1,059
Views
0
CrossRef citations to date
0
Altmetric
Review

Roles of TRPM channels in glioma

, , , , , , , , ORCID Icon & show all
Article: 2338955 | Received 03 Dec 2023, Accepted 01 Apr 2024, Published online: 29 Apr 2024

ABSTRACT

Gliomas are the most common type of primary brain tumor. Despite advances in treatment, it remains one of the most aggressive and deadly tumor of the central nervous system (CNS). Gliomas are characterized by high malignancy, heterogeneity, invasiveness, and high resistance to radiotherapy and chemotherapy. It is urgent to find potential new molecular targets for glioma. The TRPM channels consist of TRPM1-TPRM8 and play a role in many cellular functions, including proliferation, migration, invasion, angiogenesis, etc. More and more studies have shown that TRPM channels can be used as new therapeutic targets for glioma. In this review, we first introduce the structure, activation patterns, and physiological functions of TRPM channels. Additionally, the pathological mechanism of glioma mediated by TRPM2, 3, 7, and 8 and the related signaling pathways are described. Finally, we discuss the therapeutic potential of targeting TRPM for glioma.

KEY POINTS

•TRPM channels are widely expressed in the human body and play an important role in gliomas.

• Abnormal expression of TRPM2, 3, 7, and 8 channels in gliomas is associated with disease severity and prognosis.

•TRPM2, 3, 7, and 8 channels are effective targets in glioma.

1. Introduction

An ion channel is a protein consisting of pores that can regulate and passively ion flux through a biofilm (as determined by an electrochemical gradient).Citation1 As far, there are more than 400 known ion channels, which belong to different families and are involved in a wide range of physiological processes.Citation2 Meanwhile, abnormal expression of ion channels is closely related to many diseases, including cancer, hypertension, inflammatory pain, diabetes, and neurodegenerative diseases, suggesting that they may be fascinating therapeutic targets.Citation3–5

TRP channels are transmembrane ion channels that allow the nonselective passage of cations through the cell membrane.Citation6 So far, more than 30 members of the TRP channel family have been cloned in mammals. These channels span a range of conformational states from closed, open, and partially open.Citation7 Since 2017, significant progress has been made in studying the structural biology of the TRPM subfamily using cryo-electron microscopy.Citation8 There are four independent TRPM ion channels in which the cryo-EM structures have been solved.Citation9 The TRPM channels represent one of the largest and most diverse subfamilies of the TRP superfamilies and are expressed in almost all cell types.Citation10 In recent years, the TRPM subfamily has attracted considerable attention due to its involvement in several physiological and pathological processes, including temperature sensing,Citation11 cancer progression,Citation12 vascular development,Citation13 neurological diseases,Citation14 endothelial dysfunctionCitation15 and numerous others.Citation16,Citation17 This makes TRPM channels a fascinating group of ion channels with a high biomedical projection.Citation2 As such, the TRPM channels have inspired scientists’ enthusiasm for functional and structural studies.

Over the past decade, TRPM channels have been shown to play a critical role in tumorigenesis, including gliomas.Citation18–20 Gliomas, including anaplastic astrocytoma and glioblastoma, are the most common types of primary brain tumors.Citation21 Chemotherapy is a major treatment for glioma besides surgery. Due to the lack of drugs with blood-brain barrier permeability, the clinical efficacy of glioma is greatly limited.Citation22 Therefore, there is a need to search for new targets and drugs that are effective against glioblastoma. Among them, the TRPM channel represents a new intervention target. As epithelial cells evolve from normal to tumor growth, genetic changes can affect TRPM expression or lead to changes in TRPM channel activity.Citation23 The TRPM channel is expressed in both normal and tumor tissues of the brain, which can regulate glioma growth and improve migration and invasion ability.Citation24

Research confirmed that TRPM2 and TRPM3 have anti-tumor effects, while TRPM7 and TRPM8 may be related to glioma malignancy (such as proliferation).Citation25 For example, Ca2+ permeates TRPM2 channels to enhance H2O2-induced human A172 glioma cell death.Citation26 TRPM7 suppression significantly inhibited the growth, proliferation, migration and invasion of A172 cells.Citation27 In addition, the expression level of TRPM8 was significantly correlated with the aggressiveness of glioma cells.Citation28 These make the TRPM channels a potential prognostic indicator and therapeutic target.

This article reviews the research progress of TRPM subfamily members in recent years, focusing on the pathological function, related signaling pathways, and application of related drugs of the TRPM channels in glioma.

2. The TRP superfamily

The TRP channel superfamily was first discovered in mutant Drosophila melanogaster in 1969.Citation29,Citation30 Minke et al. named it TRP according to its electrophysiological phenotype.Citation31 Electroretinogram measurements revealed a transient response to a steady light stimulus in vision-impaired mutant flies compared to wild flies, rather than the sustained voltage response typical of wild flies.Citation32,Citation33 In 1989, Montell and Rubin discovered the TRP gene and described its molecular characteristics.Citation34 In 1992, the first homolog Trpl was found. Then, in 1993, the TRP family was identified as a new superfamily of ion channels, and its role in Ca2+ permeability was identified.Citation35 In 1995, the first human homolog, transient receptor potential channel associated protein 1 (TRPC1), was identified.Citation36 Since then, TRP channels have been found to present in large amounts in multicellular organisms, including but not limited to zebrafish,Citation37 humans,Citation38 mice,Citation39 and worms.Citation40 Unlike other ion channels, it is defined by the membrane’s sequence homology and topological structure.Citation41,Citation42 The structure of TRP has been highly conserved during the evolution from nematodes to flies and humans.Citation43 Based on the amino acid homology of TRP channels, they were further divided into seven subfamilies: vanilloid (TRPV1–6), Canonical (TRPC1–7), melastatin (TRPM1–8), Mucolipin (TRPML1–3), Polycystin (TRPP1–3), Anchor protein Type (Ankyrin, TRPA1), TRPNs and TRPA1s.Citation44 The subfamilies showed remarkable differences in the structure and sequence homology, resulting in different physiological functions.Citation29,Citation45 Some TRPs (e.g., TRPC1 and TRPM7) are widely expressed, while others are restricted to specific organs (e.g., TRPV5 is only expressed in the human kidney).Citation46 Many diseases are associated with TRP channel dysfunction, such as surface-level abnormalities, cell translocation, or mutation, suggesting its essential role in homeostasis and disease in vivo.Citation47

TRP exists in the plasma membrane as six transmembrane domain (S1-S6) polypeptide subunits. It requires four subunits to assemble homologous or heterotypic oligomeric pores into functional channels.Citation48 S4 corresponds to a region similar to a voltage sensor that senses changes in intracellular ion concentrations.Citation2 Channel hole by the S5 and S6 subunits between alpha spiral ring formation.Citation49 Differences in subunit composition may lead to differences in the biological characteristics of TRP channels.Citation50 Furthermore, TRP channels can permeate both monovalent and divalent ions, with some notable exceptions. TRPM4 and TRPM5 only pass through monovalent cations, while TRPV5 and TRPV6 show unusually high selectivity for Ca2+.Citation51,Citation52

Since Ca2+ is both the primary charge carrier and the most important second messenger, the TRP superfamily naturally becomes fundamental to the survival and function of cells.Citation53 Moreover, TRP channels in muscle contraction, ion homeostasis, bone remodeling, and vasomotor control also play an essential role.Citation54 TRP channels have many binding partners, mainly in channel regulation.Citation55 When activated, TRP channels allow cations to flow in, leading to cell depolarization.Citation56,Citation57 Different cations have different permeability between channels, leading to various cellular effects.Citation58 For example, Ca2+ and Mg2+ ions cross the cell membrane through TRP channels and play an essential role in various physiological processes, including cell proliferation, gene transcription, muscle contraction, and cell death.Citation59 Early in the disease, the release of various endogenous substances can affect TRP pathway function, leading to infection and disease progression (for example, the release of leukotriene B4 induces TRPV1 activation).Citation60 One of the most critical pathological processes is the development of cancer cells. More and more evidence has shown that changes in TRP channel expression levels play a crucial role in the occurrence and development of tumors, including proliferation,Citation61 migration,Citation62 invasion,Citation63 and other characteristics of glioma. Under the background of brain tumors, all TRP channels have been studied for their potential role in cancer progression or as therapeutic targets.

3. TRPM subfamily

As mentioned, the TRPM channels represent one of the TRP superfamily’s largest and most diverse subfamilies.Citation64 Since TRPM1 was first identified in 1989, much progress has been made in identifying new members and functions of the TRPM channels.Citation2 TRPM subfamily consists of eight members, ranging from TRPM1 to TRPM8.Citation65 These genes are widely expressed in the human body and involve numerous physiological and pathological features, particularly in ion homeostasisCitation29 and cell development.Citation66 TRPM channels promote intracellular Ca2 + signal in response to lipid, ion concentration, or second messenger to regulate these functions.Citation10 Besides TRPM6 and TRPM7 being physically associated with bivalent on the membrane potential of selective, most of the channels are nonselective channels.Citation67 Except for TRPM4 and TRPM5, other TRPM channels can be permeated by Ca2+, which are univalent cation channels activated by Ca2+ and functionally guide Na+ ions.Citation68 Despite some shared structural features, members of the TRPM channels are less conserved than other members of the TRP family.Citation69 According to sequence similarity, they were divided into four subgroups: 1) TRPM1 & TRPM3, 2) TRPM6 & TRPM7, 3) TRPM4 & TRPM5, 4) TRPM2 & TRPM8.Citation70 summarizes their relationship.

Figure 1. Members and general structure of the TRPM channels.

Figure 1. Members and general structure of the TRPM channels.

4. Structure and domain organization of the TRPM channels

Due to the highly sequentially conservative nature of the TRPM subfamily, the overall structure of most TRPM members is similar.Citation71 However, they also have some differences, which will be reviewed in detail from the following aspects: S1-S6 domain, N-terminal domain, and C-terminal domain, as shown in . The pore domain on the cell membrane is connected to S1-S4 by the S4-S5 domain. The S1-S4 domain contains ligand-binding sites, such as the agonist Ca2+. In addition, the S1-S4 domains of K+ and Ca2+ channels contain voltage sensors. The S5 and S6 domains form ionic conducting pores with the p ring.Citation10 As for the N-terminal, all TRPM channels have about 700 amino acids.Citation72 As for the C-terminus, except for TRPM1 and TRPM2, all other TRPM members contain a highly conserved TRP box and phosphatidylinositol 4,5-bisphosphate (PIP2) binding site. However, there are some structural differences between TRPM1 and TRPM2. For example, TRPM1 and TRPM2 have the binding site of PIP2,Citation7 but TRPM2 also has a NUDT9-H domain that can act as a binding site for ADP-ribose (ADPR).Citation73 In addition, calmodulin (CAM) binding sites and ATP binding sites were also found at the TRPM4 and TRPM5 channels, and these channels could affect the channel’s Ca2+ sensitivity.Citation74

Interestingly, the presence of C-terminal enzyme domains in 3 TRPM members, such as TRPM2, TRPM6, and TRPM7, is particularly notable, the only characteristic of all known ion channels to date. Specifically, TRPM2 possesses a domain at this location with a large amount of homology with NUDT9, a Nudix enzyme with ADP-nuclear hydrolase activity, while TRPM6 and TRPM7 also contain kinase domains and belong to atypical α-protein kinases.Citation75

5. Activation mechanisms of the TRPM channels

The TRPM channels are essential cellular receptors that can permeate Mg2+, Ca2+, Zn2+, and Na+ cations. As shown in , different physical or chemical stimuli, such as temperature, osmolality, and PH, can activate TRPM channels.Citation76

Figure 2. Activation mechanisms of the TRPM channels.

Figure 2. Activation mechanisms of the TRPM channels.

Although TRPM1 was first discovered, the mechanism of TRPM1 channel activation has yet to be fully elucidated. Studies have found that the change in ion concentration can also negatively impact channel activity. [Ca2+] cyt inhibits the increase of TRPM1 channels, and intracellular Zn2+ blocks the increase of TRPM1 channels.Citation77

Because TRPM2 is widely distributed and has different physiological effects, its activation involves a variety of external stimuli. There is some controversy in the literature regarding TRPM2 activation, particularly nucleotide activation. ADPR generally has the highest affinity of TRPM2 receptor agonists (EC50, 10–80 μM).Citation78,Citation79 Other more controversial activators include nicotinate adenine dinucleotide (NAAD), cyclic ADPR (cADPR), nicotinamide adenine dinucleotide (NAD), and NAAD-phosphate (NAADP).Citation80 Due to the low affinity of some of these activators, their direct role in TRPM2 activation has been controversial. However, most of them use ADPR as the activation mechanism. Recent evidence suggests that TRPM2 channel activation is related to its enzymatic domain.Citation81 Extracellular signaling stimulates the production of ADPR in mitochondria by activating poly (ADPR) polymerase (PARP) or poly (ADPR) glycohydrolase (PARG), which subsequently binds to the C-terminal NUDT9-H domain of TRPM2 to activate the channel.Citation82,Citation83 Previous studies have found that cADPR and pyridine dinucleotides can activate TRPM2 or promote ADPR activation. However, when contaminated ADPR was purified with either nucleotide phosphatase or ADPR hydrolase, neither stimulated TRPM2 activation.Citation80 These evidence suggests that ADPR is an activator of TRPM2. In addition to ADPR, increased intracellular Ca2+ positively modulates TRPM2.Citation84 The initial calcium entry via ADPR-bound TRPM2 or a Ca2+ spark activation channel starts from cytosolic activity.Citation7 Ca2±bound calmodulin then combined with the N-terminal IQ motif of TRPM2, providing positive feedback for activating the TRPM2 ion channel and increased Ca2+ influx.Citation85 However, in the absence of internal or external Ca2+, ADPR is ineffective in activating TRPM2 channels.Citation86 Furthermore, PIP2 has been found to affect the sensitivity of TRPM2 to Ca2+ activation.Citation82 Other intracellular Ca2±dependent activators make TRPM2 sensitive to ADPR by interacting with CaM and can also splice ADPR-free TRPM2 in their isoforms.Citation78 It has also been found that TRPM2 is sensitive to temperature, and the channel activity is inhibited by acidification.Citation87 Finally, other external activators include ROS, tumor necrosis factor-α (TNFα), reactive nitrogen (RNS), Zn2+, Falcon A, amyloid beta peptide, and H2O2.Citation88

TRPM3 channels can be activated by CIM0216, hypotonic solution, and pregnenolone sulfate (PS). When activated, it induces an increase in [Ca2+] cyt, leading to Ca2+/calmodulin regulation and subsequent activation of mitogen-activated protein kinases (MAPKs).Citation89 It is also activated by plant-derived and synthetic compounds, including iodine, cholesterol, troglitazone, and rosiglitazone.Citation90 In addition, changes in ion concentration can also affect channel activity. Several studies have found that [Ca2+] cyt and intracellular Mg2+ ions inhibit the activity of TRPM3 channels.Citation77,Citation91

Both TRPM4 and TRPM5 require an increase in intracellular Ca2+ levels for activation. Both are regulated by PIP2 but differ in their high sensitivity to Ca2+.Citation92 TRPM4, identified as a homolog of MLSN (TRPM1), is highly expressed in the colon, prostate, kidney, and heart.Citation93 The c-terminal CaM binding site of TRPM4 is critical for susceptibility to Ca2±dependent activation. For TRPM5, TRPM5 is 5 to 10 times more sensitive to Ca2+ than the TRPM4 channels.Citation2 Initially identified as MTR1, TRPM5 was later reported to co-express with the taste-signaling molecule α-gustducin.Citation94 Nevertheless, both TRPM4 and TRPM5 have a direct Ca2±binding site on the inner side of the cell in the S1-S4 domain. A recent study found that SOCE may be a source of Ca2+ supply, with TRPM4/5 mediated depolarization providing negative feedback for Ca2+ inflow through the SOCE channel.Citation95 Furthermore, low micromolar AMP, ADP, and ATP inhibit TRPM4 but not TRPM5 and their activity is enhanced via phosphorylation of the protein kinase C (PKC)-dependent TRP domain.Citation10

TRPM6 and its homology TRPM7 are activated when Mg2+ and Mg ATP levels are reduced in the cytoplasm.Citation96 TRPM6 channels are inactive at basal levels of Mg2+ and lack sensitivity to Mg2+. However, TRPM6 is activated in the oligomeric form in response to the phosphorylation of TRPM7.Citation97 As for TRPM7, other activators reported in the literature include Naltriben, prostaglandin E2, AMP, ADP, ATP, mechanical stimulation, etc.Citation98 Notably, although Naltriben is highly similar in structure to other opioid receptor antagonists, such as NTX, no other opioid receptor antagonists activate TRPM7 channels.Citation99

TRPM8 is probably the best-studied member of the TRPM channels. It is a cold receptor, which can be activated directly by hypothermia and chemical agonists such as menthol and modulated by critical molecules such as PIP2 and Ca2+.Citation100 The sensitive region of PIP2 located in the C-terminal region is the determining factor of temperature sensitivity. For TRPM8 channels, PIP2 is a coolant agonist and a cofactor of the cryogenic activation channel.Citation101 While PIP2 alone may be enough to activate TRPM8, the loss of base levels of PIP2 leads to channel desensitization. Moreover, activating TRPM8 by icilin requires basal cytoplasmic Ca2+ but does not require menthol activity.Citation102

6. Molecular biological functions of the TRPM channels

In humans, TRPM1 is located on chromosome 15 and consists of 27 exons with a gene length of more than 58 KB.Citation103 The TRPM1 gene has an mRNA transcription volume of about 5.4 KB and encodes the 1603 aa protein of approximately 182 kDa, expressed only in pigmentation cells of the skin and eyes.Citation104 TRPM1 also has a short N-terminal isoform called MLSN1-S, which lacks all the transmembrane domains that produce 500aa proteins.Citation105 It has been reported that MLSN1-S is located in the cytoplasm, while L-type (MLSN1-L) is located in the cell membrane.Citation20

First reported by Nagamine et al., TRPM2 was identified as LTRPC and TRPC7.Citation106 In CNS, TRPM2 is expressed in multiple regions, including the cortex, substantia nigra, spinal cord, hippocampus, and striatum.Citation107 TRPM2 can also be detected in peripheral tissues such as the heart, liver, pancreas, and lungs.Citation108 This widespread expression reflects the involvement of TRPM2 in various pathological and physiological processes. Moreover, the channel is expressed in mononuclear cell lines, including multiple cultures of macrophage cell lines, peripheral blood mononuclear cells, and neutrophils.Citation109 TRPM2 channels exhibit calcium, cation channel, and ADP-ribose pyrophosphatase activity through C-terminal enzyme domains. The domain hydrolyzes ADP-ribose to ribose-phosphate and AMP but at a much lower efficiency than the known Nudix enzymes.Citation110

The TRPM3 is expressed in the human brain, as well as in the kidneys of mice and humans.Citation111 Differences in the transcript of the TRPM3 mRNA and protein products may lead to diverse molecular mechanisms, such as the induction of short variants by hypotonic solutions or the storage depletion of lengthy variants. This fact, coupled with the specific expression of TRPM3 in the kidney, suggests that this channel is involved in renal osmotic homeostasis. Due to alternative splicing, the TRPM3 gene produces many different mRNAs.Citation112 Although Lee et al. cloned six variants from human kidneys, three transcripts of varying lengths were detected in the mouse brains.Citation113 Different starting positions and C-terminus give TRPM3 transcripts a high degree of variability. In addition, TRPM3 ion channels function independently of TRPV1’s thermal sensor.Citation114 TRPM4 and TRPM5 are calcium-activated sodium ion channels impermeable to calcium and mediate plasma membrane depolarization.Citation115 The binding sites of CaM, PKC phosphorylation sites, and ATP in TRPM4 agree with the regulatory activity of calcium, AMP, ADP, ATP, and Ca2+ sensitivities in the channel.Citation20 Furthermore, TRPM4 has ATP-binding sites that may inhibit TRPM4 by stabilizing channels in the apolipoprotein-like conformation.Citation116 Selective expression of TRPM5 was initially reported in taste buds, suggesting that it may be involved in taste transduction.Citation117 Subsequently, the immune reactivity of TRPM5 was found in the olecranon and nose. It suggested that TRPM5 is an inherent signaling component of the mammalian chemosensory organs.Citation118 TRPM4 and TRPM5 are both voltage-gated and regulated by PIP2 and phospholipase C (PLC). Moreover, these two channels play a role in T lymphocyte secretion, cytokine secretion, and myogenic contraction of cerebral arteries.Citation119

TRPM6 and TRPM7 are most closely related to TRPM1 and TRPM3. In fact, despite their different C-terminal domains, they share the same N-terminal sequence.Citation120 Furthermore, these four channels are similar in the small cytoplasmic region, the putative channel, and the transmembrane region after the sixth transmembrane region. TRPM6 and TRPM7 are closely related, and both have a protein kinase domain at the C-terminus. This domain has been shown to form autophosphorylated dimers and phosphorylate protein substrates.Citation121 TRPM6 and TRPM7 preferentially penetrate Mg2+ and Ca2+ but may be the most important for maintaining physiological Mg2+ homeostasis. In addition to a wide variety of divalent positive ions, TRPM7 channels can penetrate univalent ions, especially H +.Citation122 Interestingly, TRPM6 has been found to interact specifically with the homolog TRPM7, leading to the formation of functional TRPM6/TRPM7 complexes in cell membranes. In the presence of Mg2+ and Mg2+·ATP, the binding of mTRPM6 and mTRPM7 results in the high intrinsic activity of mTRPM6/7 at the physiological level.Citation123

TRPM8 is a homotetramer composed of 130 kDa subunits. It is located on chromosome 2 and contains 102 kilobases and 27 exons.Citation124 Multiple transcription factor binding sites on its promoter are famous, such as MYC, NKX3–1, and USF1. TRPM8 forms 11 mRNA isomers by alternative splicing. Among them, the isomers SM8α and SM8β are detected in the prostate and regulate the production of full-length proteins.Citation125 TRPM8 protein contains 1104 AA residues, which is the main conserved region of the TRP family. There are eight glycosylation sites and one immune antigen epitope. TRPM8 is expressed in a variety of cells, but mainly in the dorsal root and trigeminal ganglion of the peripheral nervous system.Citation126 In the cell, this channel shows the location of the plasma membrane and membrane raft. However, TRPM8 has also been found in the mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) in prostate cancer cells.Citation127 Thus, TRPM8 channels may have a role far beyond their role in somatosensory neurons. In addition, the protein is also found in other tissues, such as the bladder, vascular smooth muscle, the liver, and lungs.Citation89

7. Pathological functions of the TRPM channels in glioma

Most TRPM members are extensively involved in developing malignant tumors.Citation18 Among them, TRPM2, 3, 7, and 8 are closely related to the occurrence and development of glioma. TRPM2 and TRPM3 have antitumor effects, and TRPM7 and TRPM8 may be involved in the malignant transformation of glioma.Citation63 Therefore, this chapter will focus on the relevant pathological functions of TRPM2, 3, 7, and 8 in glioma.

7.1. TRPM2

Oxidative stress is a prominent feature of many pathologies, and a common mechanism is to induce cell death by disrupting intracellular ion homeostasis.Citation73 Recent evidence suggests that TRPM2 channels are susceptible to oxidative stress activation and play a vital part in oxidative stress-induced cell death, including gliomas.Citation128 For example, introducing TRPM2 into human glioblastoma multiforme (GBM) cells enhanced the hydrogen peroxide-induced apoptosis. Interestingly, TRPM2 has also been found to induce autophagy. In most cases, TRPM2, located on the lysosome membrane, causes cell death through autophagy. Moreover, studies have shown that TRPM2 may also induce autophagy by acting on the early steps of autophagy in cancer.Citation129 Apoptosis and autophagy are common forms of cytotoxicity induced by anticancer drugs. For example, the effect of selenium (Se) on docetaxel-resistant GBM cells may be to activate TRPM2 through oxidative stress and enhance the apoptotic effect of DTX.Citation19

In addition to inducing apoptosis and autophagy, TRPM2 may play a vital role in glioma progression since TRPM2 channels are also found in cerebral blood vessels.Citation130 Furthermore, abnormal TRPM2 function appears to be associated with psychiatric and neurological disorders, such as stress depression, epilepsy, ischemic brain damage, and glioma invasion.Citation131 Moreover, TRPM2-S and TRPM2-AS are also closely associated with malignant tumors such as gliomas.Citation132 TRPM2-S is an isomer of TRPM2, which activates the ERAD system to ubiquitinate TRPM2 and inhibit TRPM2 activity by interacting with TRPM2 located at the cytoplasmic membrane.Citation133 TRPM2-AS has recently been identified as an antisense lncRNA located at chromosome 21q22.3.Citation134 It is an oncogene for prostate cancer, hepatocellular carcinoma, and glioma.Citation135,Citation136 In gliomas, overexpression of TRPM2-AS is closely related to cell proliferation, invasion, and migration. The same phenomenon was found when TRPM2-AS was down-regulated by siRNA.Citation132 In conclusion, TRPM2, TRPM2-S, and TRPM2-AS are closely related to the occurrence and development of glioma.

7.2. TRPM3

TRPM3 is widely distributed in the body, especially in the brain, and the dysregulation of its expression is closely related to glioma. Still, there are few reports on the pathological function of TRPM3 in glioma. For example, TRPM3 expression was upregulated in glioma. TRPM3 may play a vital role as a tumor suppressor gene in glioma. Its loss of expression is a marker of poor prognosis in GBM patients because TRPM3 expression is significantly down-regulated with increased glioma grade.Citation63 Furthermore, DNA methylation of the TRPM3 promoter was positively correlated with glioma dryness and migration.Citation137 Moreover, TRPM3 has also been closely related to glioma growth and autophagy,Citation138 but its mechanism needs further research. In conclusion, methylation of the TRPM3 is involved in the malignant phenotype of glioma.

7.3. TRPM7

TRPM7 is widely expressed throughout the body and is closely associated with many central nervous system lesions, including glioma.Citation121,Citation139 For example, one study has found that TRPM7 channels are closely related to the angiogenesis of glioma.Citation140 Another study demonstrated TRPM7-dependent CLIC1 vesicles translocated to microvascular endothelial cells in glioma. It suggested that TRPM7 channels may promote GBM angiogenesis.Citation141 As mentioned above, TRPM7 is a bifunctional protein containing an ion channel and kinase structure.Citation142 Although TRPM7 mediates cell migration, invasion, and proliferation in glioma, migration and invasion ability are mainly increased by α-kinase domains, while proliferative capacity is increased by ion channel activity.Citation24,Citation61 These lines of evidence suggest that TRPM7 channels and kinase domains are closely related to glioma proliferation, migration, and invasion.

7.4. TRPM8

Various diseases, such as chronic cough, cancer, migraines, and irritable bowel syndrome, have been linked to TRPM8 pathology. Early studies found that in more than 20 TRP channels, compared with normal brain tissue, TRPM8 mRNA expression in GBM was upregulated to the highest level, which was closely related to the migration, invasion, and proliferation of glioma.Citation143 TRPM8 suppression or knockdown has been shown to disrupt the trigger of apoptotic cell death, cell cycle, cloning survival, and impair DNA repair.Citation144 TRPM8 channels may also regulate cell proliferation by dynamically controlling the level of glioma resting potential, which may be a crucial cell cycle regulator. Furthermore, one study found two TRPM8 variants, with a more significant menthol-induced increase in Ca2+ influx observed in migrating cells than in non-migrating cells, possibly indicating that only migrating cells express full-length TRPM8 proteins in the plasma membrane.Citation63 Nevertheless, these results must be treated with caution in future cancer treatment strategies, as TRPM8 has been found to have anti-migration activity against other cancers, such as prostate cancer.Citation145 Interestingly, ionizing radiation can also activate BK channels to induce migration. The mechanism proved that ionizing radiation activates and upregulates TRPM8 in glioma,Citation63 thus confirming the direct and indirect interaction in controlling glioma migration. Moreover, the increased invasion rate of glioma is also related to TRPM8 overexpression. In addition, HGF/SF is a multifunctional cell effector that can affect TRPM8-induced cell migration and Ca2+ homeostasis enhancement, meaning that TRPM8 is invasive and resistant. The evidence also indicates that TRPM8 may be enriched in HGF/SF and cMET, which is vital in malignant tumors, including GBM.Citation146 Furthermore, TRPM8’s effect on GBM progression was far greater than its effect on cell migration and invasion. It also dramatically affects other decisive processes such as cell survival, cell cycle, and radiation resistance.Citation144 In summary, the above evidence proves that the pathological function of TRPM8 is closely related to the migration, invasion, and proliferation of glioma.

8. Signaling pathways of the TRPM channels in glioma

Four members of the TRPM channels, TRPM2, 3, 7, and 8, are widely involved in the signal transduction process of glioma, such as cell-death-related signaling pathways, RTK/RAS/PI3K signaling pathways, MAPK/ERK signaling pathways, JAK2/STAT3/Notch signaling pathway, miR-28-5p/Rap1b axis, CaMKII, BK, and Kir4.1-K+ channels, etc. These signaling pathways are reviewed in detail below, as shown in ).

Figure 3. The main TRPMs-mediated mechanisms and signaling pathways associated with the development and progression of glioma.

Figure 3. The main TRPMs-mediated mechanisms and signaling pathways associated with the development and progression of glioma.

8.1. TRPM2-mediated cell death-related signaling pathways in glioma

Tumor cell death cannot be separated from various signaling pathways. TRPM2 is closely associated with cell death-related signaling pathways in glioma, especially apoptosis and autophagy.Citation20 TRPM2-mediated apoptosis was induced mainly through the mitochondrial pathway. TRPM2 activation leads to increased intracellular Ca2+ concentration and oxidative stress, which promotes glioma cell death through apoptosis.Citation63 Specifically, Oxidative stress is closely related to the increase of ROS. ROS increases NAD+/ADPR levels or induces oxidative stress, triggering TRPM2 activation. Subsequently, TRPM2 induces MPTP opening and mitochondrial damage through excess Ca2+ and Na+, leading to caspase-3-dependent apoptosis, apoptotic body formation, and nuclear condensation. Interestingly, mitochondrial fission may be one of the pathways through which mitochondria trigger TRPM2-mediated apoptosis. Previous studies have suggested that TRPM2 may initiate pancreatic cell mitochondrial apoptosis by mediating the lysosome Zn2+ layer.Citation73 Nevertheless, the source of elevated Zn2+ needs to be clarified in the study, and we speculate that this mechanism is also related to glioma cell apoptosis.

Protein kinase C-α can induce the decoupling of spliceosome TRPM2-S and TRPM2 and activate cell apoptosis. For example, TRPM2-S induces apoptosis in liposarcoma by increasing ROS levels.Citation82 So far, there have been no studies on glioma. In addition to inducing apoptosis, TRPM2 can induce autophagy through a mechanism that has been shown to activate a range of Ca2±dependent receptors and signaling cascades by altering cytoplasmic Ca2+ concentrations. Such as CaMKK, MAPK and JNK/c-Jun/RGS4.Citation132 Moreover, lysosomal Ca2+ signaling was found to activate AMPK and trigger autophagy via calcineurin. In conclusion, these results suggest that TRPM2 mediates glioma cell death through apoptosis and autophagy signaling pathways related to cell death.

8.2. TRPM3 plays a protective role in glioma by targeting the miR-204

MicroRNAs (miRNAs) are endogenous, small non-coding RNAs that negatively regulate gene expression.Citation147 As one of them, miR-204 originates from the 6th intron of the TRPM3 gene,Citation148 while the TRPM3 promoter controls the expression of miR-204. miR-204 has been reported to act as a tumor suppressor gene in various malignancies,Citation149 including glioma. For example, miR-204 exerts tumor-suppressive effects by promoting apoptosis, conferring cancer cell resistance to chemotherapy, inhibiting epithelial-to-mesenchymal transformation (EMT), and self-renewal of cancer stem cells (CSCs).Citation150 Interestingly, studies have shown that miR-204 is substantially down-regulated due to hypermethylation of the TRPM3 promoter, which is related to enhanced cell stemness and migration.Citation137 In addition, in nude mouse brain transplant tumors, miR-204 restoration consistently inhibited tumor invasion and improved animal survival.Citation63 However, the role of TRPM3 and the regulatory function of miR-204 have been questioned. For example, upregulation of miR-204 expression in glioma leads to reduced ezrin levels through classical interaction with MRE (miRNA recognition element) in ezrin 3’ UTR.Citation151 In conclusion, TRPM3 is closely related to the function of miR-204 and plays a protective role in glioma.

8.3. Extensive TRPM7-related signaling pathways in glioma

8.3.1. TRPM7-related RTK/RAS/PI3K and MAPK/ERK signaling pathways in glioma

RTK/RAS/RAF/MEK/ERK signal cascade is essential in intercellular and intracellular communication and is a ubiquitous signal transduction pathway.Citation152 It is often altered in disease and causes many human syndromes and conditions, including cancer.Citation153 To date, RTK/RAS/PI3K and MAPK/ERK signaling pathways related to TRPM7 are closely associated with the migration, invasion, and proliferation of glioma, which are crucial for glioma progression. For example, one study found that growth factors such as EGF and VEGF regulate TRPM7 activity through the RTKS signaling pathway.Citation154 Another study confirmed that TRPM7 enhances the MAPK/ERK and/or PI3K/Akt pathways in various cancers, including glioma.Citation155,Citation156 Furthermore, MAPK/ERK and PI3K/Akt are essential signaling pathways that show key roles in glioma migration, invasion, and proliferation. Interestingly, PI3K/Akt signaling hyperactivation occurs in approximately 45% of GBM patients.Citation157 Inhibiting the PI3K/Akt signaling pathway can inhibit glioblastoma growth. Moreover, Naltriben has been shown to enhance GBM migration and invasion by inducing TRPM7-like currents through Ca2 + influx, associated with increased activation of MAPK/ERK-related proteins.Citation99 Interestingly, Naltriben also enhanced TRPM7 channel activity, possibly by upregulating the MAPK/ERK signaling pathway and MMP-2 protein expression to enhance GBM migration and invasion. Besides Naltriben, the anti-migration and anti-proliferation effects of xylitone B on glioma are also related to Ras/MEK/MAPK and PI3K/Akt signaling pathways regulated by TRPM7.Citation121

PI3K/Akt and MAPK/ERK signaling pathways are regulated by PLC.Citation121 Therefore, TRPM7-related PLC isoenzymes may interact with the TRPM7α-serine/threonine protein kinase domain. Further research is needed to confirm these hypotheses. Interestingly, patients did not respond satisfactorily to treatment that inhibited only one of them. These evidence suggests a possible interaction between the MAPK/ERK pathway and the PI3K/Akt pathway, and a more effective strategy for treating GBM would be to target TRPM7.Citation99

8.3.2. TRPM7-related Notch and/or JAK2/STAT3 signaling pathway, and miR-28-5p/Rap1b axis in glioma

As a traditional inflammatory pathway, the JAK/STAT signaling pathway has also been found to promote the occurrence and development of cancer.Citation158 It involves various tumorigenic functions, including replication, anti-apoptosis, angiogenesis, and immunosuppression.Citation159 Abnormal activation of this pathway has been found in tumors, including gliomas, which may contribute to tumor development.Citation160 TRPM7 can enhance malignant phenotype in glioma by upregulating Notch and JAK2/STAT3 signaling pathways.Citation140 Furthermore, STAT3 binds the aldehyde dehydrogenase 1 (ALDH1) promoter, which is functionally involved in cell differentiation, self-protection, proliferation, and expansion, which may imply that TRPM7 is not only involved in migration, invasion, and proliferation but also glioma stem cell (GSC) differentiation and renewal.Citation161 Moreover, the expression of TRPM7 was associated with the decrease of miR-28-5p. As previously mentioned, miR-28-5p also is a tumor suppressor whose downregulation leads to significantly increased proliferation and invasion of glioma.Citation61 In the target of miR-28-5p, the expression of Rap1b in GBM was positively correlated with TRPM7 and negatively correlated with miR-28-5p. In fact, TRPM7 may also modulate the Notch pathway, recently implicated in integrin-mediated cell adhesion and Rap1b signaling.Citation63

8.4. TRPM8 is involved in the MAPK and Ca2+/calmodulin-dependent protein kinase II (CaMKII) signaling pathways as well as BK and Kir4.1-K+channels of glioma

As mentioned before, MAPK signaling pathways related to TRPM8 are closely related to the malignant phenotype of glioma, which also shows a vital role in the cell death of glioma. For example, one study found that MAPK signaling is involved in TRPM8-mediated apoptosis of GBM cells,Citation162 and another study confirmed that inhibition of the MAPK signaling pathway leads to growth arrest and stimulates apoptosis of GBM cells.Citation163 Furthermore, TRPM8 interferes with cell cycle control via Cdc2, CaMKII, and cdc25C.Citation144 More specifically, TRPM8-mediated Ca2+ entry increases CaMKII activity by activating the BK channels and then inhibits the Cdc2 subunit of the mitotic promoter by inhibiting Cdc25C phosphatase phosphorylation. Interestingly, TRPM8 channels also regulate GBM cell migration by activating K+ membrane ion channels (BK channels) activated by large conductance Ca2+.Citation164 In addition, TRPM8 activation can also increase Kir4.1-K+ channels. Similar to the BK channels, Kir4.1 is a K+ channel with altered expression in the glioma.Citation165 Its expression was negatively related to glioma migration and invasion behavior.Citation166 In a word, TRPM8 is closely related to glioma signaling pathways, including the MAPK signaling pathway and CaMKII, BK, and Kir4.1-K+ channels.

9. TRPM channels drug application in glioma

Ion channels are widely expressed in almost all living cells and are the third largest drug target after enzymes and receptors. Considering the enormous contribution of altered expression of the TRPM channels to glioma formation and progression, they may be promising novel therapeutic targets. and show the structure of commonly used TRPM channel drugs.

Figure 4. Structure of chemical agonists and antagonists of the TRPM channels in glioma.

Figure 4. Structure of chemical agonists and antagonists of the TRPM channels in glioma.

Table 1. Chemical inhibitors and activators of the TRPM family in glioma.

Some data show that TRPM2 could be a good candidate for gene therapy. Combining TRPM2 with γ-radiation or chemotherapy drugs can improve the efficacy of GBM treatment. Moreover, TRPM2 promotes cancer cell death, including glioma.Citation73 Next, curcumin, BTX-A, DTX, paclitaxel and resveratrol will be reviewed one by one. Previously, many studies have confirmed that the cytotoxic effect of curcumin on glioma cells is closely related to the induction of cell apoptosis and autophagy.Citation168 The combination of curcumin and cisplatin suggested that curcumin enhanced cisplatin-induced death of human laryngeal squamous cell carcinoma (LSCC) cells through TRPM2.Citation169 Cisplatin is a taxane widely used in glioma, breast cancer, and prostate cancer chemotherapy. Therefore, curcumin can enhance the efficacy in the case of cisplatin resistance, and it is an excellent drug combination for the treatment of glioma. In glioblastoma, it has been confirmed that BTX-A shows a vital protective role in glioma cell proliferation by stimulating cell apoptosis. Its molecular mechanism may be the up-regulation of the mitochondrial DRIA-related oxidative stress pathway and the activation of TRPM2.Citation167 Furthermore, selenium (Se) tested in docetaxel (DTX) resistant GBM cells may enhance the apoptotic effects of DTX by activating TRPM2 through oxidative stress. More specifically, selenium activates TRPM2-mediated Ca2+ influx by stimulating the production of oxidative stress, thereby enhancing the same Ca2±dependent apoptotic pathway induced by DTX. Interestingly, the cytotoxic effects of DTX may also arise from Ca2+ influx into cells and the formation of excessive mitochondrial ROS, which leads to DNA damage and subsequent apoptosis by triggering the overactivation of PARP. In addition, the supplementation of selenium can alleviate the side effects of docetaxel.Citation19 Recent studies have demonstrated the benefits of combined treatment with paclitaxel and resveratrol. Resveratrol was shown to enhance the efficacy of paclitaxel, particularly against paclitaxel-resistant breast cancer cells.Citation170 When it comes to brain cancer, there are all kinds of findings. Ozturk et al. studied the combined effect of paclitaxel and resveratrol on GBM cells. The results showed that TRPM2 was involved in the mechanism by which resveratrol enhanced the effect of paclitaxel.Citation171 Paclitaxel and resveratrol further increased apoptosis by increasing intracellular ROS levels and inducing mitochondrial dysfunction, suggesting that TRPM2 plays a role in regulating apoptosis in GBM cells. In conclusion, targeting TRPM2 may be a promising therapeutic approach, and researchers should actively explore its characteristics and mechanism as a therapeutic target for glioma.

Studies have found that the central pore is essential in drug transport.Citation172 In the TRP family, TRPV2 and TRPM3-related drugs were found to be closely related to the central pore. For example, one study found that TRPV2 channels act as “drug carriers” in cancer treatment. TRPM3 promotes chemotherapeutic drugs’ absorption through the central pore, thereby improving the efficacy of cancer therapy.Citation173 A similar central pore is present in TRPM3, as mentioned earlier. Other evidence is that the TRPM channels can act as a “drug carrier” due to the penetration of chemotherapy agents into cells through the pore domain. For example, activating TRPM3 channels may allow intracellular uptake of nonspecific chemotherapy agents by altering pore helical conformation.Citation57 In this context, future studies should explore whether TRPM3 can promote chemotherapeutic drug absorption via the central pore pathway.

Tumor recurrence is attributed to migration, invasion, and resistance to therapy,Citation174 and pharmacological inhibition of TRPM7 may be a novel therapy in glioma. It was also found that some TRPM7 channels, which interfere with cell division pathways, may be promising targets for drugs, making tumors more amenable to resection and local radiotherapy. For example, carvacrol affects glioma migration, invasion, and proliferation by inhibiting TRPM7 and regulating the G0-G1 cell cycle.Citation175

Increasing evidence suggests that increased activation of TRPM7-mediated MAPK/ERK and PI3K/AKT signaling pathways are critical components of tumorigenesis and other signature cancer features. Like xyloketal B, carvacrol has been reported to induce xykeob-like effects in glioma by inhibiting MAPK/ERK and PI3K/AKT signaling pathways.Citation121 In parallel experiments, it was found that carvacrol could reduce expression levels of p-Akt and p-ERK1/2, the cell viability, migration, and proliferation in GBM cells. Interestingly, Vacquinol-1 (VQ-1)-induced multitype glioblastoma cell death was in contrast to exogenous ATP-induced TRPM7 activity.Citation176 The study of TRPM7 and its specific pharmacological inhibition confirmed that inhibition of AKT or ERK inhibitors is crucial for controlling GBM growth and invasion. Moreover, Siddiqui et al. used NS8593 to demonstrate that TRPM7 inhibits the migration and invasion properties of microglial cells.Citation177 TRPM7 is a negative regulator of the angiogenic process.Citation61 It suggests that specific TRPM7 activators can target both vascularization and cancer progression. Future studies should consider specific activators of TRPM7, such as Mibefradil, to be valuable for further investigation.

The low specificity of some TRPM8 agonists may have higher efficacy by triggering multiple channels simultaneously. For example, activating TRPM8 channels with menthol and icilin has increased the opening probability of individual BK channels.Citation63 This channel mediates ion flux through the plasma membrane and supports cell contractile-driven cell migration.Citation178 Furthermore, the overexpression of BK channels was found in glioma. Pharmacological inhibition of the channels was confirmed to eliminate cell migration and menthol-stimulated intracellular Ca2+ influx.Citation179 Furthermore, ionizing radiation, which induces migration through Ca2+ -mediated activation of BK channels, has been shown to activate and upregulate TRPM8-mediated Ca2+ influx in glioma cells,Citation144 thus confirming the direct interplay of the two channels in controlling glioma migration. Moreover, another study showed that TRPM8 agonists such as menthol could increase Kir4.1-mediated cell membrane conductivity in U251 GBM cells.Citation180 Therefore, all the above evidence proves that the TRPM8 channels are a promising candidate for targeted glioma therapy.

10. Conclusions and Perspectives

With the deepening of the understanding of TRPM channels, it has been found that TRPM channels are widely involved in the physiological and pathological processes of the human body, such as temperature regulation, immune response, insulin secretion, and cold or osmotic regulation. In addition, the TRPM channels are closely related to the occurrence and development of tumors. There is increasing evidence that TRPM channels may be promising targets for cancer therapy. Studies of different TRPM members have advanced the understanding of the complexity of the TRPM channels. Since most TRPM structures are closed, much remains to be learned about the “control” of channel activation.

Here, we conclude that TRPM2, TRPM3, TRPM7, and TRPM8 isoforms are promising targets in glioma, with multiple pieces of evidence reporting their overexpression in cancer and demonstrating the requirements in cancer cell growth, survival, migration, and invasion. As described in the main text, TRPM3 and TRPV2-related drugs are closely associated with the central pore. However, whether TRPM3-related drugs can also promote the absorption of chemotherapeutic drugs through the intermediate pore, future studies should focus on these aspects.

With the increasing understanding of TRPM channel-mediated signaling pathways in tumors, many therapeutic targets involved in glioma cell proliferation, self-renewal, invasion, and angiogenesis have been identified. As a result, many novel treatments have been reported in the past decade. Few of these significantly affect glioma, even though anti-angiogenic therapy may provide some benefits for GBM patients, such as a possible delay in recurrence of about six months. Therefore, there is a need to develop other molecular targets that can bind to anti-angiogenic, cytotoxic, immunotherapy, and DNA repair inhibitors. In this case, targeting the activity of cytokines or components in the tumor microenvironment is also promising.

TRPM channels are overexpressed in tumors, including gliomas. Tumor microenvironments include a variety of cell types. Current studies are limited to tumor cells, so the cell types expressing TRPM channels in tumor microenvironments need to be further explored. TRPM channels are not universally expressed and may play a pleiotropic role. For example, TRPM channels are involved in vascular endothelial growth factor (VEGF) signaling, which influences angiogenesis and tumor growth. TRPM channels have also been demonstrated to interact with other channel families, such as the BK channels, which are directly involved in the signaling pathway of brain tumor progression. Furthermore, some researchers strongly support the use of TRPMs in antiangiogenic therapy. Since TRPM2 and TRPM7 are also found in cerebral blood vessels, it is suggested that they may promote GBM angiogenesis. Moreover, TRPM8 isoforms and TRPM2-AS may be more limited in nonmalignant human organs and thus may represent more specific targets. Certain studies did not measure intracellular ion concentrations or changes in ion fluxes that modulate the activity of specific TRPM channels, nor did they measure changes in specific TRPM currents. Therefore, further experiments are needed to discover the specific mechanisms.

Finally, in the last 3–4 years, significant discoveries have been made in the structure resolution of TRPM2, TRPM7, and TRPM8 by cryogenic EM. Due to the advances of cryo-EM methods in resolving complex receptor or ion channel structures embedded in plasma membranes, it is conceivable that we will gradually discover the structure of other TRPM channels. These data will aid in designing drugs with precise action and specific inhibitory activity, thereby expanding the potential of targeting TRPM pathways. TRPM channels can destroy human cancer cells, as shown by various evidence of their carcinogenic properties. In addition, future in vivo and in vitro evaluations and human clinical trials are needed to fully understand the role of TRPM ion channels in tumors such as gliomas. In short, the pharmaceutical industry and medical practice should consider them as new targets for therapeutic approaches.

Abbreviations

CNS=

central nervous system

TRP=

transient receptor potential

TRPM=

transient receptor potential melastatin

cADPR=

cyclic ADPR

NAD=

nicotinamide adenine dinucleotide

NAAD=

nicotinate adenine dinucleotide

NAADP=

NAAD-phosphate

RNS=

reactive nitrogen

MAPKs=

mitogen-activated protein kinases

PKC=

protein kinase C

PLC=

phospholipase C

ER=

endoplasmic reticulum

GBM=

glioblastoma multiforme

Se=

selenium

miRNAs=

MicroRNAs

CSCs=

cancer stem cells

MRE=

miRNA recognition element

EMT=

epithelial-to-mesenchymal transformation

PS=

pregnenolone sulfate

ALDH1=

aldehyde dehydrogenase 1

GSC=

glioma stem cell

CaMKII=

Ca2+/calmodulin-dependent protein kinase II

LSCC=

laryngeal squamous cell carcinoma

DTX=

docetaxel

VQ-1=

Vacquinol-1

VEGF=

vascular endothelial growth factor

PIP2=

phosphatidylinositol 4,5-bisphosphate

ADPR=

ADP-ribose

CAM=

calmodulin

TNFα=

tumor necrosis factor-α

RNS=

reactive nitrogen

PKC=

protein kinase C

Author contributions

Z.C., H.X. and J.L. wrote the main manuscript text, E.F., J.Z., R.H., Y.X. and H.W. prepared figures. E.B. and D.S. conceived and designed the study. All authors reviewed the manuscript.

Ethical approval

This is a review, and there is no ethical conflict.

Acknowledgments

We thank all participants of this study.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

This research was funded by Talent Acquisition Program of Yijishan Hospital (KY29070694), the National Natural Science Foundation of China (No. 81972348), College Excellent Youth Talent Support Program in Anhui Province (No. gxypZD2019019), Key Projects of Natural Science Research in Anhui Province (KJ2019A0267).

References

  • Lee A, Fakler B, Kaczmarek LK, Isom LL. More than a pore: ion channel signaling complexes. J Neurosci. 2014;34(46):15159–16. doi:10.1523/JNEUROSCI.3275-14.2014.
  • Jimenez I, Prado Y, Marchant F, Otero C, Eltit F, Cabello-Verrugio C, Cerda O, Simon F. TRPM channels in human diseases. Cells-Basel. 2020;9(12):2604. doi:10.3390/cells9122604.
  • Litan A, Langhans SA. Cancer as a channelopathy: ion channels and pumps in tumor development and progression. Front Cell Neurosci. 2015;9:86. doi:10.3389/fncel.2015.00086.
  • Qian LL, Sun X, Yang J, Wang X-L, Ackerman MJ, Wang R-X, Xu X, Lee H-C, Lu T. Changes in ion channel expression and function associated with cardiac arrhythmogenic remodeling by sorbs2. Biochim Biophys Acta Mol Basis Dis. 2021;1867(12):166247. doi:10.1016/j.bbadis.2021.166247.
  • Brierley S. Altered ion channel/receptor expression and function in extrinsic sensory neurons: the cause of and solution to chronic visceral pain? Adv Exp Med Biol. 2016;891:75–90. doi:10.1007/978-3-319-27592-5_9.
  • Sakaguchi R, Mori Y. Transient receptor potential (trp) channels: biosensors for redox environmental stimuli and cellular status. Free Radic Biol Med. 2020;146:36–44. doi:10.1016/j.freeradbiomed.2019.10.415.
  • Zhang Z, Toth B, Szollosi A, Chen J, Csanády L. Structure of a TRPM2 channel in complex with Ca(2+) explains unique gating regulation. Elife. 2018;7. doi:10.7554/eLife.36409.
  • Chen Y, Zhang X, Yang T, Bi R, Huang Z, Ding H, Li J, Zhang J. Emerging structural biology of trpm subfamily channels. Cell Calcium. 2019;79:75–9. doi:10.1016/j.ceca.2019.02.011.
  • Lopez-Romero AE, Hernandez-Araiza I, Torres-Quiroz F, Tovar-Y-Romo LB, Islas LD, Rosenbaum T. Trp ion channels: proteins with conformational flexibility. Channels (Austin). 2019;13(1):207–26. doi:10.1080/19336950.2019.1626793.
  • Huang Y, Fliegert R, Guse AH, Lü W, Du J. A structural overview of the ion channels of the trpm family. Cell Calcium. 2020;85:102111. doi:10.1016/j.ceca.2019.102111.
  • Held K, Voets T, Vriens J. TRPM3 in temperature sensing and beyond. Temp (Austin). 2015;2(2):201–13. doi:10.4161/23328940.2014.988524.
  • Liu Z, Wu H, Wei Z, Wang X, Shen P, Wang S, Wang A, Chen W, Lu Y. Trpm8: a potential target for cancer treatment. J Cancer Res Clin Oncol. 2016;142(9):1871–81. doi:10.1007/s00432-015-2112-1.
  • Zholos A, Johnson C, Burdyga T, Melanaphy D. TRPM channels in the vasculature. Adv Exp Med Biol. 2011;704:707–29. 10.1007/978-94-007-0265-3_37
  • Belrose JC, Jackson MF. TRPM2: a candidate therapeutic target for treating neurological diseases. Acta Pharmacol Sin. 2018;39(5):722–32. doi:10.1038/aps.2018.31.
  • Zielinska W, Zabrzynski J, Gagat M, Grzanka A. The role of trpm2 in endothelial function and dysfunction. Int J Mol Sci. 2021;22(14):7635. doi:10.3390/ijms22147635.
  • Schneider FM, Mohr F, Behrendt M, Oberwinkler J. Properties and functions of trpm1 channels in the dendritic tips of retinal on-bipolar cells. European Journal of Cell Biology. 2015;94(7–9):420–7. doi:10.1016/j.ejcb.2015.06.005.
  • Simon F, Varela D, Cabello-Verrugio C. Oxidative stress-modulated trpm ion channels in cell dysfunction and pathological conditions in humans. Cell Signal. 2013;25(7):1614–24. doi:10.1016/j.cellsig.2013.03.023.
  • Wong KK, Banham AH, Yaacob NS, Nur Husna SM. The oncogenic roles of TRPM ion channels in cancer. J Cell Physiol. 2019;234(9):14556–73. doi:10.1002/jcp.28168.
  • Santoni G, Maggi F, Morelli MB, Santoni M, Marinelli O. Transient receptor potential cation channels in cancer therapy. Med Sci (Basel). 2019;7(12):108. doi:10.3390/medsci7120108.
  • Hantute-Ghesquier A, Haustrate A, Prevarskaya N, Lehen’kyi V. Trpm family channels in cancer. Pharmaceuticals (Basel). 2018;11(2):11. doi:10.3390/ph11020058.
  • Shahmir S, Zahmatkesh N, Mirzaahmadi S, Asaadi Tehrani G. LncRNA CASC2 inhibits progression of glioblastoma by regulating the expression of AKT in T98G cell line, treated by TMZ and thiosemicarbazone complex. Asian Pac J Cancer Prev. 2023;24(5):1553–60. doi:10.31557/APJCP.2023.24.5.1553.
  • Persano F, Gigli G, Leporatti S. Natural compounds as promising adjuvant agents in the treatment of gliomas. Int J Mol Sci. 2022;23(6):3360. doi:10.3390/ijms23063360.
  • Kim BJ. Involvement of transient receptor potential melastatin 7 channels in sophorae radix-induced apoptosis in cancer cells: sophorae radix and trpm7. J Pharmacopuncture. 2012;15(3):31–8. doi:10.3831/KPI.2012.15.003.
  • Leng TD, Li MH, Shen JF, Liu M-L, Li X-B, Sun H-W, Branigan D, Zeng Z, Si H-F, Li J, et al. Suppression of trpm7 inhibits proliferation, migration, and invasion of malignant human glioma cells. CNS Neurosci Ther. 2015; 21(3):252–261. doi:10.1111/cns.12354.
  • Elias AF, Lin BC, Piggott BJ. Ion channels in gliomas—from molecular basis to treatment. Int J Mol Sci. 2023;24(3):2530. doi:10.3390/ijms24032530.
  • Lefranc F. Transient receptor potential (trp) ion channels involved in malignant glioma cell death and therapeutic perspectives. Front Cell Dev Biol. 2021;9:618961. doi:10.3389/fcell.2021.618961.
  • Chen WL, Barszczyk A, Turlova E, Deurloo M, Liu B, Yang BB, Rutka JT, Feng Z-P, Sun H-S. Inhibition of trpm7 by carvacrol suppresses glioblastoma cell proliferation, migration and invasion. Oncotarget. 2015;6(18):16321–40. doi:10.18632/oncotarget.3872.
  • So JS, Kim H, Han KS. Mechanisms of invasion in glioblastoma: extracellular matrix, Ca(2+) signaling, and glutamate. Front Cell Neurosci. 2021;15:663092. doi:10.3389/fncel.2021.663092.
  • Samanta A, Hughes T, Moiseenkova-Bell VY. Transient receptor potential (trp) channels. Subcell Biochem. 2018;87:141–65. doi:10.1007/978-981-10-7757-9_6.
  • Kaneko Y, Szallasi A. Transient receptor potential (TRP) channels: a clinical perspective. Br J Pharmacol. 2014;171(10):2474–507. doi:10.1111/bph.12414.
  • Katz B, Payne R, Minke B. TRP channels in vision. 2017:27–63. 10.4324/9781315152837-3.
  • Fleisch VC, Jametti T, Neuhauss SC. Electroretinogram (ERG) measurements in larval zebrafish. CSH Protoc. 2008;2008(3):t4973. doi:10.1101/pdb.prot4973.
  • Venkatraman P, Mills-Henry I, Padmanabhan KR, Pascuzzi P, Hassan M, Zhang J, Zhang X, Ma P, Pang CP, Dowling JE, et al. Rods contribute to visual behavior in larval zebrafish. Invest Ophthalmol Vis Sci. 2020; 61(12):11. doi:10.1167/iovs.61.12.11.
  • Montell C, Rubin GM. Molecular characterization of the drosophila trp locus: a putative integral membrane protein required for phototransduction. Neuron. 1989;2(4):1313–23. doi:10.1016/0896-6273(89)90069-x.
  • Minke B. The history of the drosophila trp channel: the birth of a new channel superfamily. J Neurogenet. 2010;24(4):216–33. doi:10.3109/01677063.2010.514369.
  • Wes PD, Chevesich J, Jeromin A, Rosenberg C, Stetten G, Montell C. Trpc1, a human homolog of a drosophila store-operated channel. Proc Natl Acad Sci USA. 1995;92(21):9652–6. doi:10.1073/pnas.92.21.9652.
  • Chen S, Chiu CN, McArthur KL, Fetcho JR, Prober DA. Trp channel mediated neuronal activation and ablation in freely behaving zebrafish. Nat Methods. 2016;13(2):147–50. doi:10.1038/nmeth.3691.
  • Bernardini M, Fiorio PA, Prevarskaya N, Gkika D. Human transient receptor potential (trp) channel expression profiling in carcinogenesis. Int J Dev Biol. 2015;59(7–8–9):399–406. doi:10.1387/ijdb.150232dg.
  • Desai BN, Clapham DE. Trp channels and mice deficient in trp channels. Pflugers Arch Eur J Physiol. 2005;451(1):11–18. doi:10.1007/s00424-005-1429-z.
  • Venkatachalam K, Luo J, Montell C. Evolutionarily conserved, multitasking trp channels: lessons from worms and flies. Handb Exp Pharmacol. 2014;223:937–62. doi:10.1007/978-3-319-05161-1_9.
  • Pedersen SF, Owsianik G, Nilius B. Trp channels: an overview. Cell Calcium. 2005;38(3–4):233–52. doi:10.1016/j.ceca.2005.06.028.
  • Clapham DE, Runnels LW, Strubing C. The trp ion channel family. Nat Rev Neurosci. 2001;2(6):387–96. doi:10.1038/35077544.
  • Liao M, Cao E, Julius D, Cheng Y. Structure of the TRPV1 ion channel determined by electron cryo-microscopy. Nature. 2013;504(7478):107–12. doi:10.1038/nature12822.
  • Miller BA. TRPC2. Handb Exp Pharmacol. 2014;222:53–65. doi:10.1007/978-3-642-54215-2_3.
  • Dietrich A. Transient receptor potential (trp) channels in health and disease. Cells-Basel. 2019;8. doi:10.3390/cells8050413.
  • Haustrate A, Prevarskaya N, Lehen’kyi V. Role of the trpv channels in the endoplasmic reticulum calcium homeostasis. Cells-Basel. 2020;9(2):317. doi:10.3390/cells9020317.
  • Smani T, Shapovalov G, Skryma R, Prevarskaya N, Rosado JA. Functional and physiopathological implications of trp channels. Biochimica et Biophysica Acta (BBA) – Mol Cell Res. 2015;1853(8):1772–82. doi:10.1016/j.bbamcr.2015.04.016.
  • Takahashi N, Mori Y. Trp channels as sensors and signal integrators of redox status changes. Front Pharmacol. 2011;2:58. doi:10.3389/fphar.2011.00058.
  • Theile JW, Cummins TR. Recent developments regarding voltage-gated sodium channel blockers for the treatment of inherited and acquired neuropathic pain syndromes. Front Pharmacol. 2011;2:54. doi:10.3389/fphar.2011.00054.
  • Choi JH, Jeong SY, Oh MR, Allen PD, Lee EH. Trpcs: influential mediators in skeletal muscle. Cells-Basel. 2020;9(4):850. doi:10.3390/cells9040850.
  • Cao E. Structural mechanisms of transient receptor potential ion channels. J Gen Physiol. 2020;152(3):152. doi:10.1085/jgp.201811998.
  • Gwanyanya A, Mubagwa K. Emerging role of transient receptor potential (trp) ion channels in cardiac fibroblast pathophysiology. Front Physiol. 2022;13:968393. doi:10.3389/fphys.2022.968393.
  • Nagarkatti N, Deshpande LS, DeLorenzo RJ. Development of the calcium plateau following status epilepticus: role of calcium in epileptogenesis. Expert Rev Neurother. 2009;9:813–24. doi:10.1586/ern.09.21.
  • Nishimura H, Kawasaki M, Tsukamoto M, Menuki K, Suzuki H, Matsuura T, Baba K, Motojima Y, Fujitani T, Ohnishi H, et al. Transient receptor potential vanilloid 1 and 4 double knockout leads to increased bone mass in mice. Bone Rep. 2020;12:100268. doi:10.1016/j.bonr.2020.100268.
  • Marquez-Nogueras KM, Hortua TM, Chasen NM, Kuo IY, Moreno SN. Calcium signaling through a transient receptor channel is important for toxoplasma gondii growth. Elife. 2021;10. doi:10.7554/eLife.63417.
  • Young A, Machacek DW, Dhara SK, MacLeish PR, Benveniste M, Dodla MC, Sturkie CD, Stice SL. Ion channels and ionotropic receptors in human embryonic stem cell derived neural progenitors. Neuroscience. 2011;192:793–805. doi:10.1016/j.neuroscience.2011.04.039.
  • Caterina MJ, Pang Z. Trp channels in skin biology and pathophysiology. Pharmaceuticals (Basel). 2016;9(4):77. doi:10.3390/ph9040077.
  • Peralvarez-Marin A, Donate-Macian P, Gaudet R. What do we know about the transient receptor potential vanilloid 2 (trpv2) ion channel? FEBS J. 2013;280(21):5471–87. doi:10.1111/febs.12302.
  • Nilius B, Owsianik G. The transient receptor potential family of ion channels. Genome Biol. 2011;12(3):218. doi:10.1186/gb-2011-12-3-218.
  • Xu X, Yu C, Xu L, Xu J. Emerging roles of keratinocytes in nociceptive transduction and regulation. Front Mol Neurosci. 2022;15:982202. doi:10.3389/fnmol.2022.982202.
  • Wan J, Guo AA, Chowdhury I, Guo S, Hibbert J, Wang G, Liu M. TRPM7 induces mechanistic target of rap1b through the downregulation of mir-28-5p in glioma proliferation and invasion. Front Oncol. 2019;9:1413. doi:10.3389/fonc.2019.01413.
  • Santoni G, Amantini C, Maggi F, Marinelli O, Santoni M, Nabissi M, Morelli MB. The trpv2 cation channels: from urothelial cancer invasiveness to glioblastoma multiforme interactome signature. Lab Invest. 2020;100(2):186–98. doi:10.1038/s41374-019-0333-7.
  • Chinigo G, Castel H, Chever O, Gkika D. Trp channels in brain tumors. Front Cell Dev Biol. 2021;9:617801. doi:10.3389/fcell.2021.617801.
  • Aroke EN, Powell-Roach KL, Jaime-Lara RB, Tesfaye M, Roy A, Jackson P, Joseph P. Taste the pain: the role of trp channels in pain and taste perception. Int J Mol Sci. 2020;21(16):5929. doi:10.3390/ijms21165929.
  • Ishimaru Y, Matsunami H. Transient receptor potential (trp) channels and taste sensation. J Dent Res. 2009;88(3):212–8. doi:10.1177/0022034508330212.
  • Shi R, Fu Y, Zhao D, Boczek T, Wang W, Guo F. Cell death modulation by transient receptor potential melastatin channels trpm2 and trpm7 and their underlying molecular mechanisms. Biochem Pharmacol. 2021;190:114664. doi:10.1016/j.bcp.2021.114664.
  • Castillo K, Diaz-Franulic I, Canan J, Gonzalez-Nilo F, Latorre R. Thermally activated trp channels: molecular sensors for temperature detection. Phys Biol. 2018;15(2):21001. doi:10.1088/1478-3975/aa9a6f.
  • Li T, Chen J, Zeng Z. Pathophysiological role of calcium channels and transporters in the multiple myeloma. Cell Commun Signal. 2021;19(1):99. doi:10.1186/s12964-021-00781-4.
  • Kunert-Keil C, Bisping F, Kruger J, Brinkmeier H. Tissue-specific expression of trp channel genes in the mouse and its variation in three different mouse strains. BMC Genomics. 2006;7(1):159. doi:10.1186/1471-2164-7-159.
  • Cohen MR, Moiseenkova-Bell VY. Structure of thermally activated trp channels. Curr Top Membr. 2014;74:181–211. doi:10.1016/B978-0-12-800181-3.00007-5.
  • Mei ZZ, Xia R, Beech DJ, Jiang L-H. Intracellular coiled-coil domain engaged in subunit interaction and assembly of melastatin-related transient receptor potential channel 2. J Biol Chem. 2006;281(50):38748–56. doi:10.1074/jbc.M607591200.
  • Guo J, She J, Zeng W, Chen Q, Bai X-C, Jiang Y. Structures of the calcium-activated, non-selective cation channel trpm4. Nature. 2017;552(7684):205–9. doi:10.1038/nature24997.
  • Malko P, Jiang LH. Trpm2 channel-mediated cell death: an important mechanism linking oxidative stress-inducing pathological factors to associated pathological conditions. Redox Biol. 2020;37:101755. doi:10.1016/j.redox.2020.101755.
  • Combe CL, Upchurch CM, Canavier CC, Gasparini S. Cholinergic modulation shifts the response of ca1 pyramidal cells to depolarizing ramps via trpm4 channels with potential implications for place field firing. Elife. 2023;12. doi:10.7554/eLife.84387.
  • Guo H, Carlson JA, Slominski A. Role of trpm in melanocytes and melanoma. Exp Dermatol. 2012;21(9):650–4. doi:10.1111/j.1600-0625.2012.01565.x.
  • Canales J, Morales D, Blanco C, Rivas J, Cerda O. A TR(I)P to cell migration: new roles of trp channels in mechanotransduction and cancer. Front Physiol. 2019;10:757. doi:10.3389/fphys.2019.00757.
  • Bouron A, Kiselyov K, Oberwinkler J. Permeation, regulation and control of expression of trp channels by trace metal ions. Pflugers Arch Eur J Physiol. 2015;467(6):1143–164. doi:10.1007/s00424-014-1590-3.
  • Turlova E, Feng ZP, Sun HS. The role of trpm2 channels in neurons, glial cells and the blood-brain barrier in cerebral ischemia and hypoxia. Acta Pharmacol Sin. 2018;39(5):713–21. doi:10.1038/aps.2017.194.
  • Yu P, Cai X, Liang Y, Wang M, Yang W. Roles of NAD(+) and its metabolites regulated calcium channels in cancer. Molecules. 2020;25(20):4826. doi:10.3390/molecules25204826.
  • Rosenbaum T. Activators of trpm2: getting it right. J Gen Physiol. 2015;145(6):485–7. doi:10.1085/jgp.201511405.
  • Pires PW, Earley S. Redox regulation of transient receptor potential channels in the endothelium. Microcirculation. 2017;24(3). doi:10.1111/micc.12329.
  • Miller BA. Trpm2 in cancer. Cell Calcium. 2019;80:8–17. doi:10.1016/j.ceca.2019.03.002.
  • Syed MS, Wang L, Li D, Jiang L-H. Trpm2 channel-mediated ros-sensitive CA(2+) signaling mechanisms in immune cells. Front Immunol. 2015;6:407. doi:10.3389/fimmu.2015.00407.
  • Hirschler-Laszkiewicz I, Chen SJ, Bao L, Wang J, Zhang X-Q, Shanmughapriya S, Keefer K, Madesh M, Cheung JY, Miller BA, et al. The human ion channel trpm2 modulates neuroblastoma cell survival and mitochondrial function through PYK2, CREB, and MCU activation. Am J Physiol Cell Physiol. 2018; 315(4):C571–C586. doi:10.1152/ajpcell.00098.2018.
  • Tan CH, McNaughton PA. Trpm2 and warmth sensation. Pflugers Arch - Eur J Physiol. 2018;470(5):787–798. doi:10.1007/s00424-018-2139-7.
  • Starkus J, Beck A, Fleig A, Penner R. Regulation of trpm2 by extra- and intracellular calcium. J Gen Physiol. 2007;130(4):427–440. doi:10.1085/jgp.200709836.
  • Jiang LH, Gamper N, Beech DJ. Properties and therapeutic potential of transient receptor potential channels with putative roles in adversity: focus on TRPC5, TRPM2 and TRPA1. Curr Drug Targets. 2011;12(5):724–736. doi:10.2174/138945011795378568.
  • Hu X, Wu L, Liu X, Zhang Y, Xu M, Fang Q, Lu L, Niu J, Abd El-Aziz TM, Jiang L-H, et al. Deficiency of ros-activated trpm2 channel protects neurons from cerebral ischemia-reperfusion injury through upregulating autophagy. Oxid Med Cell Longev. 2021;2021:1–12. doi:10.1155/2021/7356266.
  • Ferrandiz-Huertas C, Mathivanan S, Wolf CJ, Devesa I, Ferrer-Montiel A. Trafficking of thermotrp channels. Membranes (Basel). 2014;4(3):525–564. doi:10.3390/membranes4030525.
  • Cabanas H, Muraki K, Staines D, Marshall-Gradisnik S. Naltrexone restores impaired transient receptor potential melastatin 3 ion channel function in natural killer cells from myalgic encephalomyelitis/chronic fatigue syndrome patients. Front Immunol. 2019;10:2545. doi:10.3389/fimmu.2019.02545.
  • Becker A, Gotz C, Montenarh M, Philipp SE. Control of TRPM3 Ion Channels by Protein Kinase CK2-Mediated Phosphorylation in Pancreatic β-Cells of the Line INS-1. Int J Mol Sci. 2021;22(23):22. doi:10.3390/ijms222313133.
  • Liu Y, Lyu Y, Wang H. Trp channels as molecular targets to relieve endocrine-related diseases. Front Mol Biosci. 2022;9:895814. doi:10.3389/fmolb.2022.895814.
  • Nilius B, Vennekens R. From cardiac cation channels to the molecular dissection of the transient receptor potential channel trpm4. Pflugers Arch Eur J Physiol. 2006;453(3):313–321. doi:10.1007/s00424-006-0088-z.
  • Liman ER. Trpm5 and taste transduction. Handb Exp Pharmacol. 2007:287–298. doi:10.1007/978-3-540-34891-7_17.
  • Kurland DB, Gerzanich V, Karimy JK, Woo SK, Vennekens R, Freichel M, Nilius B, Bryan J, Simard JM. The SUR1-TRPM4 channel regulates nos2 transcription in tlr4-activated microglia. J Neuroinflammation. 2016;13(1):130. doi:10.1186/s12974-016-0599-2.
  • Chubanov V, Mittermeier L, Gudermann T. Role of kinase-coupled trp channels in mineral homeostasis. Pharmacol Ther. 2018;184:159–176. doi:10.1016/j.pharmthera.2017.11.003.
  • Ryazanova LV, Hu Z, Suzuki S, Chubanov V, Fleig A, Ryazanov AG. Elucidating the role of the trpm7 alpha-kinase: trpm7 kinase inactivation leads to magnesium deprivation resistance phenotype in mice. Sci Rep. 2014;4(1):7599. doi:10.1038/srep07599.
  • Demeuse P, Penner R, Fleig A. Trpm7 channel is regulated by magnesium nucleotides via its kinase domain. J Gen Physiol. 2006;127(4):421–434. doi:10.1085/jgp.200509410.
  • Wong R, Turlova E, Feng ZP, Rutka JT, Sun H-S. Activation of trpm7 by naltriben enhances migration and invasion of glioblastoma cells. Oncotarget. 2017;8(7):11239–11248. doi:10.18632/oncotarget.14496.
  • Andersson KE. Trp channels as lower urinary tract sensory targets. Med Sci (Basel). 2019;7(5):67. doi:10.3390/medsci7050067.
  • Yin Y, Lee SY. Current view of ligand and lipid recognition by the menthol receptor trpm8. Trends Biochem Sci. 2020;45(9):806–819. doi:10.1016/j.tibs.2020.05.008.
  • Yin Y, Le SC, Hsu AL, Borgnia MJ, Yang H, Lee S-Y. Structural basis of cooling agent and lipid sensing by the cold-activated trpm8 channel. Science. 2019;363(6430). doi:10.1126/science.aav9334.
  • Duvoisin RM, Haley TL, Ren G, Strycharska-Orczyk I, Bonaparte JP, Morgans CW. Autoantibodies in melanoma-associated retinopathy recognize an epitope conserved between trpm1 and trpm3. Invest Ophthalmol Vis Sci. 2017;58(5):2732–2738. doi:10.1167/iovs.17-21443.
  • Maddodi N, Setaluri V. Prognostic significance of melanoma differentiation and trans-differentiation. Cancers Basel. 2010;2(2):989–999. doi:10.3390/cancers2020989.
  • Shiels A. TRPM3_MIR-204: a complex locus for eye development and disease. Hum Genomics. 2020;14(1):7. doi:10.1186/s40246-020-00258-4.
  • Sita G, Hrelia P, Graziosi A, Ravegnini G, Morroni F. Trpm2 in the brain: role in health and disease. Cells-Basel. 2018;7(7):82. doi:10.3390/cells7070082.
  • Xie YF, Belrose JC, Lei G, Tymianski M, Mori Y, MacDonald JF, Jackson MF. Dependence of NMDA/GSK-3β mediated metaplasticity on TRPM2 channels at Hippocampal CA3-CA1 synapses. Mol Brain. 2011;4(1):44. doi:10.1186/1756-6606-4-44.
  • Du J, Xie J, Yue L. Modulation of trpm2 by acidic PH and the underlying mechanisms for PH sensitivity. J Gen Physiol. 2009;134(6):471–488. doi:10.1085/jgp.200910254.
  • McQueen CM, Whitfield-Cargile CM, Konganti K, Blodgett GP, Dindot SV, Cohen ND. Trpm2 SNP genotype previously associated with susceptibility to rhodococcus equi pneumonia in quarter horse foals displays differential gene expression identified using RNA-seq. BMC Genomics. 2016;17(1):993. doi:10.1186/s12864-016-3345-3.
  • Kondratskyi A, Yassine M, Kondratska K, Skryma R, Slomianny C, Prevarskaya N. Calcium-permeable ion channels in control of autophagy and cancer. Front Physiol. 2013;4:272. doi:10.3389/fphys.2013.00272.
  • Brown RL, Xiong WH, Peters JH, Tekmen-Clark M, Strycharska-Orczyk I, Reed BT, Morgans CW, Duvoisin RM. Trpm3 expression in mouse retina. PLoS One. 2015;10(2):e117615. doi:10.1371/journal.pone.0117615.
  • Wagner TF, Drews A, Loch S, Mohr F, Philipp SE, Lambert S, Oberwinkler J. Trpm3 channels provide a regulated influx pathway for zinc in pancreatic beta cells. Pflugers Arch. 2010;460(4):755–765. doi:10.1007/s00424-010-0838-9.
  • Lee N, Chen J, Sun L, Wu S, Gray KR, Rich A, Huang M, Lin J-H, Feder JN, Janovitz EB, et al. Expression and characterization of human transient receptor potential melastatin 3 (htrpm3). J Biol Chem. 2003; 278(23):20890–20897. doi:10.1074/jbc.M211232200.
  • Vriens J, Voets T. Sensing the heat with trpm3. Pflugers Arch Eur J Physiol. 2018;470(5):799–807. doi:10.1007/s00424-017-2100-1.
  • Choudhary S, Kashyap SS, Martin RJ, Robertson AP. Advances in our understanding of nematode ion channels as potential anthelmintic targets. Int J Parasitol Drugs Drug Resist. 2022;18:52–86. doi:10.1016/j.ijpddr.2021.12.001.
  • Gonzales AL, Earley S. Regulation of cerebral artery smooth muscle membrane potential by Ca 2+-activated cation channels. Microcirculation. 2013;20(4):337–347. doi:10.1111/micc.12023.
  • Zhang H, Zhang X, Wang X, Sun H, Hou C, Yu Y, Wang S, Yin F, Yang Z. Comprehensive analysis of TRP channel–related genes in patients with triple-negative breast cancer for guiding prognostic prediction. Front Oncol. 2022;12:941283. doi:10.3389/fonc.2022.941283.
  • Kaske S, Krasteva G, Konig P, Kummer W, Hofmann T, Gudermann T, Chubanov V. Trpm5, a taste-signaling transient receptor potential ion-channel, is a ubiquitous signaling component in chemosensory cells. BMC Neurosci. 2007;8(1):49. doi:10.1186/1471-2202-8-49.
  • Dhakal S, Lee Y. Transient receptor potential channels and metabolism. Mol Cells. 2019;42(8):569–578. doi:10.14348/molcells.2019.0007.
  • Gaudet R. Divide and conquer: high resolution structural information on trp channel fragments. J Gen Physiol. 2009;133(3):231–7. doi:10.1085/jgp.200810137.
  • Chen WL, Turlova E, Sun CL, Kim J-S, Huang S, Zhong X, Guan Y-Y, Wang G-L, Rutka J, Feng Z-P, et al. Xyloketal b suppresses glioblastoma cell proliferation and migration in vitro through inhibiting trpm7-regulated pi3k/akt and mek/erk signaling pathways. Mar Drugs. 2015; 13(4):2505–25. doi:10.3390/md13042505.
  • Yee NS, Kazi AA, Yee RK. Cellular and developmental biology of trpm7 channel-kinase: implicated roles in cancer. Cells-Basel. 2014;3(3):751–77. doi:10.3390/cells3030751.
  • Ledeganck KJ, Boulet GA, Bogers JJ, Verpooten GA, De Winter BY. The trpm6/egf pathway is downregulated in a rat model of cisplatin nephrotoxicity. PLoS One. 2013;8(2):e57016. doi:10.1371/journal.pone.0057016.
  • Bidaux G, Borowiec AS, Dubois C, Delcourt P, Schulz C, Abeele FV, Lepage G, Desruelles E, Bokhobza A, Dewailly E, et al. Targeting of short trpm8 isoforms induces 4TM-TRPM8-dependent apoptosis in prostate cancer cells. Oncotarget. 2016; 7(20):29063–80. doi:10.18632/oncotarget.8666.
  • Peng M, Wang Z, Yang Z, Tao L, Liu Q, Yi Lu, Wang X. Overexpression of short TRPM8 variant α promotes cell migration and invasion, and decreases starvation-induced apoptosis in prostate cancer LNCaP cells. Oncol Lett. 2015;10(3):1378–84. doi:10.3892/ol.2015.3373.
  • Iraci N, Ostacolo C, Medina-Peris A, Ciaglia T, Novoselov AM, Altieri A, Cabañero D, Fernandez-Carvajal A, Campiglia P, Gomez-Monterrey I, et al. In vitro and in vivo pharmacological characterization of a novel trpm8 inhibitor chemotype identified by small-scale preclinical screening. Int J Mol Sci. 2022; 23(4):2070. doi:10.3390/ijms23042070.
  • Yudin Y, Rohacs T. Regulation of trpm8 channel activity. Mol Cell Endocrinol. 2012;353(1–2):68–74. doi:10.1016/j.mce.2011.10.023.
  • Ishii M, Oyama A, Hagiwara T, Miyazaki A, Mori Y, Kiuchi Y, Shimizu S. Facilitation of H2O2-INDUCED A172 human glioblastoma cell death by insertion of oxidative stress-sensitive trpm2 channels. Anticancer Res. 2007;27(6B):3987–92.
  • Zhao Q, Li J, Ko WH, Kwan Y-W, Jiang L, Sun L, Yao X. Trpm2 promotes autophagic degradation in vascular smooth muscle cells. Sci Rep. 2020;10(1):20719. doi:10.1038/s41598-020-77620-y.
  • Park L, Wang G, Moore J, Girouard H, Zhou P, Anrather J, Iadecola C. The key role of transient receptor potential melastatin-2 channels in amyloid-β-induced neurovascular dysfunction. Nat Commun. 2014;5(1):5318. doi:10.1038/ncomms6318.
  • Mahmuda NA, Yokoyama S, Munesue T, Hayashi K, Yagi K, Tsuji C, Higashida H. One single nucleotide polymorphism of the trpm2 channel gene identified as a risk factor in bipolar disorder associates with autism spectrum disorder in a Japanese population. Diseases. 2020;8(1):4. doi:10.3390/diseases8010004.
  • Bao MH, Lv QL, Szeto V, Wong R, Zhu S-Z, Zhang Y-Y, Feng Z-P, Sun H-S. Trpm2-as inhibits the growth, migration, and invasion of gliomas through JNK, C-JUN, and RGS4. J Cell Physiol. 2020;235(5):4594–604. doi:10.1002/jcp.29336.
  • Yamamoto S, Ishii T, Mikami R, Numata T, Shimizu S. Short trpm2 prevents the targeting of full-length trpm2 to the surface transmembrane by hijacking to ER associated degradation. Biochem Biophys Res Commun. 2019;520(3):520–5. doi:10.1016/j.bbrc.2019.10.065.
  • Cui D, Feng Y, Shi K, Zhang H, Qian R. Long non-coding RNA trpm2-as sponges microRNA-138-5p to activate epidermal growth factor receptor and PI3K/AKT signaling in non-small cell lung cancer. Ann Transl Med. 2020;8(20):1313. doi:10.21037/atm-20-6331.
  • Shi T, Li R, Duan P, Guan Y, Zhang D, Ding Z, Ruan X. Trpm2-as promotes paclitaxel resistance in prostate cancer by regulating FOXK1 via sponging mir-497-5p. Drug Dev Res. 2022;83(4):967–78. doi:10.1002/ddr.21924.
  • Xu C, Huang Q, Zhang C, Xu W, Xu G, Zhao X, Liu X, Du Y. Long non-coding RNA trpm2-as as a potential biomarker for hepatocellular carcinoma. Ir J Med Sci. 2018;187(3):621–8. doi:10.1007/s11845-017-1692-y.
  • Guo J, Shan C, Xu J, Li M, Zhao J, Cheng W. New insights into trp ion channels in stem cells. Int J Mol Sci. 2022;23(14):7766. doi:10.3390/ijms23147766.
  • Fallah HP, Ahuja E, Lin H, Qi J, He Q, Gao S, An H, Zhang J, Xie Y, Liang D, et al. A review on the role of trp channels and their potential as drug targets_an insight into the trp channel drug discovery methodologies. Front Pharmacol. 2022;13:914499. doi:10.3389/fphar.2022.914499.
  • Chen X, Numata T, Li M, Mori Y, Orser BA, Jackson MF, Xiong Z-G, MacDonald JF. The modulation of trpm7 currents by nafamostat mesilate depends directly upon extracellular concentrations of divalent cations. Mol Brain. 2010;3(1):38. doi:10.1186/1756-6606-3-38.
  • Liu M, Inoue K, Leng T, Guo S, Xiong Z-G. Trpm7 channels regulate glioma stem cell through STAT3 and Notch signaling pathways. Cell Signalling. 2014;26(12):2773–81. doi:10.1016/j.cellsig.2014.08.020.
  • Thuringer D, Chanteloup G, Winckler P, Garrido C. The vesicular transfer of CLIC1 from glioblastoma to microvascular endothelial cells requires trpm7. Oncotarget. 2018;9(70):33302–11. doi:10.18632/oncotarget.26048.
  • Zeitlmayr S, Zierler S, Staab-Weijnitz CA, Dietrich A, Geiger F, Horgen FD, Gudermann T, Breit A. TRPM7 restrains plasmin activity and promotes transforming growth factor-β1 signaling in primary human lung fibroblasts. Arch Toxicol. 2022;96(10):2767–83. doi:10.1007/s00204-022-03342-x.
  • Zalles M, Smith N, Saunders D, Lerner M, Fung K-M, Battiste J, Towner RA. A tale of two multi-focal therapies for glioblastoma: an antibody targeting ELTD1 and nitrone-based okn-007. J Cell Mol Med. 2022;26(2):570–82. doi:10.1111/jcmm.17133.
  • Klumpp D, Frank SC, Klumpp L, Sezgin EC, Eckert M, Edalat L, Bastmeyer M, Zips D, Ruth P, Huber SM, et al. Trpm8 is required for survival and radioresistance of glioblastoma cells. Oncotarget. 2017; 8(56):95896–913. doi:10.18632/oncotarget.21436.
  • Grolez GP, Gordiendko DV, Clarisse M, Hammadi M, Desruelles E, Fromont G, Prevarskaya N, Slomianny C, Gkika D. Trpm8-androgen receptor association within lipid rafts promotes prostate cancer cell migration. Cell Death Dis. 2019;10(9):652. doi:10.1038/s41419-019-1891-8.
  • Laterra J, Rosen E, Nam M, Ranganathan S, Fielding K, Johnston P. Scatter factor/hepatocyte growth factor expression enhances human glioblastoma tumorigenicity and growth. Biochem Biophys Res Commun. 1997;235(3):743–7. doi:10.1006/bbrc.1997.6853.
  • Wang X, Omar O, Vazirisani F, Thomsen P, Ekström K. Mesenchymal stem cell-derived exosomes have altered microRNA profiles and induce osteogenic differentiation depending on the stage of differentiation. PLoS One. 2018;13(2):e193059. doi:10.1371/journal.pone.0193059.
  • Li T, Pan H, Li R. The dual regulatory role of mir-204 in cancer. Tumour Biol. 2016;37(9):11667–77. doi:10.1007/s13277-016-5144-5.
  • Son JC, Jeong HO, Park D, No SG, Lee EK, Lee J, Chung HY. mir-10a and mir-204 as a potential prognostic indicator in low-grade gliomas. Cancer Inform. 2017;16:1885518114. doi:10.1177/1176935117702878.
  • Fan X, Zou X, Liu C, Cheng W, Zhang S, Geng X, Zhu W. microRNA expression profile in serum reveals novel diagnostic biomarkers for endometrial cancer. Biosci Rep. 2021;41(6):41. doi:10.1042/BSR20210111.
  • Xin J, Zheng LM, Sun DK, Li X, Xu P, Tian L. miR‑204 functions as a tumor suppressor gene, at least partly by suppressing CYP27A1 in glioblastoma. Oncol Lett. 2018;16:1439–48. doi:10.3892/ol.2018.8846.
  • Li L, Zhao GD, Shi Z, Qi L-L, Zhou L-Y, Fu Z-X. The RAS/RAF/MEK/ERK signaling pathway and its role in the occurrence and development of HCC. Oncol Lett. 2016;12(5):3045–3050. doi:10.3892/ol.2016.5110.
  • Wang SH, Wu HC, Badrealam KF, Kuo Y-H, Chao Y-P, Hsu H-H, Bau D-T, Viswanadha VP, Chen Y-H, Lio P-J, et al. Taiwanin E induces cell cycle arrest and apoptosis in Arecoline/4-NQO-induced oral cancer cells through modulation of the ERK signaling pathway. Front Oncol. 2019;9:1309. doi:10.3389/fonc.2019.01309.
  • Zou ZG, Rios FJ, Montezano AC, Touyz RM. Trpm7, magnesium, and signaling. Int J Mol Sci. 2019;20(8):1877. doi:10.3390/ijms20081877.
  • Lee EH, Chun SY, Kim B, Yoon BH, Lee JN, Kim BS, Yoo ES, Lee S, Song PH, Kwon TG, et al. Knockdown of trpm7 prevents tumor growth, migration, and invasion through the Src, Akt, and Jnk pathway in bladder cancer. BMC Urol. 2020; 20(1):145. doi:10.1186/s12894-020-00714-2.
  • Takayasu T, Kurisu K, Esquenazi Y, Ballester LY. Ion channels and their role in the pathophysiology of gliomas. Mol Cancer Ther. 2020;19(10):1959–1969. doi:10.1158/1535-7163.MCT-19-0929.
  • William D, Mokri P, Lamp N, Linnebacher M, Classen CF, Erbersdobler A, Schneider B. Amplification of the egfr gene can be maintained and modulated by variation of egf concentrations in in vitro models of glioblastoma multiforme. PLoS One. 2017;12(9):e185208. doi:10.1371/journal.pone.0185208.
  • Zhang Q, Shen J, Wu Y, Ruan W, Zhu F, Duan S. LINC00520: a potential diagnostic and prognostic biomarker in cancer. Front Immunol. 2022;13:845418. doi:10.3389/fimmu.2022.845418.
  • Zhang Y, Zhuang K, Yuan S, Si W, Li Y, Zhang J, Liu J. Effects of mir-195 targeted regulation of JAK2 on proliferation, invasion, and apoptosis of gastric cancer cells. Comput Math Methods Med. 2022;2022:1–8. doi:10.1155/2022/5873479.
  • Swiatek-Machado K, Kaminska B. Stat signaling in glioma cells. Adv Exp Med Biol. 2020;1202:203–222. doi:10.1007/978-3-030-30651-9_10.
  • Ciaglia T, Vestuto V, Bertamino A, González-Muñiz R, Gómez-Monterrey I. On the modulation of trpm channels: current perspectives and anticancer therapeutic implications. Front Oncol. 2022;12:1065935. doi:10.3389/fonc.2022.1065935.
  • Su Z, Han S, Jin Q, Zhou N, Lu J, Shangguan F, Yu S, Liu Y, Wang L, Lu J, et al. Ciclopirox and bortezomib synergistically inhibits glioblastoma multiforme growth via simultaneously enhancing JNK/p38 MAPK and NF-κB signaling. Cell Death Dis. 2021; 12(3):251. doi:10.1038/s41419-021-03535-9.
  • Yuan Z, Yang Z, Li W, Wu A, Su Z, Jiang B, Ganesan S. Triphlorethol-a attenuates u251 human glioma cancer cell proliferation and ameliorates apoptosis through Jak2/Stat3 and P38 Mapk/Erk signaling pathways. J Biochem Mol Toxicol. 2022;36(9):e23138. doi:10.1002/jbt.23138.
  • Maklad A, Sharma A, Azimi I. Calcium signaling in brain cancers: roles and therapeutic targeting. Cancers Basel. 2019;11(2):11. doi:10.3390/cancers11020145.
  • Higashimori H, Sontheimer H. Role of kir4.1 channels in growth control of glia. Glia. 2007;55(16):1668–1679. doi:10.1002/glia.20574.
  • Seker-Polat F, Pinarbasi DN, Solaroglu I, Bagci-Onder T. Tumor cell infiltration into the brain in glioblastoma: from mechanisms to clinical perspectives. Cancers Basel. 2022;14(2):14. doi:10.3390/cancers14020443.
  • Akpinar O, Ozsimsek A, Guzel M, Nazıroğlu M. Clostridium botulinum neurotoxin a induces apoptosis and mitochondrial oxidative stress via activation of trpm2 channel signaling pathway in neuroblastoma and glioblastoma tumor cells. J Recept Signal Transduct Res. 2020;40(6):620–632. doi:10.1080/10799893.2020.1781174.
  • Lee JE, Yoon SS, Moon EY. Curcumin-induced autophagy augments its antitumor effect against a172 human glioblastoma cells. Biomol & Therapeutics. 2019;27(5):484–491. doi:10.4062/biomolther.2019.107.
  • Gokce KS, Gokce G, Kutuk M, Gürses Cila HE, Nazıroğlu M. Curcumin enhances cisplatin-induced human laryngeal squamous cancer cell death through activation of trpm2 channel and mitochondrial oxidative stress. Sci Rep. 2019;9(1):17784. doi:10.1038/s41598-019-54284-x.
  • Sprouse AA, Herbert BS. Resveratrol augments paclitaxel treatment in MDA-MB-231 and paclitaxel-resistant Mda-Mb-231 breast cancer cells. Anticancer Res. 2014;34(10):5363–5374.
  • Ozturk Y, Gunaydin C, Yalcin F, Nazıroğlu M, Braidy N. Resveratrol enhances apoptotic and oxidant effects of paclitaxel through trpm2 channel activation in dbtrg glioblastoma cells. Oxid Med Cell Longevity. 2019;2019:1–13. doi:10.1155/2019/4619865.
  • Li T, Wu K, Yue Z, Wang Y, Zhang F, Shen H. Structural basis for the modulation of human KCNQ4 by small-molecule drugs. Mol Cell. 2021;81(1):25–37. doi:10.1016/j.molcel.2020.10.037.
  • Nabissi M, Morelli MB, Santoni M, Santoni G. Triggering of the trpv2 channel by cannabidiol sensitizes glioblastoma cells to cytotoxic chemotherapeutic agents. Carcinogenesis. 2013;34(1):48–57. doi:10.1093/carcin/bgs328.
  • Nikolova B, Antov G, Semkova S, Tsoneva I, Christova N, Nacheva L, Kardaleva P, Angelova S, Stoineva I, Ivanova J, et al. Bacterial natural disaccharide (Trehalose Tetraester): molecular modeling and in vitro study of anticancer activity on breast cancer cells. Polym (Basel). 2020; 12(2):12. doi:10.3390/polym12020499.
  • Chen J, Dou Y, Zheng X, Leng T, Lu X, Ouyang Y, Sun H, Xing F, Mai J, Gu J, et al. Trpm7 channel inhibition mediates midazolam-induced proliferation loss in human malignant glioma. Tumour Biol. 2016; 37(11):14721–14731. doi:10.1007/s13277-016-5317-2.
  • Sander P, Mostafa H, Soboh A, Schneider JM, Pala A, Baron A-K, Moepps B, Wirtz CR, Georgieff M, Schneider M, et al. Vacquinol-1 inducible cell death in glioblastoma multiforme is counter regulated by trpm7 activity induced by exogenous ATP. Oncotarget. 2017; 8(21):35124–35137. doi:10.18632/oncotarget.16703.
  • Voringer S, Schreyer L, Nadolni W, Meier MA, Woerther K, Mittermeier C, Ferioli S, Singer S, Holzer K, Zierler S, et al. Inhibition of TRPM7 blocks MRTF/SRF-dependent transcriptional and tumorigenic activity. Oncogene. 2020; 39(11):2328–2344. doi:10.1038/s41388-019-1140-8.
  • McFerrin MB, Sontheimer H. A role for ion channels in glioma cell invasion. Neuron Glia Biol. 2006;2(1):39–49. doi:10.1017/S17440925X06000044.
  • Ransom CB, Sontheimer H. BK channels in human glioma cells. J Neurophysiol. 2001;85(2):790–803. doi:10.1152/jn.2001.85.2.790.
  • Ratto D, Ferrari B, Roda E, Brandalise F, Siciliani S, De Luca F, Priori EC, Di Iorio C, Cobelli F, Veneroni P, et al. Squaring the circle: a new study of inward and outward-rectifying potassium currents in U251 GBM cells. Cell Mol Neurobiol. 2020; 40(5):813–828. doi:10.1007/s10571-019-00776-3.