Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3637 2023-10-30 11:04:25 |
2 update references and layout Meta information modification 3637 2023-10-31 02:32:31 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Marini, M.; Titiz, M.; Souza Monteiro De Araújo, D.; Geppetti, P.; Nassini, R.; De Logu, F. TRP Channels in Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/50930 (accessed on 02 September 2024).
Marini M, Titiz M, Souza Monteiro De Araújo D, Geppetti P, Nassini R, De Logu F. TRP Channels in Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/50930. Accessed September 02, 2024.
Marini, Matilde, Mustafa Titiz, Daniel Souza Monteiro De Araújo, Pierangelo Geppetti, Romina Nassini, Francesco De Logu. "TRP Channels in Cancer" Encyclopedia, https://encyclopedia.pub/entry/50930 (accessed September 02, 2024).
Marini, M., Titiz, M., Souza Monteiro De Araújo, D., Geppetti, P., Nassini, R., & De Logu, F. (2023, October 30). TRP Channels in Cancer. In Encyclopedia. https://encyclopedia.pub/entry/50930
Marini, Matilde, et al. "TRP Channels in Cancer." Encyclopedia. Web. 30 October, 2023.
TRP Channels in Cancer
Edit

Ion channels play a crucial role in a wide range of biological processes, including cell cycle regulation and cancer progression. In particular, the transient receptor potential (TRP) family of channels has emerged as a promising therapeutic target due to its involvement in several stages of cancer development and dissemination.

ion channel transient receptor potential (TRP) channels calcium

1. Introduction

According to the World Health Organization (WHO), cancer comprises a large group of diseases characterized by the rapid growth of abnormal cells that invade neighboring parts of the body, with the capacity to spread to other organs. More than 100 types of cancer have been identified so far [1]. Cancer represents a leading cause of death worldwide, causing nearly 10 million deaths in 2020 [2]. The leading cause in the development of cancer is an abnormal proliferation of cancer cells, which, rather than responding appropriately to the signals that regulate normal cell behavior, grow and divide in an uncontrolled manner. However, because of the large variability in cancers, tissues, organs of origin, predisposing or causal agents, genetic influence, and differential response to pharmacological treatments, the identification of common causes, mechanisms, and treatments is practically impossible. Notwithstanding, an improved knowledge of specific transduction signaling pathways may offer novel possibilities for innovative targeted treatments.
Ion channels are integral membrane proteins containing an aqueous pore that facilitates the mobilization of certain ions between cell compartments playing an essential role in cell functioning [3]. They regulate different cellular pathways, including cell proliferation, migration, apoptosis, and differentiation, to maintain normal tissue homeostasis. During the phenotypic changes that lead from a normal epithelial towards a cancer cell, a series of genetic/epigenetic changes, among other functions, may affect the activity of the ion channels [4]. Ion transport across the cell membrane has a crucial role in fundamental tumor cell functions [5], such as cell migration and cycle progression [6], cell volume regulation, proliferation, and death [7][8], which play critical roles in tumor cell survival and metastasis [9].
Increasing awareness of the ion channel’s role in tumor progression has led to the consideration of cancer as a channelopathy, or as a disease characterized by a profound alteration in ion channel function [10]. A special family of ion channels, called mechano-gated ion channels, includes the prototypical mechanosensitive piezo channels, which respond to mechanical stimuli such as changes in membrane tension or force [11]. Another family of ion channels, the transient receptor potential (TRP) channels, are opened by physical (mechanical and thermal) and chemical stimuli [12]. The great sensitivity of mechanosensitive ion channels to modifications in matrix stiffness is another significant feature [13]. Mechanical signals that operate through mechanosensitive ion channels during tumor growth have an impact on the microenvironment as well as cancer cells [14]. It has been reported that there is an association between gliomas and piezo channels in the regulation of tissue stiffness and tumor mitosis. Piezo1 sustains focal adhesions and supports integrin focal adhesion kinase (FAK) signaling, tissue stiffening, and extracellular matrix control [15].
Calcium (Ca2+), potassium (K+), and sodium (Na+) channels are examples of channels involved in tumor growth and metastasis [16]. Ca2+ is an important second messenger whose intracellular levels control several downstream signaling pathways, such as apoptosis and cell migration, functions that typically affect cancer growth [17]. In some hormone-sensitive cancers, such as breast cancer, the presence of channels in metastatic cells is regulated by positive feedback mechanisms, induced by hormone action [18]. Therefore, those ion channels represent a promising target for tumor treatment [19].
TRP channels are ionic channels permeable to monovalent and divalent cations, with a conserved structure and a higher selectivity for K+, Na+, and Ca2+ [20]. They have a crucial role in several pathologies, including metabolic, cardiovascular, and cancer diseases [21][22]. Recently, in a study evaluating transcriptomic and genomic alterations in TRP genes across more than 10,000 patients, it was found that 27 of 28 TRP genes are correlated with at least one hallmark of cancer in 33 different tumor types [23]. Furthermore, antagonists and agonists of TRPs have been used in association with chemotherapy in many tumor models, although side effects due to a lack of tissue specificity were observed [24][25][26][27][28].
Until now, altered levels in the function of TRP proteins in cancer have been reported, rather than mutations in the TRP genes [29]. Depending on the stage of the cancer, decreased or increased levels of the expression of the normal TRP protein can be detected [30]. Thus, these proteins could represent important markers for predicting tumor progression and, consequently, potential therapeutical targets [31][32][33][34][35]. Changes in TRP channel expression have also been associated with the staging of tumor progression [12][36][37][38].

2. TRPs in Cancer

2.1. TRPA1 in Cancer

TRPA1, the only member of the ankyrin subfamily, is a polymodal channel that can be activated by a wide variety of noxious external stimuli, such as irritants, often associated with pain and inflammation, and intense cold [39][40][41][42]. TRPA1 can also be gated by several endogenously produced reactive chemical species, including oxidative stress by-products, such as ROS, reactive nitrogen (RNS), and carbonylic (RCS) species [41][43]. TRPA1 is predominantly expressed in the primary sensory neurons of the dorsal root (DRG), vagal (VG), and trigeminal (TG) ganglia, where it signals diverse pain stimuli [44][45][46][47][48]. TRPA1 expression has also been reported in non-neuronal cells, including the mouse inner ear and the organ of Corti [49], vascular endothelial cells [50], enterochromaffin cells of the respiratory tract [39][51], keratinocytes and melanocytes, synoviocytes, and dental pulp and gingival fibroblasts [52][53], as well as mast cells, epithelial, and pancreatic β cells [54][55][56][57][58][59][60][61]. More recently, the presence of TRPA1 in glial cells, such as astrocytes [62], oligodendrocytes [63], and Schwann cells [64][65], has been reported. In addition, the expression of TRPA1 has been observed in different cancer cells, including pancreatic adenocarcinoma and melanoma cells [66][67].
In cancer, TRPA1 activation in prostate tumor endothelial cells acts as a modulator of angiogenesis, since its activation promotes neovascularization, endothelial cell migration, and tubulogenesis in vitro in models of human prostate cancer [68]. TRPA1 activation in lung epithelial cancer cells (A549) can induce a decrease in cell invasion by inhibiting the cyclooxygenase-2 (COX-2)/prostaglandin E2 pathway in hypoxic cancer cells in vitro [69]. In breast and lung cancer spheroids, TRPA1 activates Ca2+-dependent antiapoptotic pathways by promoting ROS resistance [70]. Specifically, TRPA1 upregulated by nuclear factor erythroid 2-related factor 2 (NRF2), a transcription factor that, by encoding proteins with antioxidant and anti-inflammatory functions, promotes an adaptive process involving non-canonical oxidative stress defense and canonical ROS defense mechanisms [70]. Furthermore, TRPA1 inhibition attenuates xenograft tumor growth and increases chemosensitivity [70].
The TRPA1 protein has been identified in benign human skin lesions (dermal melanocytic nevi and dysplastic nevi), in cutaneous thin (pT1) and thick (pT4) melanomas, and in two different melanoma cell lines (SK-MEL-28 and WM266-4) [67]. In samples of skin lesion and melanoma, the presence of TRPA1 has been correlated with a progressive increase in oxidative stress and melanocytic transformation, as well as tumor severity. In vitro experiments on melanoma cell lines have shown that TRPA1 activation is associated with the release of H2O2, an observation in line with previous findings that have indicated the channel as an oxidative stress sensor and amplifier, a function that might affect tumor cells and proliferation [67].

2.2. TRPA1 in Cancer in Cancer Pain

Recent studies have underlined the role of TRPA1 in cancer pain. In a mouse model of cancer induced by the subcutaneous inoculation of melanoma B16-F10 cells, neuronal TRPA1 has been proposed to mediate mechanical and cold hypersensitivity and thigmotaxis behavior [71]. However, in mouse models of neuropathic pain induced by sciatic partial nerve ligation or ischemia and reperfusion, a prominent role of TRPA1 expressed in Schwann cells has been proposed [64][72]. Hematogenic macrophages recruited by increases in C-C Motif Chemokine Ligand 2 (CCL2) at sites of nerve injury generated a first burst of oxidative stress that, targeting the peripheral glial cell TRPA1, initiated a feed-forward mechanism that, via a Ca2+-dependent NADPH oxidase-1 (NOX1), amplified the oxidative stress to sustain pain signals [64][72]. Schwann cell TRPA1 has been similarly implicated in cancer-related pain by regulating, via a macrophage colony-stimulating factor (M-CSF), macrophage expansion and oxidative stress amplification, finally targeting neuronal TRPA1 to signal pain. In this mouse cancer model evoked by the inoculation of melanoma cells in the mouse paw, neuroinflammation and mechanical/cold hypersensitivity are maintained by a feed-forward mechanism, which requires continuous interaction between Schwann cell TRPA1 and expanded endoneurial macrophages throughout the entire sciatic nerve trunk [73].
Pain is also a recurrent symptom of cancer that becomes more frequent and debilitating in the presence of bone metastases, which are a common consequence of many primary tumors, including breast cancer [74]. A prominent role for the TRPA1 channel has also been reported in the development of mechanical hypersensitivity in a mouse model of metastatic bone cancer pain induced by the intramammary inoculation of breast carcinoma cells [75][76]. More recently, TRPA1 has been shown to be a regulator of metastatic bone cancer pain via insulin-like growth factor 1 receptor (IGF-1R) signaling in Schwann cells [77]. IGF-1, derived from osteoclast activation in osteolytic lesions caused by metastatic growth, targets its receptor expressed in Schwann cells, thus promoting an endothelial nitric oxide synthase-mediated TRPA1 activation and ROS release that, via M-CSF-mediated endoneurial macrophage expansion, sustains proalgesic responses.

2.3. TRPC

There are seven known members of the TRPC family, from TRPC1 to TRPC7. TRPC channels are widely expressed in many tissues and cells, including neurons, muscle cells, and epithelial cells. TRPC channels are involved in various physiological processes, such as sensory perception, smooth muscle contraction, hormone secretion, and cell migration [28]. TRPC channels can be activated by various signals, including G-protein-coupled receptors (GPCRs), receptor tyrosine kinases, and intracellular second messengers, such as diacylglycerol (DAG) and inositol trisphosphate (IP3) [78][79]. Upon activation, TRPC channels allow for the influx of Ca2+ ions into the cytoplasm, leading to an increase in the intracellular Ca2+ concentration. This Ca2+ influx triggers downstream signaling pathways and modulates the activity of various enzymes and transcription factors, ultimately influencing cell function. Aberrant TRPC channel activity has been associated with several diseases and pathological conditions, including cardiovascular disorders, neurodegenerative diseases, and cancer. Therefore, TRPC channels have emerged as potential therapeutic targets, and efforts are underway to develop drugs that modulate their activity for the treatment of these conditions.

2.3.1. TRPC1

TRPC1 is expressed in various types of cancer, including breast, pancreatic, lung, and glioblastoma multiforme. Its dysregulation has been proposed as a prognostic marker for some types of cancer [80], including breast cancer, as its expression is modulated by tumor development and metastasis [81][82][83]. An increased expression of TRPC1 is has been positively correlated with epithelial–mesenchymal transition (EMT), a complex process that induces tumor cells to spread and fight apoptosis, thus conferring a more aggressive phenotype [84][85]. Recently, it has been reported that TRPC1 overexpression increases markers for an EMT-like phenotype, such as zinc finger proteins, SNAI1 (SNAIL) and SNAI2 (SLUG), and VIMENTIN, by increasing invasiveness in mouse breast cancer cell lines in vitro [86]. TRPC1 is also expressed in human lung carcinoma, and high protein levels have been correlated with cancer differentiation and proliferation [87].
Aberrant Ca2+ signaling has been implicated in glioma pathogenesis and cell biology influencing cell proliferation, migration, invasion, and angiogenesis [88]. TRPC1 channels, acting as Ca2+-permeable channels, have been shown to regulate Ca2+ influx in glioma cells [89]. Increased Ca2+ influx through TRPC1 channels activates downstream signaling pathways, leading to cell proliferation and survival [89]. A loss of function of TRPC1, mediated by pharmacological or genetic inhibition, was found to reduce the proliferation of multinucleated glioma cells, mainly due to the suppression of store-operated Ca2+ entry (SOCE) [89].
TRPC1 channels have been found to promote glioma cell migration through their association with focal adhesion proteins and cytoskeletal rearrangement. Furthermore, TRPC1-mediated Ca2+ signaling can activate proteases and matrix metalloproteinases, facilitating the breakdown of the extracellular matrix and promoting glioma cell invasion [90]. TRPC1 channels have been shown to promote the release of pro-angiogenic factors, such as vascular endothelial growth factor (VEGF), from glioma cells, thus inducing endothelial cell proliferation and migration and leading to the formation of new blood vessels within the tumor microenvironment [91].

2.3.2. TRPC3

The TRPC3 protein might contribute to the development of tumor senescent phenotypes. Its downregulation in stromal cells promotes cellular senescence, sustaining inflammation and tumor growth in vivo [92]. TRPC3-mediated Ca2+ influx has been suggested as an endothelial cell attraction factor in prostate cancer, thus promoting angiogenesis [68][93]. TRPC3 overexpression has been detected in triple-negative breast cancer cells, and its activation induces an RAS P21 protein activator 4-mitogen-activated protein kinase (RASA4-MAPK) signaling cascade that plays a crucial functional role in preserving proliferation and resistance to apoptosis. A TRPC3 blocker attenuates proliferation, induces apoptosis, and sensitizes cell death to chemotherapeutic agents [94]. The TRPC3 channel is also highly expressed in gastric cancer specimens, and its expression is correlated with malignant progression by modulating the calcineurin B-like 2/glycogen synthase kinase-3 beta/nuclear factor of the activated T cells 2(CNB2/GSK3β/NFATc2) signaling pathway and controlling cell cycle, apoptosis, and intracellular ROS generation [95].

2.3.3. TRPC5

An aberrant Wnt/β-catenin signaling cascade facilitates cell renewal, proliferation, and differentiation in several cancer types [96]. The activation of this intracellular pathway increases the production of the ATP-biding cassette, subfamily B, member 1 (ABCB1), a multidrug efflux transporter that attenuates the effect of cytotoxic drugs in cancer cells. TRPC5 was found to be overexpressed together with ABCB1 in colorectal cancer cells resistant to 5-fluorouracil (5-Fu). TRPC5 silencing inhibits Wnt/β-catenin signaling, thus reducing ABCB1 and consequently reverting resistance to 5-Fu [97]. In a similar manner, TRPC5 channel expression is increased in breast cancer cell lines together with P-glycoprotein (P-gp), another pump overexpressed by cancer cells to remove cytotoxic drugs. TRPC5 suppression reduces P-gp levels and causes a reversal of drug resistance in cells [98]. The mechanism by which TRPC5 regulates P-gp seems to be specifically controlled through the activation of the nuclear factor of activated T cells isoform c3 (NFATc3) [99]. In addition, in breast cancer cells, TRPC5 activation promotes autophagy and chemoresistance via the Ca2+/calmodulin-dependent protein kinase beta/adenosine monophosphate-activated protein kinase alpha/mechanistic target of rapamycin (CaMKKβ/AMPKα/mTOR) pathway [100]. It has also been shown that TRPC5 is highly expressed in human breast cancer after long-term chemotherapy treatment, and its presence has been correlated with an increase in the transcription of vascular endothelial growth factor, which, in turn, stimulates tumor angiogenesis [101]. The TRPC5 channel seems to promote metastasis in colon cancer. Colon cancer patients with a high expression of TRPC5 display poorer overall and metastasis-free survival [102]. TRPC5 overexpression, by increasing intracellular Ca2+ concentration and mesenchymal biomarker expression, promotes cell migration, invasion, and proliferation [102].

2.3.4. TRPC6

Growing evidence has reported that the pattern of the expression of TRPC6 proteins is upregulated in several pathophysiological conditions, including cancer. TRPC6 has been found to be overexpressed in breast cancer biopsy tissues compared to normal breast tissues [103]. Human breast cancer cells in vitro also display significant levels of TRPC6 expression, and its silencing results in a significant reduction in cell growth [103]. In human hepatocellular carcinoma cells, transforming growth factor beta (TGFβ) is a mediator of motility, invasion, and metastases via the stimulation of Na+/Ca2+ exchanger 1 (NCX1), and TRPC6 activation regulates TGFβ, thus inducing the formation of a TRPC6/NCX1 molecular complex [104]. The expressions of both TRPC6 and NCX1 are markedly increased in human hepatocellular carcinoma tissues, and their expression levels positively correlate with migration, invasion, and intrahepatic metastasis [104]. An increased TRPC6 channel in cervical cancer cell lines induces cell proliferation, suggesting that channel inhibition might reduce the malignant behavior of the cancer. TRPC6 might be a new target for the prevention and treatment of cervical cancer [105].

2.4. TRPM

TRPM channels are a family of ion channels that regulate sensory perception, cellular homeostasis, and signal transduction. TRPM channels respond to a broad array of stimuli, including temperature, touch, pain, osmolarity, and chemical signals. They are expressed in various tissues and cell types throughout the body, highlighting their importance in numerous physiological functions. TRPM channels have gained significant attention in the field of cancer research due to their potential involvement in tumor progression and metastasis. Several members of the TRPM channel family have been implicated in cancer development and are investigated as potential therapeutic targets [106][107].

2.4.1. TRPM1

TRPM1 gene expression has been identified in benign nevi, dysplastic nevi, and cutaneous melanomas, with a negative association between its presence and melanoma aggressiveness [108][109]. In contrast, TRPM1 protein expression is associated with tumor progression and survival in acral melanoma, supposedly because of the activation of Ca2+/calmodulin-dependent protein kinase II (CaMKII), which facilitates the binding of CaMKII with protein kinase B (AKT) and activates AKT, promoting melanoma cell colony formation, mobility, and an increase in tumor growth [110].

2.4.2. TRPM2

TRPM2 is highly expressed in many human cancers (neuroblastoma, breast, gastric, lung, pancreatic, prostate cancer, squamous cell carcinoma, and T-cell leukemia), where its activation increases malignant cell survival [111]. The modulation of TRPM2 via oxidative stress in several pathological conditions has been reported [112]. One additional product derived from oxidative stress, ADP-ribose (ADPR), binds to the C- and N-termini of TRPM2, an action that results in channel activation [113][114][115].
Channel activation also modulates oxidative stress production. TRPM2 opening modulates the hypoxia-inducible transcription factor 1/2α (HIF-1/2α) signaling cascade, including proteins involved in oxidant stress, glycolysis, and mitochondrial function in neuroblastoma xenograft models [116]. TRPM2 inhibition or depletion reduces cell and mitochondria Ca2+ influx and decreases activity, autophagy, antioxidant response, and mitochondrial function, thus impairing tumor cell survival [116][117][118][119][120][121]. TRPM2 also has immunomodulatory functions and can influence the tumor microenvironment. TRPM2 activation in immune cells, such as macrophages and dendritic cells, affects their polarization and cytokine production, leading to a modulation of the anti-tumor immune response [122]. Additionally, TRPM2-mediated Ca2+ influx influences the release of inflammatory mediators that promote tumor growth and angiogenesis.

2.4.3. TRPM3

TRPM3 plays pleiotropic roles in cellular Ca2+ signaling and homeostasis [123]. TRPM3 has been identified in several cancer types in mammals, including kidney cell carcinoma, glioma, melanoma, and melanoma-associated retinopathy (MAR) [124][125][126]. The TRPM3 channel supports the growth of clear cell renal cell carcinoma by promoting autophagy [124]. An increased expression of TRPM3 in renal cell carcinoma leads to Ca2+ influx, which elicits the activation of CaMKII, 5′-AMP-activated protein kinase (AMPK), and Unc-51-like autophagy-activating kinase 1 (ULK1), as well as the formation of phagophore [127].
MiR-204 is an intron micro-RNA (miRNA) located between exons 7 and 8 of the TRPM3 gene. A reduction in miR-204 induced by the higher methylation of host gene TRPM3 in gliomas can promote cell migration and enhance cell stemness [128]. It has been shown that TRPM3 interacts with the signal transducer and activator of transcription 3 (STAT3) via the activation of STAT3-suppressing miR-204 expression. Furthermore, the downregulation of miR-204 via the methylation of the promoter of its host gene TRPM3 leads to the activation of the Src-STAT3-NFAT pathway, promoting glioma stem cell invasion and stem cell-like phenotype [129].

2.4.4. TRPM4

In physiological conditions, TRPM4 controls cell migration [130]. It regulates the activation of T lymphocyte and mast cells, together with the migration of dendritic and mast cells [131][132]. Under inflammatory conditions, TRPM4 is involved in vascular endothelial cell migration and ROS production [133]. TRPM4-mediated effects on cell migration are at least partially due to the activation of Rac family small GTPase 1 (Rac1-GTPase), a key regulator of cytoskeletal dynamics and cell polarity [134].
TRPM4 channel expression has been described in several cancers, including prostate [135][136][137], urinary bladder [138], cervical [139], colorectal [140][141], liver [142], and large B cell lymphoma [143]. In cancer cells, TRPM4 upregulation is associated with cancer cell migration, proliferation, and invasion. A recent study has shown that TRPM4 upregulation and its conductivity control the viability and cell cycle of colorectal cancer cells [144]. Another study revealed that TRPM4 gene defects mechanically engaged intestinal barrier integrity by depressing the generation of ROS and decreasing mucus production, thus promoting chronic bowel inflammation, a risk factor for colorectal cancer [140]. A more recent study indicated that an attenuated expression of TRPM4 is associated with the development of endometrial carcinoma and breast cancer through the hyperactivation of the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway, which regulates cell transcription, translation, migration, metabolism, proliferation, and survival [145]. In addition, within normal lymphoid tissues, including the tonsils, lymph nodes, and appendix, human normal B cells express low levels of TRPM4, while in diffuse large B cell lymphoma, a higher TRPM4 protein level has been detected, which confers significantly poorer patient outcomes [143]. Similarly, a higher level of TRPM4 protein correlates with a higher risk of recurrence following radical prostatectomy [137].

2.4.5. TRPM6

TRPM6 is mostly expressed in the kidneys, distal small intestine, and colon [146]. The TRPM6 channel is permeable to magnesium (Mg2+), thus assuming a relevant role in epithelial Mg2+ transport and active Mg2+ absorption, especially in the gut and kidneys [147]. Hypomagnesemia is evidenced in cancer patients after cisplatin-based chemotherapies [148], and it has emerged as the most notable adverse effect of the anti-epidermal growth factor receptor (EGFR) monoclonal antibody, cetuximab, which is used widely for the treatment of advanced colorectal cancer cells [149]. The downregulation of the TRPM6 channel is present in 80% of primary tumors in colorectal cancer cells, whereas its high expression increases patient survival [150].

2.4.6. TRPM8

TRPM8, also defined as a “cold receptor”, as it is activated by chemical cooling agents (such as menthol) [151], exhibits an increased expression in several cancer subtypes, including colon, breast, and prostate tumors, and it is considered to be a useful prognostic marker [152][153][154]. TRPM8 channels in cancer prognosis are associated with the modulation of cell viability, proliferation, migration, and apoptosis. For example, TRPM8 activation may regulate AMPK activity by modifying cellular autophagy to control the proliferation and migration of breast cancer cells. TRPM8 knockdown decreases basal autophagy, while TRPM8 overexpression increases basal autophagy in several mammalian cancer cell types. The activation of autophagy-associated signaling pathways for AMPK and unc-51-like kinase 1 (ULK1), as well as the production of phagophores, are part of the TRPM8 strategy for controlling autophagy [155].
TRPM8 involvement in the death and apoptosis of bladder cancer cells is due to the modulation of mitochondrial activity [156][157]. It has also been reported that, in glioblastoma, TRPM8 channels modulate the expression of apoptosis-related factors through the p38/MAPK pathway [158][159]. In colon, oral, esophageal, bladder, and breast cancers, TRPM8 seems directly involved in invasiveness and metastasis via the regulation of the epithelial–mesenchymal transition process (EMT) [153][160][161][162].

References

  1. Prevarskaya, N.; Skryma, R.; Shuba, Y. Ion Channels in Cancer: Are Cancer Hallmarks Oncochannelopathies? Physiol. Rev. 2018, 98, 559–621.
  2. World Health Organization. Cancer. Available online: https://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 7 June 2023).
  3. Kessler, D.; Gruen, G.C.; Heider, D.; Morgner, J.; Reis, H.; Schmid, K.W.; Jendrossek, V. The action of small GTPases Rab11 and Rab25 in vesicle trafficking during cell migration. Cell. Physiol. Biochem. 2012, 29, 647–656.
  4. Kunzelmann, K. Ion channels and cancer. J. Membr. Biol. 2005, 205, 159–173.
  5. Huber, S.M. Oncochannels. Cell Calcium 2013, 53, 241–255.
  6. Becchetti, A. Ion channels and transporters in cancer. 1. Ion channels and cell proliferation in cancer. Am. J. Physiol. Cell Physiol. 2011, 301, C255–C265.
  7. Lang, F.; Hoffmann, E.K. Role of ion transport in control of apoptotic cell death. Compr. Physiol. 2012, 2, 2037–2061.
  8. Turner, K.L.; Sontheimer, H. Cl− and K+ channels and their role in primary brain tumour biology. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130095.
  9. Djamgoz, M.B.; Onkal, R. Persistent current blockers of voltage-gated sodium channels: A clinical opportunity for controlling metastatic disease. Recent. Pat. Anticancer. Drug Discov. 2013, 8, 66–84.
  10. Litan, A.; Langhans, S.A. Cancer as a channelopathy: Ion channels and pumps in tumor development and progression. Front. Cell. Neurosci. 2015, 9, 86.
  11. Delmas, P.; Parpaite, T.; Coste, B. PIEZO channels and newcomers in the mammalian mechanosensitive ion channel family. Neuron 2022, 110, 2713–2727.
  12. Nilius, B.; Owsianik, G.; Voets, T.; Peters, J.A. Transient receptor potential cation channels in disease. Physiol. Rev. 2007, 87, 165–217.
  13. Kostic, A.; Lynch, C.D.; Sheetz, M.P. Differential matrix rigidity response in breast cancer cell lines correlates with the tissue tropism. PLoS ONE 2009, 4, e6361.
  14. Karska, J.; Kowalski, S.; Saczko, J.; Moisescu, M.G.; Kulbacka, J. Mechanosensitive Ion Channels and Their Role in Cancer Cells. Membranes 2023, 13, 167.
  15. Chen, X.; Wanggou, S.; Bodalia, A.; Zhu, M.; Dong, W.; Fan, J.J.; Yin, W.C.; Min, H.K.; Hu, M.; Draghici, D.; et al. A Feedforward Mechanism Mediated by Mechanosensitive Ion Channel PIEZO1 and Tissue Mechanics Promotes Glioma Aggression. Neuron 2018, 100, 799–815.e797.
  16. Lang, F.; Stournaras, C. Ion channels in cancer: Future perspectives and clinical potential. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130108.
  17. Canales, J.; Morales, D.; Blanco, C.; Rivas, J.; Díaz, N.; Angelopoulos, I.; Cerda, O. A TR(i)P to Cell Migration: New Roles of TRP Channels in Mechanotransduction and Cancer. Front. Physiol. 2019, 10, 757.
  18. Fraser, S.P.; Ozerlat-Gunduz, I.; Brackenbury, W.J.; Fitzgerald, E.M.; Campbell, T.M.; Coombes, R.C.; Djamgoz, M.B. Regulation of voltage-gated sodium channel expression in cancer: Hormones, growth factors and auto-regulation. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130105.
  19. Oosterwijk, E.; Gillies, R.J. Targeting ion transport in cancer. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130107.
  20. Nilius, B.; Owsianik, G. The transient receptor potential family of ion channels. Genome Biol. 2011, 12, 218.
  21. Saldías, M.P.; Maureira, D.; Orellana-Serradell, O.; Silva, I.; Lavanderos, B.; Cruz, P.; Torres, C.; Cáceres, M.; Cerda, O. TRP Channels Interactome as a Novel Therapeutic Target in Breast Cancer. Front. Oncol. 2021, 11, 621614.
  22. Smani, T.; Shapovalov, G.; Skryma, R.; Prevarskaya, N.; Rosado, J.A. Functional and physiopathological implications of TRP channels. Biochim. Biophys. Acta 2015, 1853, 1772–1782.
  23. Pan, T.; Gao, Y.; Xu, G.; Zhou, P.; Li, S.; Guo, J.; Zou, H.; Xu, Q.; Huang, X.; Xu, J.; et al. Pan-cancer analyses reveal the genetic and pharmacogenomic landscape of transient receptor potential channels. NPJ Genom. Med. 2022, 7, 32.
  24. Santoni, G.; Maggi, F.; Morelli, M.B.; Santoni, M.; Marinelli, O. Transient Receptor Potential Cation Channels in Cancer Therapy. Med. Sci. 2019, 7, 108.
  25. Santoni, G.; Farfariello, V. TRP channels and cancer: New targets for diagnosis and chemotherapy. Endocr. Metab. Immune Disord. Drug Targets 2011, 11, 54–67.
  26. Prevarskaya, N.; Skryma, R.; Shuba, Y. Ion channels and the hallmarks of cancer. Trends Mol. Med. 2010, 16, 107–121.
  27. Steinritz, D.; Stenger, B.; Dietrich, A.; Gudermann, T.; Popp, T. TRPs in Tox: Involvement of Transient Receptor Potential-Channels in Chemical-Induced Organ Toxicity-A Structured Review. Cells 2018, 7, 98.
  28. Koivisto, A.P.; Belvisi, M.G.; Gaudet, R.; Szallasi, A. Advances in TRP channel drug discovery: From target validation to clinical studies. Nat. Rev. Drug Discov. 2022, 21, 41–59.
  29. Stokłosa, P.; Borgström, A.; Kappel, S.; Peinelt, C. TRP Channels in Digestive Tract Cancers. Int. J. Mol. Sci. 2020, 21, 1877.
  30. Prevarskaya, N.; Zhang, L.; Barritt, G. TRP channels in cancer. Biochim. Biophys. Acta 2007, 1772, 937–946.
  31. Duncan, L.M.; Deeds, J.; Hunter, J.; Shao, J.; Holmgren, L.M.; Woolf, E.A.; Tepper, R.I.; Shyjan, A.W. Down-regulation of the novel gene melastatin correlates with potential for melanoma metastasis. Cancer Res. 1998, 58, 1515–1520.
  32. Tsavaler, L.; Shapero, M.H.; Morkowski, S.; Laus, R. Trp-p8, a novel prostate-specific gene, is up-regulated in prostate cancer and other malignancies and shares high homology with transient receptor potential calcium channel proteins. Cancer Res. 2001, 61, 3760–3769.
  33. Fuessel, S.; Sickert, D.; Meye, A.; Klenk, U.; Schmidt, U.; Schmitz, M.; Rost, A.K.; Weigle, B.; Kiessling, A.; Wirth, M.P. Multiple tumor marker analyses (PSA, hK2, PSCA, trp-p8) in primary prostate cancers using quantitative RT-PCR. Int. J. Oncol. 2003, 23, 221–228.
  34. Fixemer, T.; Wissenbach, U.; Flockerzi, V.; Bonkhoff, H. Expression of the Ca2+-selective cation channel TRPV6 in human prostate cancer: A novel prognostic marker for tumor progression. Oncogene 2003, 22, 7858–7861.
  35. Bödding, M. TRP proteins and cancer. Cell. Signal 2007, 19, 617–624.
  36. Bai, S.; Wei, Y.; Liu, R.; Chen, Y.; Ma, W.; Wang, M.; Chen, L.; Luo, Y.; Du, J. The role of transient receptor potential channels in metastasis. Biomed. Pharmacother. 2023, 158, 114074.
  37. Gkika, D.; Prevarskaya, N. TRP channels in prostate cancer: The good, the bad and the ugly? Asian J. Androl. 2011, 13, 673–676.
  38. Prevarskaya, N.; Skryma, R.; Shuba, Y. Calcium in tumour metastasis: New roles for known actors. Nat. Rev. Cancer 2011, 11, 609–618.
  39. Mukhopadhyay, I.; Gomes, P.; Aranake, S.; Shetty, M.; Karnik, P.; Damle, M.; Kuruganti, S.; Thorat, S.; Khairatkar-Joshi, N. Expression of functional TRPA1 receptor on human lung fibroblast and epithelial cells. J. Recept. Signal Transduct. Res. 2011, 31, 350–358.
  40. Cvetkov, T.L.; Huynh, K.W.; Cohen, M.R.; Moiseenkova-Bell, V.Y. Molecular architecture and subunit organization of TRPA1 ion channel revealed by electron microscopy. J. Biol. Chem. 2011, 286, 38168–38176.
  41. Landini, L.; Souza Monteiro de Araujo, D.; Titiz, M.; Geppetti, P.; Nassini, R.; De Logu, F. TRPA1 Role in Inflammatory Disorders: What Is Known So Far? Int. J. Mol. Sci. 2022, 23, 4529.
  42. Souza Monteiro de Araujo, D.; Nassini, R.; Geppetti, P.; De Logu, F. TRPA1 as a therapeutic target for nociceptive pain. Expert. Opin. Ther. Targets 2020, 24, 997–1008.
  43. Trevisani, M.; Siemens, J.; Materazzi, S.; Bautista, D.M.; Nassini, R.; Campi, B.; Imamachi, N.; Andrè, E.; Patacchini, R.; Cottrell, G.S.; et al. 4-Hydroxynonenal, an endogenous aldehyde, causes pain and neurogenic inflammation through activation of the irritant receptor TRPA1. Proc. Natl. Acad. Sci. USA 2007, 104, 13519–13524.
  44. Nilius, B.; Appendino, G.; Owsianik, G. The transient receptor potential channel TRPA1: From gene to pathophysiology. Pflug. Arch. 2012, 464, 425–458.
  45. Nassini, R.; Fusi, C.; Materazzi, S.; Coppi, E.; Tuccinardi, T.; Marone, I.M.; De Logu, F.; Preti, D.; Tonello, R.; Chiarugi, A.; et al. The TRPA1 channel mediates the analgesic action of dipyrone and pyrazolone derivatives. Br. J. Pharmacol. 2015, 172, 3397–3411.
  46. Iannone, L.F.; De Logu, F.; Geppetti, P.; De Cesaris, F. The role of TRP ion channels in migraine and headache. Neurosci. Lett. 2022, 768, 136380.
  47. De Logu, F.; Li Puma, S.; Landini, L.; Tuccinardi, T.; Poli, G.; Preti, D.; De Siena, G.; Patacchini, R.; Tsagareli, M.G.; Geppetti, P.; et al. The acyl-glucuronide metabolite of ibuprofen has analgesic and anti-inflammatory effects via the TRPA1 channel. Pharmacol. Res. 2019, 142, 127–139.
  48. De Logu, F.; Trevisan, G.; Marone, I.M.; Coppi, E.; Padilha Dalenogare, D.; Titiz, M.; Marini, M.; Landini, L.; Souza Monteiro de Araujo, D.; Li Puma, S.; et al. Oxidative stress mediates thalidomide-induced pain by targeting peripheral TRPA1 and central TRPV4. BMC Biol. 2020, 18, 197.
  49. García-Añoveros, J.; Duggan, A. Frontiers in Neuroscience. TRPA1 in Auditory and Nociceptive Organs. In TRP Ion Channel Function in Sensory Transduction and Cellular Signaling Cascades; Liedtke, W.B., Heller, S., Eds.; Copyright© 2023; CRC Press: Boca Raton, FL, USA; Taylor & Francis: Oxford, UK, 2007.
  50. Earley, S.; Gonzales, A.L.; Crnich, R. Endothelium-dependent cerebral artery dilation mediated by TRPA1 and Ca2+-Activated K+ channels. Circ. Res. 2009, 104, 987–994.
  51. Nassini, R.; Pedretti, P.; Moretto, N.; Fusi, C.; Carnini, C.; Facchinetti, F.; Viscomi, A.R.; Pisano, A.R.; Stokesberry, S.; Brunmark, C.; et al. Transient receptor potential ankyrin 1 channel localized to non-neuronal airway cells promotes non-neurogenic inflammation. PLoS ONE 2012, 7, e42454.
  52. Meents, J.E.; Ciotu, C.I.; Fischer, M.J.M. TRPA1: A molecular view. J. Neurophysiol. 2019, 121, 427–443.
  53. Maglie, R.; Souza Monteiro de Araujo, D.; Antiga, E.; Geppetti, P.; Nassini, R.; De Logu, F. The Role of TRPA1 in Skin Physiology and Pathology. Int. J. Mol. Sci. 2021, 22, 3065.
  54. Andrade, E.L.; Meotti, F.C.; Calixto, J.B. TRPA1 antagonists as potential analgesic drugs. Pharmacol. Ther. 2012, 133, 189–204.
  55. Baraldi, P.G.; Preti, D.; Materazzi, S.; Geppetti, P. Transient receptor potential ankyrin 1 (TRPA1) channel as emerging target for novel analgesics and anti-inflammatory agents. J. Med. Chem. 2010, 53, 5085–5107.
  56. Bellono, N.W.; Kammel, L.G.; Zimmerman, A.L.; Oancea, E. UV light phototransduction activates transient receptor potential A1 ion channels in human melanocytes. Proc. Natl. Acad. Sci. USA 2013, 110, 2383–2388.
  57. Bellono, N.W.; Oancea, E. UV light phototransduction depolarizes human melanocytes. Channels 2013, 7, 243–248.
  58. Büch, T.R.; Schäfer, E.A.; Demmel, M.T.; Boekhoff, I.; Thiermann, H.; Gudermann, T.; Steinritz, D.; Schmidt, A. Functional expression of the transient receptor potential channel TRPA1, a sensor for toxic lung inhalants, in pulmonary epithelial cells. Chem. Biol. Interact. 2013, 206, 462–471.
  59. Earley, S. TRPA1 channels in the vasculature. Br. J. Pharmacol. 2012, 167, 13–22.
  60. Oh, M.H.; Oh, S.Y.; Lu, J.; Lou, H.; Myers, A.C.; Zhu, Z.; Zheng, T. TRPA1-dependent pruritus in IL-13-induced chronic atopic dermatitis. J. Immunol. 2013, 191, 5371–5382.
  61. Prasad, P.; Yanagihara, A.A.; Small-Howard, A.L.; Turner, H.; Stokes, A.J. Secretogranin III directs secretory vesicle biogenesis in mast cells in a manner dependent upon interaction with chromogranin A. J. Immunol. 2008, 181, 5024–5034.
  62. Takizawa, M.; Harada, K.; Nakamura, K.; Tsuboi, T. Transient receptor potential ankyrin 1 channels are involved in spontaneous peptide hormone release from astrocytes. Biochem. Biophys. Res. Commun. 2018, 501, 988–995.
  63. Hamilton, N.B.; Kolodziejczyk, K.; Kougioumtzidou, E.; Attwell, D. Proton-gated Ca2+-permeable TRP channels damage myelin in conditions mimicking ischaemia. Nature 2016, 529, 523–527.
  64. De Logu, F.; Nassini, R.; Materazzi, S.; Carvalho Gonçalves, M.; Nosi, D.; Rossi Degl’Innocenti, D.; Marone, I.M.; Ferreira, J.; Li Puma, S.; Benemei, S.; et al. Schwann cell TRPA1 mediates neuroinflammation that sustains macrophage-dependent neuropathic pain in mice. Nat. Commun. 2017, 8, 1887.
  65. De Logu, F.; Li Puma, S.; Landini, L.; Portelli, F.; Innocenti, A.; de Araujo, D.S.M.; Janal, M.N.; Patacchini, R.; Bunnett, N.W.; Geppetti, P.; et al. Schwann cells expressing nociceptive channel TRPA1 orchestrate ethanol-evoked neuropathic pain in mice. J. Clin. Investig. 2019, 129, 5424–5441.
  66. Cojocaru, F.; Şelescu, T.; Domocoş, D.; Măruţescu, L.; Chiritoiu, G.; Chelaru, N.R.; Dima, S.; Mihăilescu, D.; Babes, A.; Cucu, D. Functional expression of the transient receptor potential ankyrin type 1 channel in pancreatic adenocarcinoma cells. Sci. Rep. 2021, 11, 2018.
  67. De Logu, F.; Souza Monteiro de Araujo, D.; Ugolini, F.; Iannone, L.F.; Vannucchi, M.; Portelli, F.; Landini, L.; Titiz, M.; De Giorgi, V.; Geppetti, P.; et al. The TRPA1 Channel Amplifies the Oxidative Stress Signal in Melanoma. Cells 2021, 10, 3131.
  68. Bernardini, M.; Brossa, A.; Chinigo, G.; Grolez, G.P.; Trimaglio, G.; Allart, L.; Hulot, A.; Marot, G.; Genova, T.; Joshi, A.; et al. Transient Receptor Potential Channel Expression Signatures in Tumor-Derived Endothelial Cells: Functional Roles in Prostate Cancer Angiogenesis. Cancers 2019, 11, 956.
  69. Park, J.; Shim, M.K.; Jin, M.; Rhyu, M.R.; Lee, Y. Methyl syringate, a TRPA1 agonist represses hypoxia-induced cyclooxygenase-2 in lung cancer cells. Phytomedicine 2016, 23, 324–329.
  70. Takahashi, N.; Chen, H.Y.; Harris, I.S.; Stover, D.G.; Selfors, L.M.; Bronson, R.T.; Deraedt, T.; Cichowski, K.; Welm, A.L.; Mori, Y.; et al. Cancer Cells Co-opt the Neuronal Redox-Sensing Channel TRPA1 to Promote Oxidative-Stress Tolerance. Cancer Cell 2018, 33, 985–1003.
  71. Antoniazzi, C.T.D.; Nassini, R.; Rigo, F.K.; Milioli, A.M.; Bellinaso, F.; Camponogara, C.; Silva, C.R.; de Almeida, A.S.; Rossato, M.F.; De Logu, F.; et al. Transient receptor potential ankyrin 1 (TRPA1) plays a critical role in a mouse model of cancer pain. Int. J. Cancer 2019, 144, 355–365.
  72. De Logu, F.; De Prá, S.D.; de David Antoniazzi, C.T.; Kudsi, S.Q.; Ferro, P.R.; Landini, L.; Rigo, F.K.; de Bem Silveira, G.; Silveira, P.C.L.; Oliveira, S.M.; et al. Macrophages and Schwann cell TRPA1 mediate chronic allodynia in a mouse model of complex regional pain syndrome type I. Brain Behav. Immun. 2020, 88, 535–546.
  73. De Logu, F.; Marini, M.; Landini, L.; Souza Monteiro de Araujo, D.; Bartalucci, N.; Trevisan, G.; Bruno, G.; Marangoni, M.; Schmidt, B.L.; Bunnett, N.W.; et al. Peripheral Nerve Resident Macrophages and Schwann Cells Mediate Cancer-Induced Pain. Cancer Res. 2021, 81, 3387–3401.
  74. Rades, D.; Schild, S.E.; Abrahm, J.L. Treatment of painful bone metastases. Nat. Rev. Clin. Oncol. 2010, 7, 220–229.
  75. de Almeida, A.S.; Rigo, F.K.; De Prá, S.D.; Milioli, A.M.; Pereira, G.C.; Lückemeyer, D.D.; Antoniazzi, C.T.; Kudsi, S.Q.; Araújo, D.; Oliveira, S.M.; et al. Role of transient receptor potential ankyrin 1 (TRPA1) on nociception caused by a murine model of breast carcinoma. Pharmacol. Res. 2020, 152, 104576.
  76. de Almeida, A.S.; Pereira, G.C.; Brum, E.D.S.; Silva, C.R.; Antoniazzi, C.T.D.; Ardisson-Araújo, D.; Oliveira, S.M.; Trevisan, G. Role of TRPA1 expressed in bone tissue and the antinociceptive effect of the TRPA1 antagonist repeated administration in a breast cancer pain model. Life Sci. 2021, 276, 119469.
  77. Landini, L.; Marini, M.; Souza Monteiro de Araujo, D.; Romitelli, A.; Montini, M.; Albanese, V.; Titiz, M.; Innocenti, A.; Bianchini, F.; Geppetti, P.; et al. Schwann cell insulin-like growth factor receptor type-1 mediates metastatic bone cancer pain in mice. Brain Behav. Immun. 2023, 110, 348–364.
  78. Kim, H.; Kim, J.; Jeon, J.P.; Myeong, J.; Wie, J.; Hong, C.; Kim, H.J.; Jeon, J.H.; So, I. The roles of G proteins in the activation of TRPC4 and TRPC5 transient receptor potential channels. Channels 2012, 6, 333–343.
  79. Chen, X.; Sooch, G.; Demaree, I.S.; White, F.A.; Obukhov, A.G. Transient Receptor Potential Canonical (TRPC) Channels: Then and Now. Cells 2020, 9, 1983.
  80. Elzamzamy, O.M.; Penner, R.; Hazlehurst, L.A. The Role of TRPC1 in Modulating Cancer Progression. Cells 2020, 9, 388.
  81. Dhennin-Duthille, I.; Gautier, M.; Faouzi, M.; Guilbert, A.; Brevet, M.; Vaudry, D.; Ahidouch, A.; Sevestre, H.; Ouadid-Ahidouch, H. High expression of transient receptor potential channels in human breast cancer epithelial cells and tissues: Correlation with pathological parameters. Cell. Physiol. Biochem. 2011, 28, 813–822.
  82. Azimi, I.; Milevskiy, M.J.G.; Kaemmerer, E.; Turner, D.; Yapa, K.; Brown, M.A.; Thompson, E.W.; Roberts-Thomson, S.J.; Monteith, G.R. TRPC1 is a differential regulator of hypoxia-mediated events and Akt signalling in PTEN-deficient breast cancer cells. J. Cell Sci. 2017, 130, 2292–2305.
  83. Zhang, Y.; Lun, X.; Guo, W. Expression of TRPC1 and SBEM protein in breast cancer tissue and its relationship with clinicopathological features and prognosis of patients. Oncol. Lett. 2020, 20, 392.
  84. Van den Eynde, C.; De Clercq, K.; Vriens, J. Transient Receptor Potential Channels in the Epithelial-to-Mesenchymal Transition. Int. J. Mol. Sci. 2021, 22, 8188.
  85. Iwatsuki, M.; Mimori, K.; Yokobori, T.; Ishi, H.; Beppu, T.; Nakamori, S.; Baba, H.; Mori, M. Epithelial-mesenchymal transition in cancer development and its clinical significance. Cancer Sci. 2010, 101, 293–299.
  86. Tai, Y.K.; Chan, K.K.W.; Fong, C.H.H.; Ramanan, S.; Yap, J.L.Y.; Yin, J.N.; Yip, Y.S.; Tan, W.R.; Koh, A.P.F.; Tan, N.S.; et al. Modulated TRPC1 Expression Predicts Sensitivity of Breast Cancer to Doxorubicin and Magnetic Field Therapy: Segue Towards a Precision Medicine Approach. Front. Oncol. 2021, 11, 783803.
  87. Jiang, H.N.; Zeng, B.; Zhang, Y.; Daskoulidou, N.; Fan, H.; Qu, J.M.; Xu, S.Z. Involvement of TRPC channels in lung cancer cell differentiation and the correlation analysis in human non-small cell lung cancer. PLoS ONE 2013, 8, e67637.
  88. Maklad, A.; Sharma, A.; Azimi, I. Calcium Signaling in Brain Cancers: Roles and Therapeutic Targeting. Cancers 2019, 11, 145.
  89. Bomben, V.C.; Sontheimer, H. Disruption of transient receptor potential canonical channel 1 causes incomplete cytokinesis and slows the growth of human malignant gliomas. Glia 2010, 58, 1145–1156.
  90. Bomben, V.C.; Turner, K.L.; Barclay, T.T.; Sontheimer, H. Transient receptor potential canonical channels are essential for chemotactic migration of human malignant gliomas. J. Cell Physiol. 2011, 226, 1879–1888.
  91. Wang, B.; Li, W.; Meng, X.; Zou, F. Hypoxia up-regulates vascular endothelial growth factor in U-87 MG cells: Involvement of TRPC1. Neurosci. Lett. 2009, 459, 132–136.
  92. Farfariello, V.; Gordienko, D.V.; Mesilmany, L.; Touil, Y.; Germain, E.; Fliniaux, I.; Desruelles, E.; Gkika, D.; Roudbaraki, M.; Shapovalov, G.; et al. TRPC3 shapes the ER-mitochondria Ca2+ transfer characterizing tumour-promoting senescence. Nat. Commun. 2022, 13, 956.
  93. Sakellakis, M.; Chalkias, A. The Role οf Ion Channels in the Development and Progression of Prostate Cancer. Mol. Diagn. Ther. 2023, 27, 227–242.
  94. Wang, Y.; Qi, Y.X.; Qi, Z.; Tsang, S.Y. TRPC3 Regulates the Proliferation and Apoptosis Resistance of Triple Negative Breast Cancer Cells through the TRPC3/RASA4/MAPK Pathway. Cancers 2019, 11, 558.
  95. Lin, D.C.; Zheng, S.Y.; Zhang, Z.G.; Luo, J.H.; Zhu, Z.L.; Li, L.; Chen, L.S.; Lin, X.; Sham, J.S.K.; Lin, M.J.; et al. TRPC3 promotes tumorigenesis of gastric cancer via the CNB2/GSK3β/NFATc2 signaling pathway. Cancer Lett. 2021, 519, 211–225.
  96. Zhang, Y.; Wang, X. Targeting the Wnt/β-catenin signaling pathway in cancer. J. Hematol. Oncol. 2020, 13, 165.
  97. Wang, T.; Chen, Z.; Zhu, Y.; Pan, Q.; Liu, Y.; Qi, X.; Jin, L.; Jin, J.; Ma, X.; Hua, D. Inhibition of transient receptor potential channel 5 reverses 5-Fluorouracil resistance in human colorectal cancer cells. J. Biol. Chem. 2015, 290, 448–456.
  98. Ma, X.; Cai, Y.; He, D.; Zou, C.; Zhang, P.; Lo, C.Y.; Xu, Z.; Chan, F.L.; Yu, S.; Chen, Y.; et al. Transient receptor potential channel TRPC5 is essential for P-glycoprotein induction in drug-resistant cancer cells. Proc. Natl. Acad. Sci. USA 2012, 109, 16282–16287.
  99. Dong, Y.; Pan, Q.; Jiang, L.; Chen, Z.; Zhang, F.; Liu, Y.; Xing, H.; Shi, M.; Li, J.; Li, X.; et al. Tumor endothelial expression of P-glycoprotein upon microvesicular transfer of TrpC5 derived from adriamycin-resistant breast cancer cells. Biochem. Biophys. Res. Commun. 2014, 446, 85–90.
  100. Zhang, P.; Liu, X.; Li, H.; Chen, Z.; Yao, X.; Jin, J.; Ma, X. TRPC5-induced autophagy promotes drug resistance in breast carcinoma via CaMKKβ/AMPKα/mTOR pathway. Sci. Rep. 2017, 7, 3158.
  101. Zhu, Y.; Pan, Q.; Meng, H.; Jiang, Y.; Mao, A.; Wang, T.; Hua, D.; Yao, X.; Jin, J.; Ma, X. Enhancement of vascular endothelial growth factor release in long-term drug-treated breast cancer via transient receptor potential channel 5-Ca2+-hypoxia-inducible factor 1α pathway. Pharmacol. Res. 2015, 93, 36–42.
  102. Chen, Z.; Zhu, Y.; Dong, Y.; Zhang, P.; Han, X.; Jin, J.; Ma, X. Overexpression of TrpC5 promotes tumor metastasis via the HIF-1α-Twist signaling pathway in colon cancer. Clin. Sci. 2017, 131, 2439–2450.
  103. Aydar, E.; Yeo, S.; Djamgoz, M.; Palmer, C. Abnormal expression, localization and interaction of canonical transient receptor potential ion channels in human breast cancer cell lines and tissues: A potential target for breast cancer diagnosis and therapy. Cancer Cell Int. 2009, 9, 23.
  104. Xu, J.; Yang, Y.; Xie, R.; Liu, J.; Nie, X.; An, J.; Wen, G.; Liu, X.; Jin, H.; Tuo, B. The NCX1/TRPC6 Complex Mediates TGFβ-Driven Migration and Invasion of Human Hepatocellular Carcinoma Cells. Cancer Res. 2018, 78, 2564–2576.
  105. Bai, L.P.; Chen, Y.L.; Zheng, A. Pharmacological targeting transient receptor potential canonical channel 6 modulates biological behaviors for cervical cancer HeLa and SiHA cell. Cancer Cell Int. 2022, 22, 145.
  106. Gao, Y.; Liao, P. TRPM4 channel and cancer. Cancer Lett. 2019, 454, 66–69.
  107. Zhang, M.; Ma, Y.; Ye, X.; Zhang, N.; Pan, L.; Wang, B. TRP (transient receptor potential) ion channel family: Structures, biological functions and therapeutic interventions for diseases. Signal Transduct. Target. Ther. 2023, 8, 261.
  108. Deeds, J.; Cronin, F.; Duncan, L.M. Patterns of melastatin mRNA expression in melanocytic tumors. Hum. Pathol. 2000, 31, 1346–1356.
  109. Erickson, L.A.; Letts, G.A.; Shah, S.M.; Shackelton, J.B.; Duncan, L.M. TRPM1 (Melastatin-1/MLSN1) mRNA expression in Spitz nevi and nodular melanomas. Mod. Pathol. 2009, 22, 969–976.
  110. Hsieh, C.C.; Su, Y.C.; Jiang, K.Y.; Ito, T.; Li, T.W.; Kaku-Ito, Y.; Cheng, S.T.; Chen, L.T.; Hwang, D.Y.; Shen, C.H. TRPM1 promotes tumor progression in acral melanoma by activating the Ca2+/CaMKIIδ/AKT pathway. J. Adv. Res. 2023, 43, 45–57.
  111. Miller, B.A. TRPM2 in Cancer. Cell Calcium 2019, 80, 8–17.
  112. 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, 1614–1624.
  113. Hara, Y.; Wakamori, M.; Ishii, M.; Maeno, E.; Nishida, M.; Yoshida, T.; Yamada, H.; Shimizu, S.; Mori, E.; Kudoh, J.; et al. LTRPC2 Ca2+-permeable channel activated by changes in redox status confers susceptibility to cell death. Mol. Cell 2002, 9, 163–173.
  114. Sumoza-Toledo, A.; Penner, R. TRPM2: A multifunctional ion channel for calcium signalling. J. Physiol. 2011, 589, 1515–1525.
  115. Lü, W.; Du, J. The N-terminal domain in TRPM2 channel is a conserved nucleotide binding site. J. Gen. Physiol. 2020, 152, e201912555.
  116. Bao, L.; Chen, S.J.; Conrad, K.; Keefer, K.; Abraham, T.; Lee, J.P.; Wang, J.; Zhang, X.Q.; Hirschler-Laszkiewicz, I.; Wang, H.G.; et al. Depletion of the Human Ion Channel TRPM2 in Neuroblastoma Demonstrates Its Key Role in Cell Survival through Modulation of Mitochondrial Reactive Oxygen Species and Bioenergetics. J. Biol. Chem. 2016, 291, 24449–24464.
  117. Miller, B.A.; Zhang, W. TRP channels as mediators of oxidative stress. Adv. Exp. Med. Biol. 2011, 704, 531–544.
  118. Zhang, W.; Chu, X.; Tong, Q.; Cheung, J.Y.; Conrad, K.; Masker, K.; Miller, B.A. A novel TRPM2 isoform inhibits calcium influx and susceptibility to cell death. J. Biol. Chem. 2003, 278, 16222–16229.
  119. Chen, S.J.; Zhang, W.; Tong, Q.; Conrad, K.; Hirschler-Laszkiewicz, I.; Bayerl, M.; Kim, J.K.; Cheung, J.Y.; Miller, B.A. Role of TRPM2 in cell proliferation and susceptibility to oxidative stress. Am. J. Physiol. Cell Physiol. 2013, 304, C548–C560.
  120. Chen, S.J.; Hoffman, N.E.; Shanmughapriya, S.; Bao, L.; Keefer, K.; Conrad, K.; Merali, S.; Takahashi, Y.; Abraham, T.; Hirschler-Laszkiewicz, I.; et al. A splice variant of the human ion channel TRPM2 modulates neuroblastoma tumor growth through hypoxia-inducible factor (HIF)-1/2α. J. Biol. Chem. 2014, 289, 36284–36302.
  121. Hirschler-Laszkiewicz, I.; Chen, S.J.; Bao, L.; Wang, J.; Zhang, X.Q.; Shanmughapriya, S.; Keefer, K.; Madesh, M.; Cheung, J.Y.; Miller, B.A. 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, C571–C586.
  122. Gershkovitz, M.; Caspi, Y.; Fainsod-Levi, T.; Katz, B.; Michaeli, J.; Khawaled, S.; Lev, S.; Polyansky, L.; Shaul, M.E.; Sionov, R.V.; et al. TRPM2 Mediates Neutrophil Killing of Disseminated Tumor Cells. Cancer Res. 2018, 78, 2680–2690.
  123. Grimm, C.; Kraft, R.; Sauerbruch, S.; Schultz, G.; Harteneck, C. Molecular and functional characterization of the melastatin-related cation channel TRPM3. J. Biol. Chem. 2003, 278, 21493–21501.
  124. Hall, D.P.; Cost, N.G.; Hegde, S.; Kellner, E.; Mikhaylova, O.; Stratton, Y.; Ehmer, B.; Abplanalp, W.A.; Pandey, R.; Biesiada, J.; et al. TRPM3 and miR-204 establish a regulatory circuit that controls oncogenic autophagy in clear cell renal cell carcinoma. Cancer Cell 2014, 26, 738–753.
  125. Chinigò, G.; Castel, H.; Chever, O.; Gkika, D. TRP Channels in Brain Tumors. Front. Cell Dev. Biol. 2021, 9, 617801.
  126. Duvoisin, R.M.; Haley, T.L.; Ren, G.; Strycharska-Orczyk, I.; Bonaparte, J.P.; Morgans, C.W. Autoantibodies in Melanoma-Associated Retinopathy Recognize an Epitope Conserved Between TRPM1 and TRPM3. Investig. Ophthalmol. Vis. Sci. 2017, 58, 2732–2738.
  127. Lin, C.; Blessing, A.M.; Pulliam, T.L.; Shi, Y.; Wilkenfeld, S.R.; Han, J.J.; Murray, M.M.; Pham, A.H.; Duong, K.; Brun, S.N.; et al. Inhibition of CAMKK2 impairs autophagy and castration-resistant prostate cancer via suppression of AMPK-ULK1 signaling. Oncogene 2021, 40, 1690–1705.
  128. Ying, Z.; Li, Y.; Wu, J.; Zhu, X.; Yang, Y.; Tian, H.; Li, W.; Hu, B.; Cheng, S.Y.; Li, M. Loss of miR-204 expression enhances glioma migration and stem cell-like phenotype. Cancer Res. 2013, 73, 990–999.
  129. Bian, Z.; Ji, W.; Xu, B.; Huo, Z.; Huang, H.; Huang, J.; Jiao, J.; Shao, J.; Zhang, X. Noncoding RNAs involved in the STAT3 pathway in glioma. Cancer Cell Int. 2021, 21, 445.
  130. Cáceres, M.; Ortiz, L.; Recabarren, T.; Romero, A.; Colombo, A.; Leiva-Salcedo, E.; Varela, D.; Rivas, J.; Silva, I.; Morales, D.; et al. TRPM4 Is a Novel Component of the Adhesome Required for Focal Adhesion Disassembly, Migration and Contractility. PLoS ONE 2015, 10, e0130540.
  131. Barbet, G.; Demion, M.; Moura, I.C.; Serafini, N.; Léger, T.; Vrtovsnik, F.; Monteiro, R.C.; Guinamard, R.; Kinet, J.P.; Launay, P. The calcium-activated nonselective cation channel TRPM4 is essential for the migration but not the maturation of dendritic cells. Nat. Immunol. 2008, 9, 1148–1156.
  132. Vennekens, R.; Olausson, J.; Meissner, M.; Bloch, W.; Mathar, I.; Philipp, S.E.; Schmitz, F.; Weissgerber, P.; Nilius, B.; Flockerzi, V.; et al. Increased IgE-dependent mast cell activation and anaphylactic responses in mice lacking the calcium-activated nonselective cation channel TRPM4. Nat. Immunol. 2007, 8, 312–320.
  133. Sarmiento, D.; Montorfano, I.; Cerda, O.; Cáceres, M.; Becerra, A.; Cabello-Verrugio, C.; Elorza, A.A.; Riedel, C.; Tapia, P.; Velásquez, L.A.; et al. Increases in reactive oxygen species enhance vascular endothelial cell migration through a mechanism dependent on the transient receptor potential melastatin 4 ion channel. Microvasc. Res. 2015, 98, 187–196.
  134. Chinigò, G.; Fiorio Pla, A.; Gkika, D. TRP Channels and Small GTPases Interplay in the Main Hallmarks of Metastatic Cancer. Front. Pharmacol. 2020, 11, 581455.
  135. Holzmann, C.; Kappel, S.; Kilch, T.; Jochum, M.M.; Urban, S.K.; Jung, V.; Stöckle, M.; Rother, K.; Greiner, M.; Peinelt, C. Transient receptor potential melastatin 4 channel contributes to migration of androgen-insensitive prostate cancer cells. Oncotarget 2015, 6, 41783–41793.
  136. Sagredo, A.I.; Sagredo, E.A.; Pola, V.; Echeverría, C.; Andaur, R.; Michea, L.; Stutzin, A.; Simon, F.; Marcelain, K.; Armisén, R. TRPM4 channel is involved in regulating epithelial to mesenchymal transition, migration, and invasion of prostate cancer cell lines. J. Cell Physiol. 2019, 234, 2037–2050.
  137. Berg, K.D.; Soldini, D.; Jung, M.; Dietrich, D.; Stephan, C.; Jung, K.; Dietel, M.; Vainer, B.; Kristiansen, G. TRPM4 protein expression in prostate cancer: A novel tissue biomarker associated with risk of biochemical recurrence following radical prostatectomy. Virchows Arch. 2016, 468, 345–355.
  138. Ceylan, G.G.; Önalan, E.E.; Kuloğlu, T.; Aydoğ, G.; Keleş, İ.; Tonyali, Ş.; Ceylan, C. Potential role of melastatin-related transient receptor potential cation channel subfamily M gene expression in the pathogenesis of urinary bladder cancer. Oncol. Lett. 2016, 12, 5235–5239.
  139. Narayan, G.; Bourdon, V.; Chaganti, S.; Arias-Pulido, H.; Nandula, S.V.; Rao, P.H.; Gissmann, L.; Dürst, M.; Schneider, A.; Pothuri, B.; et al. Gene dosage alterations revealed by cDNA microarray analysis in cervical cancer: Identification of candidate amplified and overexpressed genes. Genes. Chromosomes Cancer 2007, 46, 373–384.
  140. Zhu, L.; Miao, B.; Dymerska, D.; Kuswik, M.; Bueno-Martínez, E.; Sanoguera-Miralles, L.; Velasco, E.A.; Paramasivam, N.; Schlesner, M.; Kumar, A.; et al. Germline Variants of CYBA and TRPM4 Predispose to Familial Colorectal Cancer. Cancers 2022, 14, 670.
  141. Pérez-Riesgo, E.; Gutiérrez, L.G.; Ubierna, D.; Acedo, A.; Moyer, M.P.; Núñez, L.; Villalobos, C. Transcriptomic Analysis of Calcium Remodeling in Colorectal Cancer. Int. J. Mol. Sci. 2017, 18, 922.
  142. Chen, J.; Luan, Y.; Yu, R.; Zhang, Z.; Zhang, J.; Wang, W. Transient receptor potential (TRP) channels, promising potential diagnostic and therapeutic tools for cancer. Biosci. Trends 2014, 8, 1–10.
  143. Loo, S.K.; Ch’ng, E.S.; Md Salleh, M.S.; Banham, A.H.; Pedersen, L.M.; Møller, M.B.; Green, T.M.; Wong, K.K. TRPM4 expression is associated with activated B cell subtype and poor survival in diffuse large B cell lymphoma. Histopathology 2017, 71, 98–111.
  144. Stokłosa, P.; Borgström, A.; Hauert, B.; Baur, R.; Peinelt, C. Investigation of Novel Small Molecular TRPM4 Inhibitors in Colorectal Cancer Cells. Cancers 2021, 13, 5400.
  145. Wang, J.; Qiao, S.; Liang, S.; Qian, C.; Dong, Y.; Pei, M.; Wang, H.; Wan, G. TRPM4 and TRPV2 are two novel prognostic biomarkers and promising targeted therapy in UVM. Front. Mol. Biosci. 2022, 9, 985434.
  146. de Baaij, J.H.; Hoenderop, J.G.; Bindels, R.J. Magnesium in man: Implications for health and disease. Physiol. Rev. 2015, 95, 1–46.
  147. Kim, E.Y.; Lee, J.M. Transcriptional Control of Trpm6 by the Nuclear Receptor FXR. Int. J. Mol. Sci. 2022, 23, 1980.
  148. Finkel, M.; Goldstein, A.; Steinberg, Y.; Granowetter, L.; Trachtman, H. Cisplatinum nephrotoxicity in oncology therapeutics: Retrospective review of patients treated between 2005 and 2012. Pediatr. Nephrol. 2014, 29, 2421–2424.
  149. Hofheinz, R.D.; Segaert, S.; Safont, M.J.; Demonty, G.; Prenen, H. Management of adverse events during treatment of gastrointestinal cancers with epidermal growth factor inhibitors. Crit. Rev. Oncol. Hematol. 2017, 114, 102–113.
  150. Xie, B.; Zhao, R.; Bai, B.; Wu, Y.; Xu, Y.; Lu, S.; Fang, Y.; Wang, Z.; Maswikiti, E.P.; Zhou, X.; et al. Identification of key tumorigenesis-related genes and their microRNAs in colon cancer. Oncol. Rep. 2018, 40, 3551–3560.
  151. Bautista, D.M.; Siemens, J.; Glazer, J.M.; Tsuruda, P.R.; Basbaum, A.I.; Stucky, C.L.; Jordt, S.E.; Julius, D. The menthol receptor TRPM8 is the principal detector of environmental cold. Nature 2007, 448, 204–208.
  152. Chodon, D.; Guilbert, A.; Dhennin-Duthille, I.; Gautier, M.; Telliez, M.S.; Sevestre, H.; Ouadid-Ahidouch, H. Estrogen regulation of TRPM8 expression in breast cancer cells. BMC Cancer 2010, 10, 212.
  153. Liu, J.; Chen, Y.; Shuai, S.; Ding, D.; Li, R.; Luo, R. TRPM8 promotes aggressiveness of breast cancer cells by regulating EMT via activating AKT/GSK-3β pathway. Tumour Biol. 2014, 35, 8969–8977.
  154. Yu, S.; Xu, Z.; Zou, C.; Wu, D.; Wang, Y.; Yao, X.; Ng, C.F.; Chan, F.L. Ion channel TRPM8 promotes hypoxic growth of prostate cancer cells via an O2 -independent and RACK1-mediated mechanism of HIF-1α stabilization. J. Pathol. 2014, 234, 514–525.
  155. Huang, Y.; Li, S.; Jia, Z.; Zhao, W.; Zhou, C.; Zhang, R.; Ali, D.W.; Michalak, M.; Chen, X.Z.; Tang, J. Transient Receptor Potential Melastatin 8 (TRPM8) Channel Regulates Proliferation and Migration of Breast Cancer Cells by Activating the AMPK-ULK1 Pathway to Enhance Basal Autophagy. Front. Oncol. 2020, 10, 573127.
  156. Li, Q.; Wang, X.; Yang, Z.; Wang, B.; Li, S. Menthol induces cell death via the TRPM8 channel in the human bladder cancer cell line T24. Oncology 2009, 77, 335–341.
  157. Xiao, N.; Jiang, L.M.; Ge, B.; Zhang, T.Y.; Zhao, X.K.; Zhou, X. Over-expression of TRPM8 is associated with poor prognosis in urothelial carcinoma of bladder. Tumour Biol. 2014, 35, 11499–11504.
  158. Alptekin, M.; Eroglu, S.; Tutar, E.; Sencan, S.; Geyik, M.A.; Ulasli, M.; Demiryurek, A.T.; Camci, C. Gene expressions of TRP channels in glioblastoma multiforme and relation with survival. Tumour Biol. 2015, 36, 9209–9213.
  159. Zeng, J.; Wu, Y.; Zhuang, S.; Qin, L.; Hua, S.; Mungur, R.; Pan, J.; Zhu, Y.; Zhan, R. Identification of the role of TRPM8 in glioblastoma and its effect on proliferation, apoptosis and invasion of the U251 human glioblastoma cell line. Oncol. Rep. 2019, 42, 1517–1526.
  160. Liu, J.J.; Li, L.Z.; Xu, P. Upregulation of TRPM8 can promote the colon cancer liver metastasis through mediating Akt/GSK-3 signal pathway. Biotechnol. Appl. Biochem. 2022, 69, 230–239.
  161. Chen, L.; Xiong, Y.; Li, J.; Zheng, X.; Zhou, Q.; Turner, A.; Wu, C.; Lu, B.; Jiang, J. PD-L1 Expression Promotes Epithelial to Mesenchymal Transition in Human Esophageal Cancer. Cell. Physiol. Biochem. 2017, 42, 2267–2280.
  162. Wang, G.; Cao, R.; Qian, K.; Peng, T.; Yuan, L.; Chen, L.; Cheng, S.; Xiong, Y.; Ju, L.; Wang, X.; et al. TRPM8 Inhibition Regulates the Proliferation, Migration and ROS Metabolism of Bladder Cancer Cells. Onco Targets Ther. 2020, 13, 8825–8835.
More
Information
Subjects: Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 262
Revisions: 2 times (View History)
Update Date: 31 Oct 2023
1000/1000
ScholarVision Creations