Hostname: page-component-848d4c4894-m9kch Total loading time: 0 Render date: 2024-05-20T09:32:50.137Z Has data issue: false hasContentIssue false

Roles of TRP and PIEZO receptors in autoimmune diseases

Published online by Cambridge University Press:  25 April 2024

Yang Baoqi
Affiliation:
Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
Ma Dan
Affiliation:
Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
Zhu Xueqing
Affiliation:
Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
Wu Zewen
Affiliation:
Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
An Qi
Affiliation:
Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
Zhao Jingwen
Affiliation:
Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
Gao Xinnan
Affiliation:
Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
Zhang Liyun*
Affiliation:
Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
*
Corresponding author: Zhang Liyun; Email: 1315710223@qq.com
Rights & Permissions [Opens in a new window]

Abstract

Autoimmune diseases are pathological autoimmune reactions in the body caused by various factors, which can lead to tissue damage and organ dysfunction. They can be divided into organ-specific and systemic autoimmune diseases. These diseases usually involve various body systems, including the blood, muscles, bones, joints and soft tissues. The transient receptor potential (TRP) and PIEZO receptors, which resulted in David Julius and Ardem Patapoutian winning the Nobel Prize in Physiology or Medicine in 2021, attracted people's attention. Most current studies on TRP and PIEZO receptors in autoimmune diseases have been carried out on animal model, only few clinical studies have been conducted. Therefore, this study aimed to review existing studies on TRP and PIEZO to understand the roles of these receptors in autoimmune diseases, which may help elucidate novel treatment strategies.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Author(s), 2024. Published by Cambridge University Press

Introduction

Autoimmune diseases refer to clinical disorders caused by internal and external factors, such as genetic and environmental factors. These lead to the destruction of autoimmune tolerance or abnormal regulation of autoimmune cells, and the continuous response of the immune system to self-antigens, resulting in damage or dysfunction. Autoimmune diseases are divided into organ-specific and systemic autoimmune diseases. Organ-specific autoimmune diseases include Hashimoto's thyroiditis, exophthalmic goitre and insulin-dependent diabetes mellitus. Systemic autoimmune diseases include rheumatoid arthritis (RA), osteoarthritis (OA), systemic lupus erythematosus (SLE), and Sjögren's syndrome (SS). The 2021 Nobel Prize in Physiology or Medicine drew people's attention to the transient receptor potential (TRP) ion channels and PIEZO receptors. Both TRP and PIEZO have been studied in autoimmune diseases. Therefore, this study aimed to review the most recent literature on TRP and PIEZO in autoimmune diseases.

TRP and autoimmune diseases

TRP ion channels are a class of channel proteins widely distributed in the peripheral and central nervous systems. To date, more than 30 members of the TRP channel family have been cloned in mammals. TRP channels are six-transmembrane proteins with intracellular N-termini and C-termini. Additionally, the fifth and sixth transmembrane domains together constitute a non-selective cation channel. The TRP channel family includes seven subfamilies, divided into two categories. The first category includes TRPC (typical), TRPV (capsaicin), TRPM (M-type), TRPN (no mechanoreceptor potential) and TRPA (ankyrin). The second category includes TRPP (polycystin) and TRPML (mucolipoprotein). Six TRP channel subfamilies (excluding TRPN) have been confirmed in mammals. These channels can be regulated by many factors, including temperature, osmotic pressure, pH, mechanical force, endogenous and exogenous ligands and intracellular signalling molecules. Currently, the most recognized function is to mediate the transmission of sensory signals, including temperature, pain, pressure, vision and taste, and to regulate cellular calcium balance and affect development. TRP channels are widely expressed in the sensory neurons, skin and brain. Furthermore, studies have shown that TRPV1 and TRPA1 can be endogenously expressed in T cells and participate in T-cell activation and cytokine secretion (Refs Reference Sahoo1, Reference Bertin2). TRP channels in the free nerve terminals of sensory neurons convert painful thermal (TRPV1, TRPA1, TRPM8 and TRPM3), chemical (TRPA1) and mechanical (TRPC1, TRPC3, TRPC6, TRPA1, TRPV2 and TRPV4) stimuli into electrophysiological stimuli. Some TRP channels are not involved in physiological sensation but do play a role in pathological conditions. For example, TRPC5 in mechanical sensation, and TRPV1 and TRPA1 in keratinocytes and satellite glia are involved in the transduction of pain stimuli. In addition, TRP mainly senses temperature changes. Imbalances in temperature perception can shift the comfort temperature from innocuous to noxious stimuli, resulting in pathological pain conditions. Since the gating mechanism of TRP is also regulated by certain inflammatory mediators, these channels are thought to be involved in other pathological pain states, such as inflammatory hyperalgesia and diabetic neuropathy (Ref. Reference Laing and Dhaka3).

Most autoimmune diseases present with symptoms of pain. Opioids are currently used in pain treatment but have addictive side effects and are prone to abuse. In contrast, commonly used painkillers for autoimmune diseases, such as non-steroidal anti-inflammatory drugs, are associated with the risk of cardiovascular diseases and gastrointestinal problems. Therefore, there is an urgent need for safe and effective painkillers. Since TRP is widely expressed in sensory neurons, skin and brain, targeting TRP may represent a novel therapeutic approach for pain treatment. TRP receptors have also been studied in autoimmune diseases. The following is a review of research on TRP in common autoimmune diseases.

Organ-specific autoimmune diseases

Diabetes

Diabetes is a group of metabolic diseases characterized by hyperglycaemia. Hyperglycaemia is caused by defective insulin secretion, impaired biological action or both. Long-term hyperglycaemia leads to chronic damage and dysfunction of various tissues, particularly the eyes, kidneys, heart, blood vessels and nerves. Diabetes can be divided into type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). T1DM develops at a young age (usually <30 years old), with sudden onset and obvious symptoms of polydipsia, polyuria, polyphagia, weight loss and high blood sugar level. Many patients also experience ketoacidosis as the first symptom and demonstrate low serum insulin, C-peptide levels, islet cell antibodies (ICA), insulin autoantibodies (IAA) and glutamate decarboxylase antibodies (GAD-Ab). Oral medication alone is ineffective, and insulin therapy is required. Additionally, T2DM is common in middle-aged and older individuals, and the incidence of obesity is high in these individuals. T2DM is often accompanied by hypertension, dyslipidaemia, arteriosclerosis and other diseases, with an insidious onset, asymptomatic early stages or only mild fatigue, thirst and no obvious increase in blood sugar levels. A glucose tolerance test can be performed for diagnosis. Serum insulin levels are normal or increased in the early stages and decreased in the late stage.

T1DM. Animal models have shown that TRPC1 gene expression is significantly reduced during the late stages of diabetic nephropathy (DN), however clinical studies demonstrated that TRPC1 gene polymorphism may not fundamentally contribute to the development of DN (Refs Reference Zhang4, Reference Niehof and Borlak5).

Additionally, TRPC6 expression was increased in DM rats and mice, and TRPC6 gene knockout was associated with decreased albuminuria in young animals (12-16 weeks of age) in a T1DM mouse model. However, this protective effect was no longer observed once the animals reached 20 weeks of age. A combined knockout of TRPC3, TRPC6, and TRPC7 reduced albuminuria and histological kidney injury. In the rat model, TRPC6 knockout had no significant protective effect against hyperglycaemia, albuminuria, histological kidney injury, blood urea nitrogen, or serum creatinine (Refs Reference Dryer and Kim6, Reference Staruschenko7).

In a spontaneous T1DM model, non-obese diabetic (NOD) mice were subcutaneously injected with high-dose capsaicin, which can inactivate TRPV1 sensory nerve endings (Ref. Reference Jancso and Kiraly8), significantly reduced islet inflammation, delayed the onset of diabetes, and had no effect on the autoimmune invasion of other tissues. In other words, in the absence of TRPV1-positive neurons in the pancreas, protection against diabetes was not due to the elimination or dysregulation of the diabetic T cell population. Capsaicin-treated NOD mice still possess similar numbers of islet-reactive T cells compared with the NOD control mice (Ref. Reference Razavi9). Individuals with T1DM have a lower bone mineral density and a higher risk of fractures. The role of osteoblasts in diabetes-related osteoporosis is well established; however the role of osteoclasts (OCL) is unclear. Furthermore, in vitro studies have shown that diabetes causes local acidosis in the bone marrow, which stimulates OCL formation by activating TRPV1 (Ref. Reference Reni10).

Compared with healthy controls, TRPM7 expression was increased in the hippocampus of T1DM mice. TRPM7 gene silencing can significantly inhibit the decrease of body weight and fasting insulin; inhibit the increase of blood glucose, ICA, IAA, and GAD-Ab levels in streptozotocin-induced; improve spatial cognitive function; and protect hippocampal neurogenesis in T1DM mice (Ref. Reference Zhang, Li and Zhang11).

In conclusion, TRPV1 and TRPM7 activation promotes T1DM. At the same time, TRPV1 can also stimulate OCL formation, leading to osteoporosis secondary to T1DM, and TRPM7 activation is also related to cognitive dysfunction in T1DM (Fig. 1).

Figure 1. Roles of TRP receptor subtypes in T1DM.

Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. DN, diabetic nephropathy; ICA, islet cell antibodies; IAA, insulin autoantibodies; GAD-Ab, glutamate decarboxylase antibodies.

T2DM. The TRPC1 single nucleotide polymorphisms (SNPs) rs7638459 and rs953239 were susceptibility loci associated with T2DM and T2DM without DN, respectively. Among the rs7638459 polymorphisms, the CC genotype significantly increased the risk of T2DM compared with the TT genotype. rs953239 was significantly associated with DN in T2DM, and the CC genotype significantly reduces the risk of DN compared with the AA genotype (Ref. Reference Chen12).

TRPC6 is expressed in podocytes and mediates podocyte injury induced by high glucose levels, a study shows that tacrolimus protects podocytes during the progression of type 2 DN, possibly ameliorating podocyte apoptosis by downregulating the expression of TRPC6 (Ref. Reference Ma13). TRPC6 knockout may reduce the intracellular calcium overload in the brains of T2DM mice, thereby reducing amyloid beta protein deposition and neuroinflammation, and ultimately delaying the development of T2DM-related cognitive dysfunction (Ref. Reference Korayem14).

In rodent models of T2DM, pharmacological blockade of TRPV1 by small-molecule antagonists inhibited calcitonin gene-related peptide (CGRP) secretion, stimulated insulin secretion, reduced insulin resistance and prevented disease progression (Refs Reference Gram, Holst and Szallasi15, Reference Tanaka16, Reference Lee17). However, it was shown that the TRPV1 agonist capsaicin reduces obesity-induced inflammation, insulin resistance and hepatic steatosis in obese mice fed a high-fat diet (HFD), and reduces fasting blood glucose levels (Refs Reference Derbenev and Zsombok18, Reference Panchal, Bliss and Brown19). Simultaneous TRPV1 activation also enhances intestinal glucagon-like peptide-1 (GLP-1) secretion and improves glucose homeostasis (Ref. Reference Wang20).

In rodent models of T2DM, TRPV4 agonists promote vasodilation and improve cardiovascular function, whereas TRPV4 antagonists reduce HFD-induced obesity, insulin resistance, DN, retinopathy and neuropathy (Ref. Reference Hu21).

TRPM2 silencing significantly reduces fibrosis and inflammation in the kidneys of HFD-induced diabetic mice, largely by inhibiting transforming growth factor-β1 (TGF-β1) activation (Ref. Reference Hu22).

TRPM5 is a non-selective monovalent cation channel expressed in islet β cells and activated by an increase in intracellular calcium ions (Ca2+). Islets in TRPM5-knockout mice have shown a significant reduction in glucose-induced calcium activity insulin release, insulin secretion and glucose clearance in both intraperitoneal glucose tolerance tests and oral glucose tolerance tests (Ref. Reference Colsoul23). In addition to glucose-induced insulin secretion, L-arginine-induced insulin secretion (in the presence of low glucose levels) is also impaired in TRPM5-knockout mice, but this effect can be suppressed by the TRPM5 inhibitor triphenylphosphine oxide (TPPO). Furthermore, TPPO blockade of TRPM5 inhibits GLP-1-enhanced glucose-induced insulin release (Refs Reference Krishnan24, Reference Brixel25, Reference Shigeto26). These findings suggest that TRPM5 expression levels inversely correlate with T2DM development (Ref. Reference Vennekens, Mesuere and Philippaert27).

There is no evidence of an association between common TRPM6 and TRPM7 haplotypes and diabetes risk. Compared with non-carriers, TRPM61393Ile-1584Glu haplotype carriers have an increased risk of T2DM only with low magnesium intake (<250 mg/day) (Ref. Reference Song28).

Moreover, allyl isothiocyanate, a potent TRPA1 agonist, may have beneficial effects on glucose uptake and amelioration of impaired insulin signalling through TRPA1 activation. The protective effect against insulin resistance has also been associated with increased mitochondrial activity (Ref. Reference Derbenev and Zsombok18).

In conclusion, TRPC1 SNPs are associated with an increased risk of developing T2DM and DN in T2DM. TRPM5 and TRPA1 activation can improve insulin secretion and negatively correlate with T2DM development, whereas TRPC6, TRPV4 and TRPM2 antagonism can slow down diabetes progression. However, studies on TRPV1 in T2DM have contradictory results; therefore, further research is needed. Studies on the other TRP subfamilies in T2DM are also lacking (Fig. 2).

Figure 2. Roles of TRP receptor subtypes in T2DM.

Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. CGRP, calcitonin gene-related peptide; TGF-β1, transforming growth factor-β1.

Inflammatory bowel disease (IBD)

IBD is a group of idiopathic intestinal inflammatory diseases that involve the ileum, rectum and colon. Clinical manifestations include diarrhoea, abdominal pain and bloody stools. This group includes ulcerative colitis (UC) and Crohn's disease (CD). UC is characterized by continuous inflammation of the colonic mucosa and submucosa, usually first involving the rectum with gradually spreading to the entire colon. CD can involve the entire digestive tract and is a discontinuous full-thickness inflammation. The most involved sites are the terminal ileum, colon and anus.

Intestinal fibrosis is a refractory complication of CD in which TGF-β1 significantly upregulates TRPC6 mRNA and protein expression in patients with CD. Upregulation of TRPC6 expression is critical to the formation of α-smooth muscle actin stress fibres and N-cadherin-mediated adhesion junctions, which enable myofibroblasts to acquire contractile force and strengthen the interconnection between cells simultaneously. Increased Ca2+ influx is involved in the development of intestinal fibrosis by negatively regulating the synthesis of anti-fibrotic factors (Refs Reference Kurahara29, Reference Kurahara30).

Studies have shown that TRPV1 has a pro-inflammatory effect, and inflammatory mediators activate TRPV1 receptors and induce neurogenic inflammatory components through the release of substance P (SP), neurotensin, vasoactive intestinal polypeptide and galanin. In particular, TRPV1 gene-deficient mice exhibit less dextran sodium sulphate (DSS)-induced colitis (Refs Reference Szitter31, Reference Utsumi32). Intraperitoneal and intrathecal administration of TRPV1 and TRPA1 antagonists exerts analgesic effects in a rat colitis model that emphasizes central nervous system mechanisms (Ref. Reference Vermeulen33). However, Massa et al. showed that more severe dinitrobenzenesulfonate (DNBS)-induced colitis was observed in TRPV1-knockout mice, suggesting a protective effect of TRPV1 (Ref. Reference Massa34). In addition, oral curcumin alleviates visceral hyperalgesia by inhibiting TRPV1 phosphorylation in a rat model of UC (Ref. Reference Yang35). In a clinical trial, TRPV1 nerve fibres were increased in quiescent IBD with irritable bowel syndrome-like symptoms and correlated with pain severity. Further studies have shown that TRPV1 expression is increased in inflammatory tissues of patients with active UC compared with non-inflamed tissues and is associated with disease recurrence and persistent activity (Refs Reference Akbar36, Reference Toledo-Maurino37). Another clinical study showed that TRPV1 expression was significantly upregulated in the colonic epithelium of patients with IBD compared with controls, and TRPV1 expression was not significantly different between groups with UC and CD. Although TRPV1 was highly expressed in epithelial cells and infiltrating inflammatory cells in colon biopsies from patients with active IBD, TRPV1 expression was not significantly associated with disease severity (Ref. Reference Luo38). Activation of TRPV1 receptors on sensory nerve terminals mediates neurogenic inflammation through the release of SP and CGRP, resulting in increased vascular permeability, plasma protein extravasation and inflammatory cell activation. While anti-inflammatory sensory neuropeptides, such as somatostatin and opioid peptides, released simultaneously from the same nerve terminal, exert anti-inflammatory and analgesic effects locally and systemically by entering the circulation. Therefore, the role of TRPV1 in IBD depends on a variety of neurotransmitters and cytokine interactions (Ref. Reference Cseko39).

TRPV4 activation triggers pro-inflammatory signals in the gut. After TRPV4 agonists are injected into the colon cavity of mice, the expression of pro-inflammatory chemokines and cytokines upregulates in mouse tissues, myeloperoxidase activity significantly increases and inflammatory cells infiltrate. Inhibition of TRPV4 activation is protective in a colitis model (Refs Reference Vergnolle40, Reference Fichna41, Reference Matsumoto42).

TRPM2 may play a pro-inflammatory role in colitis through its essential roles in macrophages and nuclear factor kappa-B (NF-κB) signalling. Immune cell infiltration and intestinal inflammation severity have been improved in TRPM2-knockout mice with DSS-induced colitis (Ref. Reference Yamamoto43).

In animal experiments, TRPM8 exerted a protective effect in the intestine by inhibiting the release of tumour necrosis factor alpha (TNF-α), interleukin (IL)-1, IL-6, monocyte chemoattractant protein-1 and CGRP. TRPM8 activation also reduces TRPV1-dependent CGRP release in the gut; therefore, TRPM8 is capable of suppressing TRPV1-related inflammatory cascades (Refs Reference Ramachandran44, Reference de Jong45).

In an animal study, mice with experimental colitis exhibited increased TRPA1-mediated release of colonic neuropeptides, whereas symptoms were reduced after TRPA1 inhibition by antagonists or gene deletions (Ref. Reference Engel46). Other studies have shown that TRPA1 plays a protective role in the gastrointestinal tract, and carnabiverine, a potent TRPA1 agonist, improves DNBS- and DSS-induced neutrophil infiltration, intestinal permeability, and cytokine, neuropeptide and chemokine production, and alters the dysbiosis of the gut microbiota (Refs Reference Pagano47, Reference Kun48). Compared with the control group, TRPA1-knockout mice showed more significant intestinal fibrosis, potentially related to the anti-fibrotic effect of TRPA1 in intestinal myofibroblasts (Refs Reference Hiraishi49, Reference Kurahara50). A clinical study showed that carnabiverine reduced intestinal inflammation in children with active UC (Ref. Reference Pagano47). TRPA1 promoter methylation is dysregulated in patients with CD, and abnormal TRPA1 expression may lead to typical symptoms of CD (Ref. Reference Gombert51). Therefore, TRPA1 may play a protective or destructive role in colitis.

In conclusion, TRPC6 promotes the development of intestinal fibrosis. Additionally, TRPV4 and TRPM2 play a pro-inflammatory role in IBD, while TRPM8 inhibits the inflammatory response. The research results of TRPV1 and TRPA1 roles in IBD are, however, inconsistent; they may promote or inhibit disease progression. There are no relevant studies on the roles of the remaining TRP subfamilies in IBD (Fig. 3).

Figure 3. Roles of TRP receptor subtypes in IBD.

Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. DSS, dextran sodium sulfate; DNBS, dinitrobenzenesulfonate; CGRP, calcitonin gene-related peptide; NF-κB, nuclear factor kappa-B; TNF-α, tumour necrosis factor alpha; IL, interleukin; MCP-1, monocyte chemoattractant protein-1.

Multiple sclerosis (MS)

MS is the most common demyelinating disease of the central nervous system. It is characterized by acute active central nerve white matter with multiple inflammatory demyelinating spots and old lesions of calcified spots due to glial fibre hyperplasia. The disease is also marked by multiple lesions, remission and recurrence. It is more likely to occur in the optic nerve, spinal cord and brain stem and is more common in young and middle-aged women than in men.

TRPV1 channels regulate ATP-induced NLR family pyrin domain containing 3 (NLRP3) inflammasome activation by regulating calcium influx and protein phosphatase 2A phosphorylation in microglia. Furthermore, TRPV1 deletion attenuates experimental autoimmune encephalomyelitis (EAE) and neuroinflammation in mice by inhibiting NLRP3 inflammasome activation (Ref. Reference Zhang52). TRPV1 may play a role in pain and weakness associated with influenza-like syndrome in INFβ-induced relapsing-remitting MS (RRMS), especially in GG carriers of the TRPV1 SNP rs222747 (Ref. Reference Buttari53). In vitro, capsaicin stimulation of TRPV1 significantly reduced TNF and IL-6 release from activated microglia and in patients with MS. There was a significant correlation between the TRPV1 SNP rs222747 and cerebrospinal fluid (CSF) TNF levels; in particular, the presence of the G allele known to increase TRPV1 protein expression and function is associated with reduced CSF levels of TNF (Ref. Reference Stampanoni54). The findings suggest that TRPV2 may play a key role in myelination and could be an interesting clinical target for the treatment of demyelinating diseases (Ref. Reference Enrich-Bengoa55).

Genetic deletion of TRPM2 prevents dicyclohexanone oxalyl dihydrazone-induced demyelination, synapse loss, microglial activation, NLRP3 inflammasome activation and pro-inflammatory cytokine production, ultimately leading to improved cognitive decline (Ref. Reference Shao56). In TRPM2-knockout mice, a reduced CXCL2 expression in the CNS inhibits neutrophil infiltration and slows down EAE progression (Ref. Reference Tsutsui57).

TRPM4 is expressed in mouse and human neuronal bodies, as well as in axons from inflammatory CNS injury in mouse EAE and human MS tissue. The pharmacological inhibition of TRPM4 by the antidiabetic drug glyburide resulted in reduced axonal and neuronal degeneration and lower clinical disease scores in EAE, but this did not alter EAE-related immune function (Ref. Reference Schattling58).

TRPM7 overexpression in MS astrocytes inhibits axonal growth by regulating the production of chondroitin sulphate proteoglycans, a key component of gliosis scarring (Ref. Reference Kamermans59). TRPM7 kinase mutation reduces cytokine production, including IL-17 and interferon gamma (IFN-γ), in mouse T cells, which in turn affects MS progression (Ref. Reference Romagnani60).

TRPA1 is expressed in astrocytes in the central nervous system of mice. A previous study demonstrated that TRPA1 deficiency significantly attenuates dicyclohexanone oxalyl dihydrazone-induced demyelination by reducing apoptosis of mature oligodendrocytes (Ref. Reference Saghy61). TRPA1 is involved in the development of neuropathic pain, and pain of central nervous origin is the main symptom of spinal cord injury in RRMS. Another study showed that TRPA1 is associated with the development of mechanical and cold allodynia in a mouse model of relapsing–remitting EAE (RR-EAE)-induced central neuropathic pain (Refs Reference Dalenogare62, Reference Dalenogare63). Headaches are also frequent in patients with progressive MS (PMS). Enhanced TRPA1 endogenous agonists and NADPH oxidase activity have been detected in the trigeminal ganglia of PMS-EAE and RR-EAE mice, activating TRPA1 in trigeminal nociceptors, thereby inducing periorbital mechanical hyperalgesia (Refs Reference Dalenogare64, Reference Dalenogare65). The PMS-EAE model induces depressive and anxiety-like behaviours; however, a selective TRPA1 antagonist (A-967079) reverses these behaviours, suggesting that TRPA1 plays a fundamental role in depression and anxiety-like behaviours (Ref. Reference Peres66). Lysophosphatidylcholine (LPC) is a key inducer of MS caused by neuronal inflammation and demyelinating syndrome. TRPA1 can mediate calcium overload, reactive oxygen species (ROS) generation, mitochondrial membrane depolarization, nitric oxide increase, superoxide production and cytotoxicity in OLN-93 oligodendrocytes induced by LPC, suggesting that TRPA1 plays an important role in LPC-induced oxidative stress and cell damage in OLN-93 oligodendrocytes. TRPA1 inhibition may protect against LPC-induced demyelination (Refs Reference Tian67, Reference Giacco68).

In summary, findings on the role of TRPV1 in MS are contradictory, and further research is needed. TRPV2 may be involved in myelination, TRPM2, TRPM4, TRPM7 and TRPA1 all play a harmful role in MS; there are no related studies on the roles of the other TRP subfamilies in MS (Fig. 4).

Figure 4. Roles of different TRP receptor subtypes in MS.

Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. EAE, experimental autoimmune encephalomyelitis; PMS, progressive multiple sclerosis; RR-EAE, relapsing-remitting experimental autoimmune encephalomyelitis; CSPG, chondroitin sulphate proteoglycans; IL, interleukin; IFN-γ, interferon gamma; PP2A, protein phosphatase 2A.

Anti-glomerular basement membrane autoimmune glomerulonephritis (GN)

In a rat model, the expression of TRPC6 in the glomerulus of rats treated with anti-glomerular basement membrane serum had significantly increased compared to rats treated with control serum. TRPC6 gene knockout could alleviate the pathological changes such as glomerular sclerosis; however, it does not reduce the related indexes such as total albuminuria, blood urea nitrogen and immunoglobulin (Refs Reference Dryer and Kim6, Reference Kim, Shotorbani and Dryer69).

Systemic autoimmune diseases

RA

RA is a chronic multifactorial autoimmune disease mainly characterized by inflammatory synovitis and involves multiple systems. It is also characterized by polyarticular, symmetrical and invasive inflammation of the small joints of the hands and feet, often accompanied by extra-articular organ involvement and positive serum rheumatoid factor, resulting in joint deformity and loss of function.

TRPC5 activation may be associated with the endogenous anti-inflammatory pathways that limit disease progression. In mice with complete Freund's adjuvant (CFA)-induced arthritis, inhibition of TRPC5 through gene deletion or pharmacological antagonism resulted in increased neutrophil infiltration; increased concentrations of cytokines such as IFN-γ, TNF-α and IL-10; and synovial angiogenesis, leading to increased active joint inflammation, hyperalgesia and synovitis. Compared with a non-arthritic control group, TRPC5 expression in patients with RA showed a decreasing trend, but this was not significant. TNF receptor and vascular adhesion molecule-1 expression was significantly increased in arthritic synovium (Ref. Reference Alawi70). Both mRNA and protein expression of TRPC6 were found somewhat higher levels in RA-FLSs than in OA-FLSs. Moreover, inhibiting expression of TRPC6 in vitro reduced proliferation of, as well as inflammatory mediator and protease production by, RA-FLSs, attenuates FLS-mediated synovial inflammation and joint destruction in RA (Ref. Reference Liu71).

In a CFA-induced arthritis model, TRPV1 deletion reduced joint and paw swelling, synovial inflammation, bone erosion and cartilage damage in early stages (≤5 weeks) and suppressed RA-related pain later (>8 weeks) (Refs Reference Barton72, Reference Fernandes73, Reference Szabo74, Reference Hsieh75). TNF-α plays an important role in RA with elevated levels in the synovial fluid of patients with RA (Ref. Reference Miller, Rogers and Muirden76). TNF-α-mediated hyperalgesia is dependent on TRPV1 channels (Ref. Reference Russell77). However, a previous study showed that SA13353 (1-[2-(1-adamantyl)ethyl]-1-pentyl-3-[3-(4-pyridyl)propyl]urea), an active TRPV1 agonist, inhibits TNF-α production by activating TRPV1-mediated capsaicin-sensitive afferent neurons, and reduces hind paw swelling and joint destruction in rats with collagen-induced arthritis (Ref. Reference Murai78). TRPV1 activation in synovial fibroblasts (SF) from patients with RA lead to the production of the inflammatory molecules prostaglandin E2, IL-6 and IL-8, which mediate pain in inflamed joints (Refs Reference Engler79, Reference Terenzi80). Synthetic cannabinoids (WIN) below 1μM play an anti-inflammatory role by reducing the production of cytokines such as IL-6, IL-8 and matrix metalloproteinase (MMP)-3 in RA SF through TRPV1- and TRPA1-dependent pathways (Ref. Reference Lowin, Pongratz and Straub R81). Despite the increased functional expression of the TRPV2 gene in synovial cells from patients with RA, TRPV2 agonists reduced the in vitro invasiveness of these SF (Ref. Reference Galindo, Reyna and Weyer82). In a rat model of RA, the synovial fluid in the joint is abnormal, accompanied by decreased tension, increased acidity and accumulation of various inflammatory mediators, which induce Ca2+ influx by activating the mechanically sensitive TRPV4 channel in SF, accelerating ATP release and ROS production, ultimately enhancing RA synovial cell proliferation (Ref. Reference Hu83). IL-17A increased TRPV4 expression and neuronal hyperexcitability, and TRPV4 may play an important role in hyperalgesia (Ref. Reference Schaible84). However, a study has shown that TRPV4 activation in the presence of IL-1 reduces IL-8 production, implying that IL-8 production is regulated by TRPV4 under inflammatory conditions (Ref. Reference Itoh85).

Compared with wild-type antigen-induced arthritis (AIA) mice, TRPM2-knockout AIA mice had more obvious knee joint swelling, hyperplasia of knee joint synovial cells and significantly increased synovial tissue inflammatory cell infiltration, IL-6, IL-8, and chemokine (C-X-C motif) ligand 6 mRNA expression levels also increased significantly (Ref. 86). The SF of patients with RA express TRPM3 ion channels. Progesterone sulphate, a TRPM3 agonist, stimulates TRPM3 and inhibits the production/secretion of hyaluronic acid (HA) in RA-FLSs (Ref. Reference Ciurtin87). HA is secreted by fibroblast-like synovial cells and is a recognized physiological joint lubricant, but excess HA and HA degradation products contribute to RA progression (Ref. Reference Stuhlmeier88). TRPM7 is highly expressed in chondrocytes and articular cartilage in an adjuvant arthritis (AA) rat model. Its blockade alleviates chondrocyte apoptosis and articular cartilage damage in AA rats by regulating the Indian Hedgehog signalling pathway (Refs Reference Ma89, Reference Zhou90). TRPM7 is involved in CD147 (extracellular MMP inducer)-induced chemotaxis, adhesion and invasiveness of neutrophils in patients with RA, leading to progressive cartilage destruction (Ref. Reference Wang91). In RA synovium, SF are the main cell population of the invasive synovium. TRPM7 expression is significantly enhanced in SF, and TRPM7 channel inhibition can induce SF apoptosis through endoplasmic reticulum stress and exert anti-inflammatory effects (Ref. Reference Li92). Menthol-induced SF apoptosis results from TRPM8-mediated extracellular calcium entry, intracellular ROS generation and mitochondrial membrane depolarization (Ref. Reference Zhu93).

TRPA1 activation in sensory nerve fibres causes pain. In a CFA-induced mouse model of chronic arthritis, TRPA1 contributes to joint pain and inflammation (Refs Reference Fernandes73, Reference Horvath94, Reference Pereira95). TRPA1 levels are elevated in leukocytes of patients with RA, TNF also upregulates TRPA1 under inflammatory conditions in RA SF, and its activation is accompanied by enhanced calcium influx, decreased proliferation and increased necrosis (Ref. Reference Lowin96).

In conclusion, TRPC5, TRPV2, TRPM2 and TRPM8 activation can delay the progression of RA by reducing inflammation and SF invasiveness. However, TRPC6, TRPM7 and TRPA1 promoted RA progression. TRPV1, TRPV4 and TRPM3 have positive and negative effects on RA, which need to be further studied. Other TRP family subtypes have not been identified in patients with RA (Fig. 5).

Figure 5. Roles of TRP receptor subtypes in RA.

Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. TNF-α, tumour necrosis factor alpha; IL, interleukin; PGE2, prostaglandin E2; ROS, reactive oxygen species; NE, neutrophil; Mø, macrophage.

OA

OA is a degenerative disease caused by degeneration of the articular cartilage and reactive hyperplasia of the joint edge and subchondral bone. It often occurs due to ageing, obesity, strain, trauma, joint congenital abnormality, joint deformity and among various other factors. The disease is more common in middle-aged and older adults, as well as in weight-bearing joints and joints with more activities (such as the cervical spine, lumbar spine, knee joint and hip joint). Excessive weight bearing or the use of these joints can promote degenerative changes. Clinical manifestations include a slow developing joint pain, tenderness, stiffness, joint swelling, limited motion and joint deformity. A previous study demonstrated that a variety of immune cells such as macrophages, neutrophils and T cells are involved in OA pathogenesis (Ref. Reference Nedunchezhiyan97).

TRPC5 expression is reduced in the synovial tissue of patients with OA. TRPC5 deletion can promote the release of MMP, a key enzyme for chondrocyte degradation and coordinate immune cell migration and infiltration (Ref. Reference de Sousa98).

TRPV1 is expressed in SF from patients with OA. TRPV1 activation can induce the production of inflammatory factors, such as IL-6, thereby helping to regulate the pain sensation in inflamed joints (Ref. Reference Engler79). TRPV1 expression and M1 macrophage infiltration were simultaneously increased in both human and rat OA synovium. More than 90% of the infiltrated M1 macrophages expressed TRPV1. In the rat OA model, intra-articular injection of capsaicin, a specific TRPV1 agonist, significantly attenuated OA phenotypes, including joint swelling, synovitis, cartilage damage and osteophyte formation. Capsaicin treatment markedly reduced M1 macrophage infiltration in the synovium (Ref. Reference Lv99). Studies have shown that the infiltration of M1 synovial macrophages and the expression of TRPV4 were increased significantly in OA synovium, inhibition of TRPV4 delays OA progression by inhibiting M1 synovial macrophage polarization through the ROS/NLRP3 pathway (Ref. Reference Sun100). Meanwhile specific TRPV4 knockout in chondrocytes can reduce OA severity caused by ageing in adult mice and intra-articular administration of a TRPV4 antagonist suppresses pain-related behaviours in a monoiodoacetic acid (MIA)-induced OA pain model (MIA rats), not due to inhibition of knee joint injury or inflammation caused by OA in MIA rats (Refs 101, Reference O'Conor102, Reference Hinata103, Reference Soga104). However, others have shown that TRPV4 agonists can stimulate chondrocytes to produce extracellular matrix, thereby alleviating articular cartilage damage and having a cartilage-protective effect (Refs Reference Atobe105, Reference Clark106). TRPV5 expression is upregulated in MIA-induced articular cartilage OA. TRPV5 can inhibit cell autophagy by mediating the influx of extracellular Ca2+ and increase the production of calmodulin and phosphorylation of calmodulin-dependent protein kinase II (p-CAMK II). Activated p-CAMK II promotes chondrocyte apoptosis through mitogen-activated protein kinases and Akt/mTOR pathways. Previous studies also showed that TRPV5 inhibitors slow down the progression of joint destruction, further demonstrating the role of TRPV5 in OA (Refs Reference Wei107, Reference Wei108, Reference Wei109). In an OA rat model, TRPV6 expression significantly downregulated, resulting in a decreased secretion of extracellular matrix by chondrocytes and a significantly increased expression of MMP-1 and MMP-13, which inhibits chondrocyte proliferation and promotes chondrocyte apoptosis, thereby participating in OA pathogenesis (Ref. Reference Song110).

TRPM8 is expressed in human cartilage tissue and on the chondrocyte membrane. Further research has demonstrated that TRPM8 expression in patients with OA was significantly higher than that in healthy people, suggesting that TRPM8 may be related to OA. Cold activation of TRPM8 causes Ca2+ overload, increased intracellular ROS production and depolarization of the mitochondrial membrane potential, leading to increased cell necrosis and apoptosis, a potential mechanism (Ref. 111).

TRPA1 mediates sensitization in an OA rodent model (Ref. Reference Mcgaraughty112) and induces IL-6 expression in chondrocytes, which in turn upregulates MMP-1 and MMP-13 to promote OA progression (Ref. Reference Nummenmaa113).

In summary, the downregulated expression of TRPC5 and TRPV6 in OA may participate in the pathogenesis of OA by promoting MMP release, inhibiting chondrocyte proliferation, and promoting chondrocyte apoptosis. TRPV5, TRPM8 and TRPA1 expression in OA can also contribute to OA pathogenesis by promoting the production of pro-inflammatory factors, and the release of MMP, inhibiting chondrocyte proliferation, and promoting chondrocyte apoptosis. The role of TRPV1and TRPV4 in OA is either positive or negative and requires further study. There are no relevant studies on the roles of other TRP subtypes in OA (Fig. 6).

Figure 6. Roles of TRP receptor subtypes in OA.

Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. MMP, matrix metalloproteinase; IL, interleukin; ROS, reactive oxygen species.

SLE

SLE is an autoimmune inflammatory connective tissue disease that affects multiple organs and occurs mostly in young women. It has a slow onset, insidious occurrence and various clinical manifestations. SLE can involve the skin, serosa, joints, kidneys and central nervous system.

Lupus kidney is the most common visceral lesion in SLE, and the severity of renal lesions directly affects the prognosis. Several studies have shown that TRPC6 gene inhibition can reduce pathological changes such as renal fibrosis (Refs Reference Dryer and Kim6, Reference Kim, Shotorbani and Dryer69, Reference Zheng114, Reference Wu115) and may delay the progression of lupus kidney. Furthermore, neuropsychiatric lupus erythematosus (NPSLE) is a group of serious complications related to a poor prognosis and high mortality caused by SLE lesions with nervous system involvement, resulting in neurological and/or psychiatric symptoms. Studies have shown that the TRPC6 genotype is associated with NPSLE incidence. Patients with SLE and the TT genotype of the rs7925662 SNP in the TRPC6 gene have an increased risk of developing NPSLE during follow-up, whereas patients with the C allele have a lower NPSLE incidence (Refs Reference Ramirez116, Reference Ramirez117).

SS

SS is a chronic inflammatory autoimmune disease mainly involving the exocrine glands. In addition to impaired function of the salivary and lacrimal glands, resulting in dry mouth and eyes, other exocrine glands and organs outside of the gland are involved, resulting in symptoms of multi-system damage.

In the salivary gland (SG) epithelial cells, IL-17 downregulates TRPC1 expression by inhibiting acetylcholine-induced calcium motility. TRPC1 deletion results in marked attenuation of agonist-induced calcium motility in mice and a 70% loss of salivary secretion (Refs Reference Xiao118, Reference Liu119). Knockdown of endogenous TRPC1 also significantly reduces store-operated Ca2+ entry (SOCE) in the human SG cell lines, mouse pancreatic and submandibular cells. TRPC3 is also involved in SOCE. Mice lacking TRPC1 or TRPC3 show reduced SOCE and glandular secretion, and TRPC3 exerts its function in a TRPC1-dependent manner. TRPC1-knockout mice do not show TRPC3-dependent SOCE (Ref. Reference Lee120).

TRPV4 activation leads to increased fluid secretion from SG acinar cells by increasing intracellular Ca2+. Muscarinic stimulation of salivary and tear secretion was downregulated in TRPV4-deficient mice and acinar cells treated with a TRPV4-specific antagonist (HC-067047). Infusion of the entire submandibular gland with the TRPV1 agonist capsaicin (1 μM) via the submandibular artery significantly increased carbacol-induced salivation, whereas infusion of TRPM8 and TRPA1 agonists decreased it. Moreover, radiation-induced loss of SG fluid secretion is mediated through a TRPM2-dependent pathway that affects mitochondrial function and leads to an irreversible loss of SOCE (Ref. Reference Liu, Ong and Ambudkar121).

In conclusion, TRPC1, TRPC3, TRPV1 and TRPV4 all contribute to SG secretion, whereas TRPM2, TRPM8 and TRPA1 inhibit SG secretion. The remaining TRP subfamilies were not related to salivary SG (Fig. 7).

Figure 7. Roles of TRP receptor subtypes in SS.

Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. SOCE, store-operated Ca2 + entry.

Gout

Gout is a common and complex type of arthritis that affects all age groups, with a higher incidence in men than in women. Patients with gout often experience sudden nocturnal joint pain. The onset is acute, and patients experience joint pain, oedema, redness and inflammation. The pain is gradually relieved until it disappears and lasts several days or weeks. Gout is related to an increased concentration of uric acid in the body, and it forms urate deposits in the joint cavity and other locations, thereby causing acute joint pain. Studies have shown that urate deposition in gout can mediate NLRP3 inflammasome activation, leading to the dysfunction of T-cell subsets and the initiation and progression of an autoimmune attack (Ref. Reference Wang122).

In rats with gouty arthritis, noxious stimulus inducement increases in cerebral blood volume in the primary somatosensory cortex and thalamus. This increase correlates with the upregulation of TRPV1 protein expression and pain behaviour. Selective blockade of central TRPV1 channel activity by intrathecal administration reverses the induced pain and abrogates the cerebral blood volume increase in the thalamocortical region (Refs Reference Chen123, Reference Xu124).

Studies have shown that inhibition of TRPM2 channels significantly attenuates monosodium urate (MSU)-induced activation of the NLRP3 inflammasome and macrophages secrete the bioactive substance IL-1β. In an animal model of gout, TRPM2 depletion significantly attenuated MSU-induced inflammation dominated by neutrophil infiltration (Ref. Reference Zhong125).

Furthermore, in a mouse model, IL-33 promotes neutrophil migration and triggers neutrophil-dependent ROS production, which in turn activates TRPA1 channels in the dorsal root ganglion neurons and produces pain sensation (Ref. Reference Yin126). In an additional animal model of MSU-induced gout, MSU injection increased tissue hydrogen peroxide levels, which stimulated TRPA1 and TRPV1 expression on sensory nerve endings, enhanced cellular infiltration and IL-1β levels, produced pain sensation and resulted in joint swelling. Both pharmacological inhibition and gene knockout of TRPA1 channels abrogated pain and oedema induced by MSU in animal models (Refs Reference Trevisan127, Reference Moilanen128, Reference Trevisan129).

In conclusion, TRPV1, TRPM2 and TRPA1 gene knockout or pharmacological inhibition can reduce joint pain and oedema in mice with gouty arthritis. There is no relevant research on the role of the other TRP family subtypes in gout (Fig. 8).

Figure 8. Roles of TRP receptor subtypes in gout.

Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. MSU, monosodium urate; IL, interleukin.

PIEZO and autoimmune diseases

PIEZO refers to pressure. The PIEZO family has two members, PIEZO1 and PIEZO2. PIEZO1 exists in many cells and tissues of the body, whereas PIEZO2 is mainly related to touch, proprioception, visceral sense and mechanical force perception.

PIEZO1 is widely expressed in the cardiovascular system. It is expressed on vascular endothelial cells, red blood cells, epithelial cells and cardiomyocytes. It also plays an important role in sensing drastic mechanical changes produced by cardiac systolic and diastolic relaxation, thereby regulating cardiac stability. PIEZO1 expression is upregulated in patients with heart disease (Ref. Reference Jiang130). Loss of human PIEZO1 gene function can lead to congenital lymphatic dysplasia, whereas gain-of-function mutations can lead to haemolytic anaemia and hereditary stem cell hyperplasia (Ref. Reference Alper131). Moreover, PIEZO1 is important for bone formation and remodelling. Long-term bed rest or weightlessness in space can lead to severe bone loss. The reason bones can feel gravity is due to PIEZO1 expressed in osteoblasts. Therefore, PIEZO1 deficiency causes loss of bone mass and spontaneous fractures along with increased bone resorption. Furthermore, PIEZO1-deficient mice have also been shown to develop severe bone loss (Ref. Reference Wang132).

In contrast, PIEZO2 mediates touch perception and plays an important role in human physiological processes, such as mechanical pain, urination, respiration, blood pressure and bone remodelling. Touching, hugging and the use of various tools, such as the screen of a mobile phone, all require the participation of PIEZO2. There is also ‘proprioception’, our own muscle state, such as maintaining balance when walking, as PIEZO2 in the nervous system can monitor muscle tension (Refs Reference Chesler133, Reference Woo134). Additionally, mechanical changes in internal organs, such as blood pressure, respiration and bladder filling, are also monitored by PIEZO2 (Refs Reference Marshall135, Reference Nonomura136, Reference Ghitani and Chesler137). Loss-of-function mutations in the human PIEZO2 gene cause autosomal recessive muscular dystrophy syndrome with perinatal respiratory distress, joint curvature, scoliosis and possibly urinary abnormalities due to the inability to sense bladder fullness. Gain-of-function mutations, however, cause an autosomal dominant distal joint curvature (Ref. Reference Alper131).

Organ-specific autoimmune diseases

Animal studies have shown that the exercise pressor reflex is exaggerated in early T1DM (Refs Reference Grotle138, Reference Matteucci139), and PIEZO channels may play an important role in this phenomenon. GsMTx4, a biologically derived peptide that specifically inhibits mechanically activated cation channels, can also inhibit heterologous co-expression of PIEZO1/2, attenuate the exaggerated exercise pressor reflex in T1DM rats and effectively reduce the greater cardiovascular strain caused by intermittent muscle contraction (Ref. Reference Grotle140).

In MS, PIEZO1 channel activation in axons negatively regulates central nervous system myelination (Ref. Reference Velasco-Estevez141). PIEZO1 depletion in T cells can selectively promote regulatory T-cell proliferation, thereby reducing EAE severity. However, it does not affect thymus development, lymph node homing, T-cell receptor priming, T-cell proliferation and differentiation (Refs Reference Jairaman142, Reference Yang143).

Systemic autoimmune diseases

A previous study demonstrated that in OA, inflammation enhances mechanical stimulation-induced PIEZO channel currents. However, knee OA does not affect the expression levels of PIEZO1 or PIEZO2 mRNA, suggesting that PIEZO channel function may be enhanced in knee OA without altering the mRNA expression levels (Ref. Reference Ikeda, Arimura and Saito144). Though, in OA, IL-1α is increased and IL-1α inflammatory signalling in articular chondrocytes increases PIEZO1 expression and function in chondrocytes, resulting in increased mechanically induced Ca2+ influx, leading to increased sensitivity to mechanical stimuli (Ref. Reference Lee145). The PIEZO channel mediates the signal transduction of damaging mechanical stimuli. PIEZO1 and PIEZO2 co-expression has a synergistic effect, resulting in increased sensitivity to mechanical stimuli, which in turn promotes chondrocyte injury. However, GsMTx4 attenuates the response of chondrocytes to injury-inducing mechanical stimulation (Refs Reference Lee146, Reference Lee, Guilak and Liedtke147).

Summary and outlook

The announcement of the 2021 Nobel Prize in Physiology or Medicine brought TRP and PIEZO receptors to our attention. This article summarizes the roles of TRP and PIEZO receptors in various autoimmune diseases. Different subtypes have different roles in autoimmune diseases, some have specific protective effects, and some have harmful effects (Tables 1 and 2). However, most current studies on TRP and PIEZO receptors in autoimmune diseases are animal studies, and there are currently few clinical studies. Therefore, understanding the roles of different receptor subtypes in autoimmune diseases provides a new method for the treatment of autoimmune diseases.

Table 1. Role of TRP receptors in organ-specific autoimmune diseases

Table 2. Role of TRP receptors in systemic autoimmune disease

Acknowledgements

Not applicable.

Author contributions

M. D. designed and supervised the study, and Y. B. reviewed the literature, wrote the original manuscript and revised the manuscript. Z. X., W. Z., A. Q., Z. J. and G. X. helped with the literature search. M. D. and Z. L. modified the article. All authors contributed to the article and approved the submitted version.

Funding statement

This work was supported by the basic research project of Shanxi Science and Technology Department[grant number 202103021224342]; and Key Research and Development (R&D) Projects of Shanxi Province (2021XM01).

Competing interests

No conflict of interest among all authors.

Ethical standards

Not applicable.

Footnotes

*

Note: Yang Baoqi and Ma Dan have the same contribution.

References

Sahoo, SS et al. (2019) Transient receptor potential ankyrin1 channel is endogenously expressed in T cells and is involved in immune functions[J]. Bioscience Reports. 39, BSR20191437.CrossRefGoogle Scholar
Bertin, S et al. (2014) The ion channel TRPV1 regulates the activation and proinflammatory properties of CD4(+) T cells[J]. Nature Immunology 15, 10551063.CrossRefGoogle ScholarPubMed
Laing, RJ and Dhaka, A (2016) ThermoTRPs and pain[J]. Neuroscientist 22, 171187.CrossRefGoogle ScholarPubMed
Zhang, D et al. (2009) Evaluation of genetic association and expression reduction of TRPC1 in the development of diabetic nephropathy[J]. American Journal of Nephrology 29, 244251.CrossRefGoogle ScholarPubMed
Niehof, M and Borlak, J (2008) HNF4 alpha and the Ca-channel TRPC1 are novel disease candidate genes in diabetic nephropathy[J]. Diabetes 57, 10691077.CrossRefGoogle ScholarPubMed
Dryer, SE and Kim, EY (2022) The effects of TRPC6 knockout in animal models of kidney disease[J]. Biomolecules 12, 1710.CrossRefGoogle ScholarPubMed
Staruschenko, A et al. (2023) Ion channels and channelopathies in glomeruli[J]. Physiological Reviews 103, 787854.CrossRefGoogle ScholarPubMed
Jancso, G and Kiraly, E (1981) Sensory neurotoxins: chemically induced selective destruction of primary sensory neurons[J]. Brain Research 210, 8389.CrossRefGoogle ScholarPubMed
Razavi, R et al. (2006) TRPV1+ sensory neurons control beta cell stress and islet inflammation in autoimmune diabetes[J]. Cell 127, 11231135.CrossRefGoogle ScholarPubMed
Reni, C et al. (2016) Diabetes stimulates osteoclastogenesis by acidosis-induced activation of transient receptor potential cation channels[J]. Scientific Reports 6, 30639.CrossRefGoogle ScholarPubMed
Zhang, QJ, Li, J and Zhang, SY (2018) Effects of TRPM7/miR-34a gene silencing on spatial cognitive function and hippocampal neurogenesis in mice with type 1 diabetes mellitus[J]. Molecular Neurobiology 55, 15681579.CrossRefGoogle ScholarPubMed
Chen, K et al. (2013) Association of TRPC1 gene polymorphisms with type 2 diabetes and diabetic nephropathy in Han Chinese population[J]. Endocrine Research 38, 5968.CrossRefGoogle ScholarPubMed
Ma, R et al. (2021) Tacrolimus protects podocytes from apoptosis via downregulation of TRPC6 in diabetic nephropathy[J]. Journal of Diabetes Research 2021, 8832114.CrossRefGoogle ScholarPubMed
Korayem, GB et al. (2022) The prescribing pattern of sodium-glucose cotransporter-2 inhibitors and glucagon-like peptide-1 receptor agonists in patient with type two diabetes mellitus: a two-center retrospective cross-sectional study[J]. Frontiers in Public Health 10, 1031306.CrossRefGoogle ScholarPubMed
Gram, DX, Holst, JJ and Szallasi, A (2017) TRPV1: a potential therapeutic target in type 2 diabetes and comorbidities?[J]. Trends in Molecular Medicine 23, 10021013.CrossRefGoogle ScholarPubMed
Tanaka, H et al. (2011) Enhanced insulin secretion and sensitization in diabetic mice on chronic treatment with a transient receptor potential vanilloid 1 antagonist[J]. Life Sciences 88, 559563.CrossRefGoogle ScholarPubMed
Lee, E et al. (2015) Transient receptor potential vanilloid type-1 channel regulates diet-induced obesity, insulin resistance, and leptin resistance[J]. FASEB Journal 29, 31823192.CrossRefGoogle ScholarPubMed
Derbenev, AV and Zsombok, A (2016) Potential therapeutic value of TRPV1 and TRPA1 in diabetes mellitus and obesity[J]. Seminars in Immunopathology 38, 397406.CrossRefGoogle ScholarPubMed
Panchal, SK, Bliss, E and Brown, L (2018) Capsaicin in metabolic syndrome[J]. Nutrients 10, 630.CrossRefGoogle ScholarPubMed
Wang, P et al. (2012) Transient receptor potential vanilloid 1 activation enhances gut glucagon-like peptide-1 secretion and improves glucose homeostasis[J]. Diabetes 61, 21552165.CrossRefGoogle ScholarPubMed
Hu, W et al. (2020) Transient receptor potential vanilloid 4 channels as therapeutic targets in diabetes and diabetes-related complications[J]. Journal of Diabetes Investigation 11, 757769.CrossRefGoogle ScholarPubMed
Hu, F et al. (2021) Knockdown of transient receptor potential melastatin 2 reduces renal fibrosis and inflammation by blocking transforming growth factor-beta1-activated JNK1 activation in diabetic mice[J]. Aging 13, 2460524620.CrossRefGoogle ScholarPubMed
Colsoul, B et al. (2010) Loss of high-frequency glucose-induced Ca2+ oscillations in pancreatic islets correlates with impaired glucose tolerance in Trpm5-/-mice[J]. Proceedings of the National Academy of Sciences of the USA 107, 52085213.CrossRefGoogle ScholarPubMed
Krishnan, K et al. (2014) Role of transient receptor potential melastatin-like subtype 5 channel in insulin secretion from rat beta-cells[J]. Pancreas 43, 597604.CrossRefGoogle ScholarPubMed
Brixel, LR et al. (2010) TRPM5 regulates glucose-stimulated insulin secretion[J]. Pflugers Archiv 460, 6976.CrossRefGoogle ScholarPubMed
Shigeto, M et al. (2015) GLP-1 stimulates insulin secretion by PKC-dependent TRPM4 and TRPM5 activation[J]. Journal of Clinical Investigation 125, 47144728.CrossRefGoogle ScholarPubMed
Vennekens, R, Mesuere, M and Philippaert, K (2018) TRPM5 in the battle against diabetes and obesity[J]. Acta Physiologica 222.CrossRefGoogle ScholarPubMed
Song, Y et al. (2009) Common genetic variants of the ion channel transient receptor potential membrane melastatin 6 and 7 (TRPM6 and TRPM7), magnesium intake, and risk of type 2 diabetes in women[J]. BMC Medical Genomics 10, 4.CrossRefGoogle ScholarPubMed
Kurahara, LH et al. (2016) Significant contribution of TRPC6 channel-mediated Ca(2+) influx to the pathogenesis of Crohn's disease fibrotic stenosis[J]. Journal of Smooth Muscle Research 52, 7892.CrossRefGoogle Scholar
Kurahara, LH et al. (2015) Intestinal myofibroblast TRPC6 channel may contribute to stenotic fibrosis in Crohn's disease[J]. Inflammatory Bowel Diseases 21, 496506.CrossRefGoogle ScholarPubMed
Szitter, I et al. (2014) Role of neurokinin 1 receptors in dextran sulfate-induced colitis: studies with gene-deleted mice and the selective receptor antagonist netupitant[J]. Inflammation Research 63, 399409.CrossRefGoogle ScholarPubMed
Utsumi, D et al. (2018) Transient receptor potential vanilloid 1 and transient receptor potential ankyrin 1 contribute to the progression of colonic inflammation in dextran sulfate sodium-induced colitis in mice: links to calcitonin gene-related peptide and substance P[J]. Journal of Pharmacological Sciences 136, 121132.CrossRefGoogle Scholar
Vermeulen, W et al. (2013) Role of TRPV1 and TRPA1 in visceral hypersensitivity to colorectal distension during experimental colitis in rats[J]. European Journal of Pharmacology 698, 404412.CrossRefGoogle ScholarPubMed
Massa, F et al. (2006) Vanilloid receptor (TRPV1)-deficient mice show increased susceptibility to dinitrobenzene sulfonic acid induced colitis[J]. Journal of Molecular Medicine 84, 142146.CrossRefGoogle ScholarPubMed
Yang, M et al. (2017) Oral administration of curcumin attenuates visceral hyperalgesia through inhibiting phosphorylation of TRPV1 in rat model of ulcerative colitis[J]. Molecular Pain 13, 1940339937.CrossRefGoogle ScholarPubMed
Akbar, A et al. (2010) Expression of the TRPV1 receptor differs in quiescent inflammatory bowel disease with or without abdominal pain[J]. Gut 59, 767774.CrossRefGoogle ScholarPubMed
Toledo-Maurino, JJ et al. (2018) The transient receptor potential vanilloid 1 is associated with active inflammation in ulcerative colitis[J]. Mediators of Inflammation 2018, 6570371.CrossRefGoogle ScholarPubMed
Luo, C et al. (2017) Upregulation of the transient receptor potential vanilloid 1 in colonic epithelium of patients with active inflammatory bowel disease[J]. International Journal of Clinical and Experimental Pathology 10, 1133511344.Google ScholarPubMed
Cseko, K et al. (2019) Role of TRPV1 and TRPA1 ion channels in inflammatory bowel diseases: potential therapeutic targets?[J]. Pharmaceuticals 12, 48.CrossRefGoogle ScholarPubMed
Vergnolle, N (2014) TRPV4: new therapeutic target for inflammatory bowel diseases[J]. Biochemical Pharmacology 89, 157161.CrossRefGoogle ScholarPubMed
Fichna, J et al. (2012) Transient receptor potential vanilloid 4 blockade protects against experimental colitis in mice: a new strategy for inflammatory bowel diseases treatment?[J]. Neurogastroenterology & Motility 24, e557e560.CrossRefGoogle ScholarPubMed
Matsumoto, K et al. (2018) Transient receptor potential vanilloid 4 channel regulates vascular endothelial permeability during colonic inflammation in dextran sulphate sodium-induced murine colitis[J]. British Journal of Pharmacology 175, 8499.CrossRefGoogle ScholarPubMed
Yamamoto, S et al. (2008) TRPM2-mediated Ca2+ influx induces chemokine production in monocytes that aggravates inflammatory neutrophil infiltration[J]. Nature Medicine 14, 738747.CrossRefGoogle ScholarPubMed
Ramachandran, R et al. (2013) TRPM8 activation attenuates inflammatory responses in mouse models of colitis[J]. Proceedings of the National Academy of Sciences of the USA 110, 74767481.CrossRefGoogle ScholarPubMed
de Jong, PR et al. (2015) TRPM8 on mucosal sensory nerves regulates colitogenic responses by innate immune cells via CGRP[J]. Mucosal Immunology 8, 491504.CrossRefGoogle ScholarPubMed
Engel, MA et al. (2011) TRPA1 and substance P mediate colitis in mice[J]. Gastroenterology 141, 13461358.CrossRefGoogle ScholarPubMed
Pagano, E et al. (2019) The non-euphoric phytocannabinoid cannabidivarin counteracts intestinal inflammation in mice and cytokine expression in biopsies from UC pediatric patients[J]. Pharmacological Research 149, 104464.CrossRefGoogle ScholarPubMed
Kun, J et al. (2014) Upregulation of the transient receptor potential ankyrin 1 ion channel in the inflamed human and mouse colon and its protective roles[J]. PLoS ONE 9, e108164.CrossRefGoogle ScholarPubMed
Hiraishi, K et al. (2018) Daikenchuto (Da-Jian-Zhong-Tang) ameliorates intestinal fibrosis by activating myofibroblast transient receptor potential ankyrin 1 channel[J]. World Journal of Gastroenterology 24, 40364053.CrossRefGoogle ScholarPubMed
Kurahara, LH et al. (2018) Activation of myofibroblast TRPA1 by steroids and pirfenidone ameliorates fibrosis in experimental Crohn's disease[J]. Cellular and Molecular Gastroenterology and Hepatology 5, 299318.CrossRefGoogle ScholarPubMed
Gombert, S et al. (2020) Transient receptor potential ankyrin 1 promoter methylation and peripheral pain sensitivity in Crohn's disease[J]. Clinical Epigenetics 12, 1.CrossRefGoogle Scholar
Zhang, Y et al. (2021) TRPV1 channel mediates NLRP3 inflammasome-dependent neuroinflammation in microglia[J]. Cell Death & Disease 12, 1159.CrossRefGoogle ScholarPubMed
Buttari, F et al. (2017) TRPV1 polymorphisms and risk of interferon beta-induced flu-like syndrome in patients with relapsing-remitting multiple sclerosis[J]. Journal of Neuroimmunology 305, 172174.CrossRefGoogle ScholarPubMed
Stampanoni, BM et al. (2019) Transient receptor potential vanilloid 1 modulates central inflammation in multiple sclerosis[J]. Frontiers in Neurology 10, 30.CrossRefGoogle Scholar
Enrich-Bengoa, J et al. (2022) TRPV2: a Key player in myelination disorders of the central nervous system[J]. International Journal of Molecular Sciences 23, 3617.CrossRefGoogle ScholarPubMed
Shao, Y et al. (2021) TRPM2 contributes to neuroinflammation and cognitive deficits in a cuprizone-induced multiple sclerosis model via NLRP3 inflammasome[J]. Neurobiology of Disease 160, 105534.CrossRefGoogle Scholar
Tsutsui, M et al. (2018) TRPM2 exacerbates central nervous system inflammation in experimental autoimmune encephalomyelitis by increasing production of CXCL2 chemokines[J]. Journal of Neuroscience 38, 84848495.CrossRefGoogle ScholarPubMed
Schattling, B et al. (2012) TRPM4 cation channel mediates axonal and neuronal degeneration in experimental autoimmune encephalomyelitis and multiple sclerosis[J]. Nature Medicine 18, 18051811.CrossRefGoogle ScholarPubMed
Kamermans, A et al. (2019) Reactive astrocytes in multiple sclerosis impair neuronal outgrowth through TRPM7-mediated chondroitin sulfate proteoglycan production[J]. Glia 67, 6877.CrossRefGoogle ScholarPubMed
Romagnani, A et al. (2017) TRPM7 kinase activity is essential for T cell colonization and alloreactivity in the gut[J]. Nature Communications 8, 1917.CrossRefGoogle Scholar
Saghy, E et al. (2016) TRPA1 deficiency is protective in cuprizone-induced demyelination – a new target against oligodendrocyte apoptosis[J]. Glia 64, 21662180.CrossRefGoogle ScholarPubMed
Dalenogare, DP et al. (2020) TRPA1 activation mediates nociception behaviors in a mouse model of relapsing-remitting experimental autoimmune encephalomyelitis[J]. Experimental Neurology 328, 113241.CrossRefGoogle Scholar
Dalenogare, DP et al. (2023) Neuropathic-like nociception and spinal cord neuroinflammation are dependent on the TRPA1 channel in multiple sclerosis models in mice[J]. Cells. 12, 1511.CrossRefGoogle ScholarPubMed
Dalenogare, DP et al. (2021) Periorbital nociception in a progressive multiple sclerosis mouse model is dependent on TRPA1 channel activation[J]. Pharmaceuticals. 14, 831.CrossRefGoogle Scholar
Dalenogare, DP et al. (2022) Transient receptor potential ankyrin 1 mediates headache-related cephalic allodynia in a mouse model of relapsing-remitting multiple sclerosis[J]. Pain 163, 13461355.CrossRefGoogle Scholar
Peres, DS et al. (2021) TRPA1 involvement in depression- and anxiety-like behaviors in a progressive multiple sclerosis model in mice[J]. Brain Research Bulletin 175, 115.CrossRefGoogle Scholar
Tian, C et al. (2020) Transient receptor potential ankyrin 1 contributes to the lysophosphatidylcholine-induced oxidative stress and cytotoxicity in OLN-93 oligodendrocyte[J]. Cell Stress & Chaperones 25, 955968.CrossRefGoogle Scholar
Giacco, V et al. (2023) Transient receptor potential ankyrin-1 (TRPA1) agonists suppress myelination and induce demyelination in organotypic cortical slices[J]. Glia 71, 14021413.CrossRefGoogle ScholarPubMed
Kim, EY, Shotorbani, PY and Dryer, SE (2019) TRPC6 inactivation does not affect loss of renal function in nephrotoxic serum glomerulonephritis in rats, but reduces severity of glomerular lesions[J]. Biochemistry and Biophysics Reports 17, 139150.CrossRefGoogle Scholar
Alawi, KM et al. (2017) Transient receptor potential canonical 5 (TRPC5) protects against pain and vascular inflammation in arthritis and joint inflammation[J]. Annals of the Rheumatic Diseases 76, 252260.CrossRefGoogle ScholarPubMed
Liu, G et al. (2021) Inhibition of transient receptor potential canonical 6 attenuates fibroblast-like synoviocytes mediated synovial inflammation and joint destruction in rheumatoid arthritis[J]. Clinical and Experimental Rheumatology 39, 115124.CrossRefGoogle ScholarPubMed
Barton, NJ et al. (2006) Attenuation of experimental arthritis in TRPV1R knockout mice[J]. Experimental and Molecular Pathology 81, 166170.CrossRefGoogle ScholarPubMed
Fernandes, ES et al. (2011) A distinct role for transient receptor potential ankyrin 1, in addition to transient receptor potential vanilloid 1, in tumor necrosis factor alpha-induced inflammatory hyperalgesia and Freund's complete adjuvant-induced monarthritis[J]. Arthritis and Rheumatism 63, 819829.CrossRefGoogle ScholarPubMed
Szabo, A et al. (2005) Role of transient receptor potential vanilloid 1 receptors in adjuvant-induced chronic arthritis: in vivo study using gene-deficient mice[J]. Journal of Pharmacology and Experimental Therapeutics 314, 111119.CrossRefGoogle ScholarPubMed
Hsieh, WS et al. (2017) TDAG8, TRPV1, and ASIC3 involved in establishing hyperalgesic priming in experimental rheumatoid arthritis[J]. Scientific Reports 7, 8870.CrossRefGoogle ScholarPubMed
Miller, VE, Rogers, K and Muirden, KD (1993) Detection of tumour necrosis factor alpha and interleukin-1 beta in the rheumatoid osteoarthritic cartilage-pannus junction by immunohistochemical methods[J]. Rheumatology International 13, 7782.CrossRefGoogle ScholarPubMed
Russell, FA et al. (2009) Tumour necrosis factor alpha mediates transient receptor potential vanilloid 1-dependent bilateral thermal hyperalgesia with distinct peripheral roles of interleukin-1beta, protein kinase C and cyclooxygenase-2 signalling[J]. Pain 142, 264274.CrossRefGoogle ScholarPubMed
Murai, M et al. (2008) SA13353 (1-[2-(1-adamantyl)ethyl]-1-pentyl-3-[3-(4-pyridyl)propyl]urea) inhibits TNF-alpha production through the activation of capsaicin-sensitive afferent neurons mediated via transient receptor potential vanilloid 1 in vivo[J]. European Journal of Pharmacology 588, 309315.CrossRefGoogle ScholarPubMed
Engler, A et al. (2007) Expression of transient receptor potential vanilloid 1 (TRPV1) in synovial fibroblasts from patients with osteoarthritis and rheumatoid arthritis[J]. Biochemical and Biophysical Research Communications 359, 884888.CrossRefGoogle ScholarPubMed
Terenzi, R et al. (2013) Neuropeptides activate TRPV1 in rheumatoid arthritis fibroblast-like synoviocytes and foster IL-6 and IL-8 production[J]. Annals of the Rheumatic Diseases 72, 11071109.CrossRefGoogle ScholarPubMed
Lowin, T, Pongratz, G and Straub R, H (2016) The synthetic cannabinoid WIN55,212-2 mesylate decreases the production of inflammatory mediators in rheumatoid arthritis synovial fibroblasts by activating CB2, TRPV1, TRPA1 and yet unidentified receptor targets[J]. Journal of Inflammation 13, 15.CrossRefGoogle ScholarPubMed
Galindo, T, Reyna, J and Weyer, A (2018) Evidence for transient receptor potential (TRP) channel contribution to arthritis pain and pathogenesis[J]. Pharmaceuticals 11, 105.CrossRefGoogle ScholarPubMed
Hu, F et al. (2017) Hypotonic stress promotes ATP release, reactive oxygen species production and cell proliferation via TRPV4 activation in rheumatoid arthritis rat synovial fibroblasts[J]. Biochemical and Biophysical Research Communications 486, 108115.CrossRefGoogle ScholarPubMed
Schaible, HG (2014) Nociceptive neurons detect cytokines in arthritis[J]. Arthritis Research & Therapy 16, 470.CrossRefGoogle ScholarPubMed
Itoh, Y et al. (2009) An environmental sensor, TRPV4 is a novel regulator of intracellular Ca2+ in human synoviocytes[J]. American Journal of Physiology. Cell Physiology 297, C1082C1090.CrossRefGoogle ScholarPubMed
李明遥,赵毅,罗玉斌等. (2019) 瞬时受体电位 M_2 对抗原诱导关节炎小鼠的作用及机制研究初探[J]. 中华内科杂志 2019, 911912.Google Scholar
Ciurtin, C et al. (2010) TRPM3 channel stimulated by pregnenolone sulphate in synovial fibroblasts and negatively coupled to hyaluronan[J]. BMC Musculoskeletal Disorders 11, 111.CrossRefGoogle ScholarPubMed
Stuhlmeier, KM (2006) Aspects of the biology of hyaluronan, a largely neglected but extremely versatile molecule[J]. Wiener Medizinische Wochenschrift 156, 563568.CrossRefGoogle ScholarPubMed
Ma, G et al. (2021) Blockade of TRPM7 alleviates chondrocyte apoptosis and articular cartilage damage in the adjuvant arthritis rat model through regulation of the Indian hedgehog signaling pathway[J]. Frontiers in Pharmacology 12, 655551.CrossRefGoogle ScholarPubMed
Zhou, R et al. (2022) TRPM7 channel inhibition attenuates rheumatoid arthritis articular chondrocyte ferroptosis by suppression of the PKCalpha-NOX4 axis[J]. Redox Biology 55, 102411.CrossRefGoogle ScholarPubMed
Wang, CH et al. (2014) CD147 up-regulates calcium-induced chemotaxis, adhesion ability and invasiveness of human neutrophils via a TRPM-7-mediated mechanism[J]. Rheumatology 53, 22882296.CrossRefGoogle Scholar
Li, X et al. (2014) Inhibition of transient receptor potential melastatin 7 (TRPM7) channel induces RA FLSs apoptosis through endoplasmic reticulum (ER) stress[J]. Clinical Rheumatology 33, 15651574.CrossRefGoogle ScholarPubMed
Zhu, S et al. (2014) Involvement of transient receptor potential melastatin-8 (TRPM8) in menthol-induced calcium entry, reactive oxygen species production and cell death in rheumatoid arthritis rat synovial fibroblasts[J]. European Journal of Pharmacology 725, 19.CrossRefGoogle ScholarPubMed
Horvath, A et al. (2016) Transient receptor potential ankyrin 1 (TRPA1) receptor is involved in chronic arthritis: in vivo study using TRPA1-deficient mice[J]. Arthritis Research & Therapy 18, 6.CrossRefGoogle ScholarPubMed
Pereira, I et al. (2017) Transient receptor potential ankyrin 1 channel expression on peripheral blood leukocytes from rheumatoid arthritic patients and correlation with pain and disability[J]. Frontiers in Pharmacology 8, 53.CrossRefGoogle ScholarPubMed
Lowin, T et al. (2018) Selective killing of proinflammatory synovial fibroblasts via activation of transient receptor potential ankyrin (TRPA1)[J]. Biochemical Pharmacology 154, 293302.CrossRefGoogle Scholar
Nedunchezhiyan, U et al. (2022) Obesity, inflammation, and immune system in osteoarthritis[J]. Frontiers in Immunology 13, 907750.CrossRefGoogle ScholarPubMed
de Sousa, VJ et al. (2020) Examining the role of transient receptor potential canonical 5 (TRPC5) in osteoarthritis[J]. Osteoarthritis and Cartilage Open 2, 100119.CrossRefGoogle Scholar
Lv, Z et al. (2021) TRPV1 alleviates osteoarthritis by inhibiting M1 macrophage polarization via Ca(2+)/CaMKII/Nrf2 signaling pathway[J]. Cell Death & Disease 12, 504.CrossRefGoogle ScholarPubMed
Sun, H et al. (2022) Blocking TRPV4 ameliorates osteoarthritis by inhibiting M1 macrophage polarization via the ROS/NLRP3 signaling pathway[J]. Antioxidants. 11, 2315.CrossRefGoogle ScholarPubMed
姚旺祥,戴晗豪,董佩龙等. (2020) TRPV4 在骨关节炎与正常软骨中的表达差异及意义[J]. 中国修复重建外科杂志 34, 6368.Google Scholar
O'Conor, CJ et al. (2016) Cartilage-specific knockout of the mechanosensory ion channel TRPV4 decreases age-related osteoarthritis[J]. Scientific Reports 6, 29053.CrossRefGoogle ScholarPubMed
Hinata, M et al. (2018) Sensitization of transient receptor potential vanilloid 4 and increasing its endogenous ligand 5,6-epoxyeicosatrienoic acid in rats with monoiodoacetate-induced osteoarthritis[J]. Pain 159, 939947.CrossRefGoogle ScholarPubMed
Soga, M et al. (2021) Suppression of joint pain in transient receptor potential vanilloid 4 knockout rats with monoiodoacetate-induced osteoarthritis[J]. Pain Reports 6, e951.CrossRefGoogle ScholarPubMed
Atobe, M et al. (2019) Discovery of novel transient receptor potential vanilloid 4 (TRPV4) agonists as regulators of chondrogenic differentiation: identification of quinazolin-4(3 H)-ones and in vivo studies on a surgically induced rat model of osteoarthritis[J]. Journal of Medicinal Chemistry 62, 14681483.CrossRefGoogle ScholarPubMed
Clark, AL et al. (2010) Chondroprotective role of the osmotically sensitive ion channel transient receptor potential vanilloid 4: age- and sex-dependent progression of osteoarthritis in Trpv4-deficient mice[J]. Arthritis and Rheumatism 62, 29732983.CrossRefGoogle ScholarPubMed
Wei, Y et al. (2018) Transient receptor potential channel, vanilloid 5, induces chondrocyte apoptosis in a rat osteoarthritis model through the mediation of Ca2 + influx[J]. Cellular Physiology and Biochemistry 46, 687698.CrossRefGoogle Scholar
Wei, Y et al. (2017) Transient receptor potential vanilloid 5 mediates Ca2 + influx and inhibits chondrocyte autophagy in a rat osteoarthritis model[J]. Cellular Physiology and Biochemistry 42, 319332.CrossRefGoogle Scholar
Wei, Y et al. (2018) The transient receptor potential channel, vanilloid 5, induces chondrocyte apoptosis via Ca2 + CaMKII-dependent MAPK and Akt/mTOR pathways in a rat osteoarthritis model[J]. Cellular Physiology and Biochemistry 51, 23092323.CrossRefGoogle Scholar
Song, T et al. (2017) Regulation of chondrocyte functions by transient receptor potential cation channel V6 in osteoarthritis[J]. Journal of Cellular Physiology 232, 31703181.CrossRefGoogle ScholarPubMed
沙一帆,邓国英,王秋根等. (2016) 瞬时受体电位通道 M8 在冷刺激致骨关节炎中的作用[J]. 国际骨科学杂志 37, 383387.Google Scholar
Mcgaraughty, S et al. (2010) TRPA1 modulation of spontaneous and mechanically evoked firing of spinal neurons in uninjured, osteoarthritic, and inflamed rats[J]. Molecular Pain 6, 14.CrossRefGoogle ScholarPubMed
Nummenmaa, E et al. (2020) Transient receptor potential ankyrin 1 (TRPA1) is involved in upregulating interleukin-6 expression in osteoarthritic chondrocyte models[J]. International Journal of Molecular Sciences 22, 87.CrossRefGoogle ScholarPubMed
Zheng, Z et al. (2022) In vivo inhibition of TRPC6 by SH045 attenuates renal fibrosis in a New Zealand obese (NZO) mouse model of metabolic syndrome[J]. International Journal of Molecular Sciences 23, 6870.CrossRefGoogle Scholar
Wu, YL et al. (2017) Inhibition of TRPC6 channels ameliorates renal fibrosis and contributes to renal protection by soluble klotho[J]. Kidney International 91, 830841.CrossRefGoogle ScholarPubMed
Ramirez, GA et al. (2015) TRPC6 gene variants and neuropsychiatric lupus[J]. Journal of Neuroimmunology 288, 2124.CrossRefGoogle ScholarPubMed
Ramirez, GA et al. (2018) The TRPC6 intronic polymorphism, associated with the risk of neurological disorders in systemic lupus erythematous, influences immune cell function[J]. Journal of Neuroimmunology 325, 4353.CrossRefGoogle ScholarPubMed
Xiao, F et al. (2021) IL-17 drives salivary gland dysfunction via inhibiting TRPC1-mediated calcium movement in Sjogren's syndrome[J]. Clinical & Translational Immunology 10, e1277.CrossRefGoogle ScholarPubMed
Liu, X et al. (2007) Attenuation of store-operated Ca2+current impairs salivary gland fluid secretion in TRPC1(−/−) mice[J]. Proceedings of the National Academy of Sciences of the USA 104, 1754217547.CrossRefGoogle ScholarPubMed
Lee, KP et al. (2014) Molecular determinants mediating gating of transient receptor potential canonical (TRPC) channels by stromal interaction molecule 1 (STIM1)[J]. Journal of Biological Chemistry 289, 63726382.CrossRefGoogle Scholar
Liu, X, Ong, HL and Ambudkar, I (2018) TRP channel involvement in salivary glands – some good, some bad[J]. Cells 7, 74.CrossRefGoogle ScholarPubMed
Wang, B et al. (2020) Role of T cells in the pathogenesis and treatment of gout[J]. International Immunopharmacology 88, 106877.CrossRefGoogle ScholarPubMed
Chen, CC et al. (2019) fMRI indicates cortical activation through TRPV1 modulation during acute gouty attacks[J]. Scientific Reports 9, 12348.CrossRefGoogle ScholarPubMed
Xu, X et al. (2022) Regulation of TRPV1 channel in monosodium urate-induced gouty arthritis in mice[J]. Inflammation Research 71, 485495.CrossRefGoogle ScholarPubMed
Zhong, Z et al. (2013) TRPM2 links oxidative stress to NLRP3 inflammasome activation[J]. Nature Communications 4, 1611.CrossRefGoogle ScholarPubMed
Yin, C et al. (2020) IL-33/ST2 induces neutrophil-dependent reactive oxygen species production and mediates gout pain[J]. Theranostics 10, 1218912203.CrossRefGoogle ScholarPubMed
Trevisan, G et al. (2013) Transient receptor potential ankyrin 1 receptor stimulation by hydrogen peroxide is critical to trigger pain during monosodium urate-induced inflammation in rodents[J]. Arthritis and Rheumatism 65, 29842995.CrossRefGoogle ScholarPubMed
Moilanen, LJ et al. (2015) Urate crystal induced inflammation and joint pain are reduced in transient receptor potential ankyrin 1 deficient mice – potential role for transient receptor potential ankyrin 1 in gout[J]. PLoS ONE 10, e117770.CrossRefGoogle ScholarPubMed
Trevisan, G et al. (2014) TRPA1 receptor stimulation by hydrogen peroxide is critical to trigger hyperalgesia and inflammation in a model of acute gout[J]. Free Radical Biology & Medicine 72, 200209.CrossRefGoogle Scholar
Jiang, F et al. (2021) The mechanosensitive Piezo1 channel mediates heart mechano-chemo transduction[J]. Nature Communications 12, 869.CrossRefGoogle ScholarPubMed
Alper, SL (2017) Genetic diseases of PIEZO1 and PIEZO2 dysfunction[J]. Current Topics in Membranes 79, 97134.CrossRefGoogle ScholarPubMed
Wang, L et al. (2020) Mechanical sensing protein PIEZO1 regulates bone homeostasis via osteoblast-osteoclast crosstalk[J]. Nature Communications 11, 282.CrossRefGoogle ScholarPubMed
Chesler, AT et al. (2016) The role of PIEZO2 in human mechanosensation[J]. New England Journal of Medicine 375, 13551364.CrossRefGoogle ScholarPubMed
Woo, SH et al. (2015) Piezo2 is the principal mechanotransduction channel for proprioception[J]. Nature Neuroscience 18, 17561762.CrossRefGoogle ScholarPubMed
Marshall, KL et al. (2020) PIEZO2 in sensory neurons and urothelial cells coordinates urination[J]. Nature 588, 290295.CrossRefGoogle ScholarPubMed
Nonomura, K et al. (2017) Piezo2 senses airway stretch and mediates lung inflation-induced apnoea[J]. Nature 541, 176181.CrossRefGoogle ScholarPubMed
Ghitani, N and Chesler, AT (2019) The anatomy of the baroreceptor reflex[J]. Cell Reports 29, 21212122.CrossRefGoogle ScholarPubMed
Grotle, AK et al. (2020) Effects of type 1 diabetes on reflexive cardiovascular responses to intermittent muscle contraction[J]. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology 319, R358R365.CrossRefGoogle ScholarPubMed
Matteucci, E et al. (2006) Systolic blood pressure response to exercise in type 1 diabetes families compared with healthy control individuals[J]. Journal of Hypertension 24, 17451751.CrossRefGoogle ScholarPubMed
Grotle, AK et al. (2021) GsMTx-4 normalizes the exercise pressor reflex evoked by intermittent muscle contraction in early stage type 1 diabetic rats[J]. American Journal of Physiology. Heart and Circulatory Physiology 320, H1738H1748.CrossRefGoogle ScholarPubMed
Velasco-Estevez, M et al. (2020) Inhibition of Piezo1 attenuates demyelination in the central nervous system[J]. Glia 68, 356375.CrossRefGoogle ScholarPubMed
Jairaman, A et al. (2021) Piezo1 channels restrain regulatory T cells but are dispensable for effector CD4(+) T cell responses[J]. Science Advances 7, eabg5859.CrossRefGoogle ScholarPubMed
Yang, K et al. (2022) The emerging roles of piezo1 channels in animal models of multiple sclerosis[J]. Frontiers in Immunology 13, 976522.CrossRefGoogle ScholarPubMed
Ikeda, R, Arimura, D and Saito, M (2021) Expression of Piezo mRNA is unaffected in a rat model of knee osteoarthritis[J]. Molecular Pain 17, 794215339.CrossRefGoogle Scholar
Lee, W et al. (2021) Inflammatory signaling sensitizes Piezo1 mechanotransduction in articular chondrocytes as a pathogenic feed-forward mechanism in osteoarthritis[J]. Proceedings of the National Academy of Sciences of the USA 118, e2001611118.CrossRefGoogle ScholarPubMed
Lee, W et al. (2014) Synergy between Piezo1 and Piezo2 channels confers high-strain mechanosensitivity to articular cartilage[J]. Proceedings of the National Academy of Sciences of the USA 111, E5114E5122.CrossRefGoogle ScholarPubMed
Lee, W, Guilak, F and Liedtke, W (2017) Role of piezo channels in joint health and injury[J]. Current Topics in Membranes 79, 263273.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Roles of TRP receptor subtypes in T1DM.Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. DN, diabetic nephropathy; ICA, islet cell antibodies; IAA, insulin autoantibodies; GAD-Ab, glutamate decarboxylase antibodies.

Figure 1

Figure 2. Roles of TRP receptor subtypes in T2DM.Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. CGRP, calcitonin gene-related peptide; TGF-β1, transforming growth factor-β1.

Figure 2

Figure 3. Roles of TRP receptor subtypes in IBD.Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. DSS, dextran sodium sulfate; DNBS, dinitrobenzenesulfonate; CGRP, calcitonin gene-related peptide; NF-κB, nuclear factor kappa-B; TNF-α, tumour necrosis factor alpha; IL, interleukin; MCP-1, monocyte chemoattractant protein-1.

Figure 3

Figure 4. Roles of different TRP receptor subtypes in MS.Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. EAE, experimental autoimmune encephalomyelitis; PMS, progressive multiple sclerosis; RR-EAE, relapsing-remitting experimental autoimmune encephalomyelitis; CSPG, chondroitin sulphate proteoglycans; IL, interleukin; IFN-γ, interferon gamma; PP2A, protein phosphatase 2A.

Figure 4

Figure 5. Roles of TRP receptor subtypes in RA.Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. TNF-α, tumour necrosis factor alpha; IL, interleukin; PGE2, prostaglandin E2; ROS, reactive oxygen species; NE, neutrophil; Mø, macrophage.

Figure 5

Figure 6. Roles of TRP receptor subtypes in OA.Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. MMP, matrix metalloproteinase; IL, interleukin; ROS, reactive oxygen species.

Figure 6

Figure 7. Roles of TRP receptor subtypes in SS.Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. SOCE, store-operated Ca2 + entry.

Figure 7

Figure 8. Roles of TRP receptor subtypes in gout.Note: (+) means to promote the onset of disease, and (−) means to inhibit the onset of disease. MSU, monosodium urate; IL, interleukin.

Figure 8

Table 1. Role of TRP receptors in organ-specific autoimmune diseases

Figure 9

Table 2. Role of TRP receptors in systemic autoimmune disease