S, Goncalves S, et al. Metabotropic glutamate 5 receptor in the infralimbic cortex contributes to descending pain facilitation in healthy and arthritic animals. Neuroscience. 2016;312:108‐119. [DOI] [PubMed] [Google Scholar]
- 130. David‐Pereira A, Sagalajev B, Wei H, et al. The medullary dorsal reticular nucleus as a relay for descending pronociception induced by the mGluR5 in the rat infralimbic cortex. Neuroscience. 2017;349:341‐354. [DOI] [PubMed] [Google Scholar]
- 131. Firner M, Greffrath W, Treede RD. Phosphorylation of extracellular signal‐related protein kinase is required for rapid facilitation of heat‐induced currents in rat dorsal root ganglion neurons. Neuroscience. 2006;143:253‐263. [DOI] [PubMed] [Google Scholar]
- 132. Cruz CD, Neto FL, Castro‐Lopes J, et al. Inhibition of ERK phosphorylation decreases nociceptive behaviour in monoarthritic rats. Pain. 2005;116:411‐419. [DOI] [PubMed] [Google Scholar]
- 133. Chen Y, Willcockson HH, Valtschanoff JG. Vanilloid receptor TRPV1‐mediated phosphorylation of ERK in murine adjuvant arthritis. Osteoarth Cartil. 2009;17:244‐251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Pan HL, Zhang YQ, Zhao ZQ. Involvement of lysophosphatidic acid in bone cancer pain by potentiation of TRPV1 via PKCepsilon pathway in dorsal root ganglion neurons. Mol Pain. 2010;6:85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Lee SY, Lee JH, Kang KK, et al. Sensitization of vanilloid receptor involves an increase in the phosphorylated form of the channel. Arch Pharm Res. 2005;28:405‐412. [DOI] [PubMed] [Google Scholar]
- 136. Premkumar LS, Ahern GP. Induction of vanilloid receptor channel activity by protein kinase C. Nature. 2000;408:985‐990. [DOI] [PubMed] [Google Scholar]
- 137. Mandadi S, Tominaga T, Numazaki M, et al. Increased sensitivity of desensitized TRPV1 by PMA occurs through PKCepsilon‐mediated phosphorylation at S800. Pain. 2006;123:106‐116. [DOI] [PubMed] [Google Scholar]
- 138. Koda K, Hyakkoku K, Ogawa K, et al. Sensitization of TRPV1 by protein kinase C in rats with mono‐iodoacetate‐induced joint pain. Osteoarthr Cartil. 2016;24:1254‐1262. [DOI] [PubMed] [Google Scholar]
- 139. Lewin GR, Ritter AM, Mendell LM. Nerve growth factor‐induced hyperalgesia in the neonatal and adult rat. J Neurosci. 1993;13:2136‐2148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Huang J, Zhang X, McNaughton PA. Inflammatory pain: the cellular basis of heat hyperalgesia. Curr Neuropharmacol. 2006;4:197‐206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Szolcsanyi J, Sandor Z. Multisteric TRPV1 nocisensor: a target for analgesics. Trends Pharmacol Sci. 2012;33:646‐655. [DOI] [PubMed] [Google Scholar]
- 142. Kissin EY, Freitas CF, Kissin I. The effects of intraarticular resiniferatoxin in experimental knee‐joint arthritis. Anesth Analg. 2005;101:1433‐1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Honore P, Chandran P, Hernandez G, et al. Repeated dosing of ABT‐102, a potent and selective TRPV1 antagonist, enhances TRPV1‐mediated analgesic activity in rodents, but attenuates antagonist‐induced hyperthermia. Pain. 2009;142:27‐35. [DOI] [PubMed] [Google Scholar]
- 144. Chu KL, Chandran P, Joshi SK, et al. TRPV1‐related modulation of spinal neuronal activity and behavior in a rat model of osteoarthritic pain. Brain Res. 2011;1369:158‐166. [DOI] [PubMed] [Google Scholar]
- 145. Okun A, Liu P, Davis P, et al. Afferent drive elicits ongoing pain in a model of advanced osteoarthritis. Pain. 2012;153:924‐933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Abdullah M, Mahowald ML, Frizelle SP, et al. The effect of intra‐articular vanilloid receptor agonists on pain behavior measures in a murine model of acute monoarthritis. J Pain Res. 2016;9:563‐570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Iadarola MJ, Sapio MR, Raithel SJ, et al. Long‐term pain relief in canine osteoarthritis by a single intra‐articular injection of resiniferatoxin, a potent TRPV1 agonist. Pain. 2018;159:2105‐2114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Hwang HS, Park IY, Hong JI, et al. Comparison of joint degeneration and pain in male and female mice in DMM model of osteoarthritis. Osteoarth Cartil. 2021;29:728‐738. [DOI] [PubMed] [Google Scholar]
- 149. Campbell JN, Stevens R, Hanson P, et al. Injectable capsaicin for the management of pain due to osteoarthritis. Molecules. 2021;26:778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Colpaert FC, Donnerer J, Lembeck F. Effects of capsaicin on inflammation and on the substance P content of nervous tissues in rats with adjuvant arthritis. Life Sci. 1983;32:1827‐1834. [DOI] [PubMed] [Google Scholar]
- 151. Jancso G, Santha P. The foundation of sensory pharmacology: Nicholas (Miklos) Jancso and the Szeged contribution. Temperature (Austin). 2015;2:152‐157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Szolcsanyi J, Pinter E. Transient receptor potential vanilloid 1 as a therapeutic target in analgesia. Expert Opin Ther Targets. 2013;17:641‐657. [DOI] [PubMed] [Google Scholar]
- 153. Holzer P. Capsaicin: cellular targets, mechanisms of action, and selectivity for thin sensory neurons. Pharmacol Rev. 1991;43:143‐201. [PubMed] [Google Scholar]
- 154. Jeffry JA, Yu SQ, Sikand P, et al. Selective targeting of TRPV1 expressing sensory nerve terminals in the spinal cord for long lasting analgesia. PLoS One. 2009;4:e7021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Yaksh TL, Farb DH, Leeman SE, Jessell TM. Intrathecal capsaicin depletes substance P in the rat spinal cord and produces prolonged thermal analgesia. Science. 1979;206:481‐483. [DOI] [PubMed] [Google Scholar]
- 156. Brown DC, Iadarola MJ, Perkowski SZ, et al. Physiologic and antinociceptive effects of intrathecal resiniferatoxin in a canine bone cancer model. Anesthesiology. 2005;103:1052‐1059. [DOI] [PubMed] [Google Scholar]
- 157. Arora V, Campbell JN, Chung MK. Fight fire with fire: neurobiology of capsaicin‐induced analgesia for chronic pain. Pharmacol Ther. 2021;220:107743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Chung MK, Campbell JN. Use of capsaicin to treat pain: mechanistic and therapeutic considerations. Pharmaceuticals (Basel). 2016;9:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Jancso G, Kiraly E, Joo F, et al. Selective degeneration by capsaicin of a subpopulation of primary sensory neurons in the adult rat. Neurosci Lett. 1985;59:209‐214. [DOI] [PubMed] [Google Scholar]
- 160. Szolcsanyi J, Jancso‐Gabor A, Joo F. Functional and fine structural characteristics of the sensory neuron blocking effect of capsaicin. Naunyn Schmiedebergs Arch Pharmacol. 1975;287:157‐169. [DOI] [PubMed] [Google Scholar]
- 161. Docherty RJ, Yeats JC, Piper AS. Capsazepine block of voltage‐activated calcium channels in adult rat dorsal root ganglion neurones in culture. Br J Pharmacol. 1997;121:1461‐1467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Liu L, Simon SA. Capsazepine, a vanilloid receptor antagonist, inhibits nicotinic acetylcholine receptors in rat trigeminal ganglia. Neuroscience Letters. 1997;228:29‐32. [DOI] [PubMed] [Google Scholar]
- 163. Nocerino E, Izzo AA, Borrelli F, et al. Relaxant effect of capsazepine in the isolated rat ileum. N‐S Arch Pharmacol. 2002;365:187‐192. [DOI] [PubMed] [Google Scholar]
- 164. Ahmed M, Bjurholm A, Srinivasan GR, et al. Capsaicin effects on substance P and CGRP in rat adjuvant arthritis. Regul Pept. 1995;55:85‐102. [DOI] [PubMed] [Google Scholar]
- 165. Miller F, Bjornsson M, Svensson O, Karlsten R. Experiences with an adaptive design for a dose‐finding study in patients with osteoarthritis. Contemp Clin Trials. 2014;37:189‐199. [DOI] [PubMed] [Google Scholar]
- 166. Angeby Moller K, Berge OG, Finn A, et al. Using gait analysis to assess weight bearing in rats with Freund's complete adjuvant‐induced monoarthritis to improve predictivity: interfering with the cyclooxygenase and nerve growth factor pathways. Eur J Pharmacol. 2015;756:75‐84. [DOI] [PubMed] [Google Scholar]
- 167. Malek N, Mrugala M, Makuch W, et al. A multi‐target approach for pain treatment: dual inhibition of fatty acid amide hydrolase and TRPV1 in a rat model of osteoarthritis. Pain. 2015;156:890‐903. [DOI] [PubMed] [Google Scholar]
- 168. Gavva NR, Treanor JJ, Garami A, et al. Pharmacological blockade of the vanilloid receptor TRPV1 elicits marked hyperthermia in humans. Pain. 2008;136:202‐210. [DOI] [PubMed] [Google Scholar]
- 169. Cathcart CJ, Johnston SA, Reynolds LR, et al. Efficacy of ABT‐116, an antagonist of transient receptor potential vanilloid type 1, in providing analgesia for dogs with chemically induced synovitis. Am J Vet Res. 2012;73:19‐26. [DOI] [PubMed] [Google Scholar]
- 170. Round P, Priestley A, Robinson J. An investigation of the safety and pharmacokinetics of the novel TRPV1 antagonist XEN‐D0501 in healthy subjects. Br J Clin Pharmacol. 2011;72:921‐931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Steiner AA, Turek VF, Almeida MC, et al. Nonthermal activation of transient receptor potential vanilloid‐1 channels in abdominal viscera tonically inhibits autonomic cold‐defense effectors. J Neurosci. 2007;27:7459‐7468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Iida T, Shimizu I, Nealen ML, et al. Attenuated fever response in mice lacking TRPV1. Neurosci Lett. 2005;378:28‐33. [DOI] [PubMed] [Google Scholar]
- 173. Zhang WY, Po ALW. The effectiveness of topically applied capsaicin. A meta‐analysis. Eur J Clin Pharmacol. 1994;46:517‐522. [DOI] [PubMed] [Google Scholar]
- 174. Lu M, Chen C, Lan Y, et al. Capsaicin‐the major bioactive ingredient of chili peppers: bio‐efficacy and delivery systems. Food Funct. 2020;11:2848‐2860. [DOI] [PubMed] [Google Scholar]
- 175. Iftinca M, Defaye M, Altier C. TRPV1‐targeted drugs in development for human pain conditions. Drugs. 2021;81:7‐27. [DOI] [PubMed] [Google Scholar]
- 176. Chen CC, Chang C, Hsu YH, et al. fMRI indicates cortical activation through TRPV1 modulation during acute gouty attacks. Sci Rep. 2019;9:12348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Xu XJ, Farkas‐Szallasi T, Lundberg JM, et al. Effects of the capsaicin analogue resiniferatoxin on spinal nociceptive mechanisms in the rat: behavioral, electrophysiological and in situ hybridization studies. Brain Res. 1997;752:52‐60. [DOI] [PubMed] [Google Scholar]
- 178. Jensen MT, Petersen KL. Gender differences in pain and secondary hyperalgesia after heat/capsaicin sensitization in healthy volunteers. J Pain. 2006;7:211‐217. [DOI] [PubMed] [Google Scholar]
- 179. Loyd DR, Sun XX, Locke EE, et al. Sex differences in serotonin enhancement of capsaicin‐evoked calcitonin gene‐related peptide release from human dental pulp. Pain. 2012;153:2061‐2067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Gazerani P, Andersen OK, Arendt‐Nielsen L. Site‐specific, dose‐dependent, and sex‐related responses to the experimental pain model induced by intradermal injection of capsaicin to the foreheads and forearms of healthy humans. J Orofac Pain. 2007;21:289‐302. [PubMed] [Google Scholar]
- 181. Bai X, Zhang X, Zhou Q. Effect of testosterone on TRPV1 expression in a model of orofacial myositis pain in the rat. J Mol Neurosci. 2018;64:93‐101. [DOI] [PubMed] [Google Scholar]
- 182. Rage M, Van Acker N, Facer P, et al. The time course of CO2 laser‐evoked responses and of skin nerve fibre markers after topical capsaicin in human volunteers. Clin Neurophysiol. 2010;121:1256‐1266. [DOI] [PubMed] [Google Scholar]
- 183. Salas MM, Hargreaves KM, Akopian AN. TRPA1‐mediated responses in trigeminal sensory neurons: interaction between TRPA1 and TRPV1. Eur J Neurosci. 2009;29:1568‐1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Bautista DM, Movahed P, Hinman A, et al. Pungent products from garlic activate the sensory ion channel TRPA1. Proc Natl Acad Sci U S A. 2005;102:12248‐12252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. McCleane G. The analgesic efficacy of topical capsaicin is enhanced by glyceryl trinitrate in painful osteoarthritis: a randomized, double blind, placebo controlled study. Eur J Pain. 2000;4:355‐360. [DOI] [PubMed] [Google Scholar]
- 186. Schnitzer TJ, Pelletier JP, Haselwood DM, et al. Civamide cream 0.075% in patients with osteoarthritis of the knee: a 12‐week randomized controlled clinical trial with a longterm extension. J Rheumatol. 2012;39:610‐620. [DOI] [PubMed] [Google Scholar]
- 187. Mayorga AJ, Flores CM, Trudeau JJ, et al. A randomized study to evaluate the analgesic efficacy of a single dose of the TRPV1 antagonist mavatrep in patients with osteoarthritis. Scand J Pain. 2017;17:134‐143. [DOI] [PubMed] [Google Scholar]
- 188. Manitpisitkul P, Flores CM, Moyer JA, et al. A multiple‐dose double‐blind randomized study to evaluate the safety, pharmacokinetics, pharmacodynamics and analgesic efficacy of the TRPV1 antagonist JNJ‐39439335 (mavatrep). Scand J Pain. 2018;18:151‐164. [DOI] [PubMed] [Google Scholar]
- 189. Arsenault P, Chiche D, Brown W, et al. NEO6860, modality‐selective TRPV1 antagonist: a randomized, controlled, proof‐of‐concept trial in patients with osteoarthritis knee pain. Pain Rep. 2018;3:e696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Stevens RM, Ervin J, Nezzer J, et al. Randomized, double‐blind, placebo‐controlled trial of intraarticular trans‐capsaicin for pain associated with osteoarthritis of the knee. Arthritis Rheumatol. 2019;71:1524‐1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. McCarthy GM, McCarty DJ. Effect of topical capsaicin in the therapy of painful osteoarthritis of the hands. J Rheumatol. 1992;19:604‐607. [PubMed] [Google Scholar]
- 192. Persson MSM, Stocks J, Walsh DA, et al. The relative efficacy of topical non‐steroidal anti‐inflammatory drugs and capsaicin in osteoarthritis: a network meta‐analysis of randomised controlled trials. Osteoarthr Cartil. 2018;26:1575‐1582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Garami A, Shimansky YP, Pakai E, et al. Contributions of different modes of TRPV1 activation to TRPV1 antagonist‐induced hyperthermia. J Neurosci. 2010;30:1435‐1440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Reilly RM, McDonald HA, Puttfarcken PS, et al. Pharmacology of modality‐specific transient receptor potential vanilloid‐1 antagonists that do not alter body temperature. J Pharmacol Exp Ther. 2012;342:416‐428. [DOI] [PubMed] [Google Scholar]
- 195. Trevisan G, Hoffmeister C, Rossato MF, et al. Transient receptor potential ankyrin 1 receptor stimulation by hydrogen peroxide is critical to trigger pain during monosodium urate‐induced inflammation in rodents. Arthritis Rheum. 2013;65:2984‐2995. [DOI] [PubMed] [Google Scholar]
- 196. Su KH, Lin SJ, Wei J, et al. The essential role of transient receptor potential vanilloid 1 in simvastatin‐induced activation of endothelial nitric oxide synthase and angiogenesis. Acta Physiol (Oxf). 2014;212:191‐204. [DOI] [PubMed] [Google Scholar]
- 197. Yu YB, Su KH, Kou YR, et al. Role of transient receptor potential vanilloid 1 in regulating erythropoietin‐induced activation of endothelial nitric oxide synthase. Acta Physiol (Oxf). 2017;219:465‐477. [DOI] [PubMed] [Google Scholar]
- 198. Nakanishi M, Morita Y, Hata K, Muragaki Y. Acidic microenvironments induce lymphangiogenesis and IL‐8 production via TRPV1 activation in human lymphatic endothelial cells. Exp Cell Res. 2016;345:180‐189. [DOI] [PubMed] [Google Scholar]
- 199. Ruan GF, Xu JH, Wang K, et al. Associations between serum IL‐8 and knee symptoms, joint structures, and cartilage or bone biomarkers in patients with knee osteoarthritis. Clin Rheumatol. 2019;38:3609‐3617. [DOI] [PubMed] [Google Scholar]
- 200. Solari E, Marcozzi C, Negrini D, Moriondo A. Fluid osmolarity modulates the rate of spontaneous contraction of lymphatic vessels and lymph flow by means of a cooperation between TRPV and VRAC channels. Biology‐Basel. 2023;12:1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Li Z, Zhou Y, Li Z. NFKB1 signalling activation contributes to TRPV1 over‐expression via repressing MiR‐375 and MiR‐455: a study on neuropathic low back pain. Folia Biol (Praha). 2022;68:105‐111. [DOI] [PubMed] [Google Scholar]
- 202. Mottaghitalab F, Rastegari A, Farokhi M, et al. Prospects of siRNA applications in regenerative medicine. Int J Pharm. 2017;524:312‐329. [DOI] [PubMed] [Google Scholar]
- 203. Benitez‐Del‐Castillo JM, Moreno‐Montanes J, Jimenez‐Alfaro I, et al. Safety and efficacy clinical trials for SYL1001, a novel short interfering RNA for the treatment of dry eye disease. Invest Ophthalmol Vis Sci. 2016;57:6447‐6454. [DOI] [PubMed] [Google Scholar]
- 204. Kasama S, Kawakubo M, Suzuki T, et al. RNA interference‐mediated knock‐down of transient receptor potential vanilloid 1 prevents forepaw inflammatory hyperalgesia in rat. Eur J Neurosci. 2007;25:2956‐2963. [DOI] [PubMed] [Google Scholar]
- 205. Zhang S, Zhao J, Meng Q. AAV‐mediated siRNA against TRPV1 reduces nociception in a rat model of bone cancer pain. Neurol Res. 2019;41:972‐979. [DOI] [PubMed] [Google Scholar]
- 206. Akhilesh, Uniyal A, Gadepalli A, et al. Unlocking the potential of TRPV1 based siRNA therapeutics for the treatment of chemotherapy‐induced neuropathic pain. Life Sci. 2022;288:120187. [DOI] [PubMed] [Google Scholar]
