1
|
Chen L, Xu N, Gou D, Song J, Zhou M, Zhang Y, Zhang H, Zhu L, Huang W, Zhu Y, Gao C, Gu D, Xu Y, Zhou H. TRPM8 overexpression suppresses hepatocellular carcinoma progression and improves survival by modulating the RTP3/STAT3 pathway. Cancer Med 2024; 13:e70109. [PMID: 39385506 PMCID: PMC11464657 DOI: 10.1002/cam4.70109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/02/2024] [Accepted: 08/04/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND AND AIMS Hepatocellular carcinoma (HCC) is a malignant tumour associated with high morbidity and mortality rates worldwide. Recently, TRPM8 was reported to play an important role in tumour progression. However, the precise role of TRPM8 in HCC remains unclear. In this study, we explored the expression levels, molecular functions and underlying mechanisms of TRPM8 in HCC. METHODS Tissue samples were used to analyse the expression of TRPM8 to assess its diagnostic value for prognosis. Cell Counting Kit-8, EdU and colony formation assays were performed to evaluate the effects of TRPM8 on cell proliferation, whereas the Transwell assay was used to assess cell migration and invasion. The role of TRPM8 in vivo was evaluated using a mouse subcutaneous xenograft tumour model. We performed PPI network analyses to understand the possible mechanisms of TRPM8 action. RESULTS TRPM8 expression was decreased in HCC tissues and was correlated with histological grade and poor patient prognosis. Functionally, TRPM8 repressed the proliferation and metastasis of HCC cells both in vitro and in vivo by modulating the RTP3/STAT3 signalling pathway. CONCLUSION Our findings underscore the critical role of the TRPM8-RTP3-STAT3 axis in maintaining the malignant progression of HCC. Moreover, our study demonstrates that AD80 is involved in anti-tumour processes by upregulating the expression of TRPM8.
Collapse
Affiliation(s)
- Lichan Chen
- Department of Laboratory MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital; Shenzhen Key Laboratory of Medical Laboratory and Molecular Diagnostics; Guangzhou Medical UniversityShenzhenChina
| | - Nansong Xu
- State key laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - DongMei Gou
- Shenzhen Third People's HospitalSouthern University of Science and TechnologyShenzhenChina
| | - Jianning Song
- Department of Laboratory MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital; Shenzhen Key Laboratory of Medical Laboratory and Molecular Diagnostics; Guangzhou Medical UniversityShenzhenChina
| | - Mingqin Zhou
- Department of Critical Care MedicineCancer Hospital of Shantou University Medical CollegeShantouChina
| | - Yajun Zhang
- Guangzhou University of Chinese MedicineGuangzhouChina
| | - Haohua Zhang
- Department of Laboratory MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital; Shenzhen Key Laboratory of Medical Laboratory and Molecular Diagnostics; Guangzhou Medical UniversityShenzhenChina
| | - Liwen Zhu
- Department of Laboratory MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital; Shenzhen Key Laboratory of Medical Laboratory and Molecular Diagnostics; Guangzhou Medical UniversityShenzhenChina
| | - Weihong Huang
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Yue Zhu
- Medicine Department of Biochemistry and Molecular BiologyMedical College of Jinan UniversityGuangzhouChina
| | - Cheng Gao
- Department of Laboratory MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital; Shenzhen Key Laboratory of Medical Laboratory and Molecular Diagnostics; Guangzhou Medical UniversityShenzhenChina
| | - Dayong Gu
- Department of Laboratory MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital; Shenzhen Key Laboratory of Medical Laboratory and Molecular Diagnostics; Guangzhou Medical UniversityShenzhenChina
| | - Yong Xu
- Shenzhen Third People's HospitalSouthern University of Science and TechnologyShenzhenChina
| | - Hongzhong Zhou
- Department of Laboratory MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital; Shenzhen Key Laboratory of Medical Laboratory and Molecular Diagnostics; Guangzhou Medical UniversityShenzhenChina
| |
Collapse
|
2
|
Qiao S, Wu F, Wang H. Genetic and immune identification and functional analysis of TRPM8 as a potential biomarker for pancreatic adenocarcinoma proliferation. Cancer Rep (Hoboken) 2024; 7:e2108. [PMID: 38837874 PMCID: PMC11150080 DOI: 10.1002/cnr2.2108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/26/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD), a member of highly lethal malignant tumors, has a poor outcome and extremely poor prognosis. The transient receptor potential (TRP) superfamily, a group of nonselective cation channels, is capable of influencing cellular functions by regulating calcium homeostasis. In addition, it has been shown that TRP channels can also affect various cellular phenotypes by regulating gene transcription levels and are involved in the development of a variety of malignant tumors. AIMS In order to find new therapeutic targets and biomarkers to improve the clinical prognosis of pancreatic cancer, we performed genetic and immunological characterization of TRP channels in PAAD, as well as related functional and prognostic analyses. METHODS AND RESULTS We investigated the expression, genetic alterations, methylation levels, and immune infiltration levels of TRP channels in PAAD, and further also analyzed the function of TRP channels in PAAD and their prognostic value for PAAD patients. Our results suggest that TRPM8 may contribute to tumor proliferation by controlling the PI3K-AKT-mTOR signaling pathway in PAAD. CONCLUSION After careful evaluation of the accumulated data, we concluded that TRPM8 has potential as a prognostic indicator and prospective therapeutic target in PAAD.
Collapse
Affiliation(s)
- Sen Qiao
- Assisted Reproduction CenterNorthwest Women's and Children's HospitalXi'anChina
| | - Fengming Wu
- School of MedicineSoutheast UniversityNanjingJiangsuChina
| | - Hongmei Wang
- School of MedicineSoutheast UniversityNanjingJiangsuChina
- Shaanxi University of Chinese MedicineXianyangChina
| |
Collapse
|
3
|
Hou YJ, Yang XX, He L, Meng HX. Pathological mechanisms of cold and mechanical stress in modulating cancer progression. Hum Cell 2024; 37:593-606. [PMID: 38538930 DOI: 10.1007/s13577-024-01049-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/22/2024] [Indexed: 04/15/2024]
Abstract
Environmental temperature and cellular mechanical force are the inherent factors that participate in various biological processes and regulate cancer progress, which have been hot topics worldwide. They occupy a dominant part in the cancer tissues through different approaches. However, extensive investigation regarding pathological mechanisms in the carcinogenic field. After research, we found cold stress via two means to manipulate tumors: neuroscience and mechanically sensitive ion channels (MICHs) such as TRP families to regulate the physiological and pathological activities. Excessive cold stimulation mediated neuroscience acting on every cancer stage through the hypothalamus-pituitary-adrenocorticoid (HPA) to reach the target organs. Comparatively speaking, mechanical force via Piezo of MICHs controls cancer development. The progression of cancer depends on the internal activation of proto-oncogenes and the external tumorigenic factors; the above two means eventually lead to genetic disorders at the molecular level. This review summarizes the interaction of bidirectional communication between them and the tumor. It covers the main processes from cytoplasm to nucleus related to metastasis cascade and tumor immune escape.
Collapse
Affiliation(s)
- Yun-Jing Hou
- Harbin Medical University, Harbin, China
- Department of Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin-Xin Yang
- Harbin Medical University, Harbin, China
- Department of Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lin He
- Department of Stomatology, Heilongjiang Provincial Hospital, Harbin, China
| | - Hong-Xue Meng
- Harbin Medical University, Harbin, China.
- Department of Pathology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, China.
| |
Collapse
|
4
|
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 2023; 12:1065935. [PMID: 36844925 PMCID: PMC9948629 DOI: 10.3389/fonc.2022.1065935] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/15/2022] [Indexed: 02/11/2023] Open
Abstract
The transient melastatin receptor potential (TRPM) ion channel subfamily functions as cellular sensors and transducers of critical biological signal pathways by regulating ion homeostasis. Some members of TRPM have been cloned from cancerous tissues, and their abnormal expressions in various solid malignancies have been correlated with cancer cell growth, survival, or death. Recent evidence also highlights the mechanisms underlying the role of TRPMs in tumor epithelial-mesenchymal transition (EMT), autophagy, and cancer metabolic reprogramming. These implications support TRPM channels as potential molecular targets and their modulation as an innovative therapeutic approach against cancer. Here, we discuss the general characteristics of the different TRPMs, focusing on current knowledge about the connection between TRPM channels and critical features of cancer. We also cover TRPM modulators used as pharmaceutical tools in biological trials and an indication of the only clinical trial with a TRPM modulator about cancer. To conclude, the authors describe the prospects for TRPM channels in oncology.
Collapse
Affiliation(s)
- Tania Ciaglia
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | - Vincenzo Vestuto
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | - Alessia Bertamino
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | | | | |
Collapse
|
5
|
Ochoa SV, Casas Z, Albarracín SL, Sutachan JJ, Torres YP. Therapeutic potential of TRPM8 channels in cancer treatment. Front Pharmacol 2023; 14:1098448. [PMID: 37033630 PMCID: PMC10073478 DOI: 10.3389/fphar.2023.1098448] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/20/2023] [Indexed: 04/11/2023] Open
Abstract
Cancer is a multifactorial process associated with changes in signaling pathways leading to cell cycle variations and gene expression. The transient receptor potential melastatin 8 (TRPM8) channel is a non-selective cation channel expressed in neuronal and non-neuronal tissues, where it is involved in several processes, including thermosensation, differentiation, and migration. Cancer is a multifactorial process associated with changes in signaling pathways leading to variations in cell cycle and gene expression. Interestingly, it has been shown that TRPM8 channels also participate in physiological processes related to cancer, such as proliferation, survival, and invasion. For instance, TRPM8 channels have an important role in the diagnosis, prognosis, and treatment of prostate cancer. In addition, it has been reported that TRPM8 channels are involved in the progress of pancreatic, breast, bladder, colon, gastric, and skin cancers, glioblastoma, and neuroblastoma. In this review, we summarize the current knowledge on the role of TRPM8 channels in cancer progression. We also discuss the therapeutic potential of TRPM8 in carcinogenesis, which has been proposed as a molecular target for cancer therapy.
Collapse
Affiliation(s)
- Sara V. Ochoa
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá, Colombia
- Semillero de Investigación, Biofísica y Fisiología de Canales Iónicos, Pontificia Universidad Javeriana, Bogotá, Colombia
- *Correspondence: Sara V. Ochoa, ; Yolima P. Torres,
| | - Zulma Casas
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Sonia L. Albarracín
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Jhon Jairo Sutachan
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Yolima P. Torres
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá, Colombia
- *Correspondence: Sara V. Ochoa, ; Yolima P. Torres,
| |
Collapse
|
6
|
Vestuto V, Di Sarno V, Musella S, Di Dona G, Moltedo O, Gomez-Monterrey IM, Bertamino A, Ostacolo C, Campiglia P, Ciaglia T. New Frontiers on ER Stress Modulation: Are TRP Channels the Leading Actors? Int J Mol Sci 2022; 24:185. [PMID: 36613628 PMCID: PMC9820239 DOI: 10.3390/ijms24010185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic structure, playing multiple roles including calcium storage, protein synthesis and lipid metabolism. During cellular stress, variations in ER homeostasis and its functioning occur. This condition is referred as ER stress and generates a cascade of signaling events termed unfolded protein response (UPR), activated as adaptative response to mitigate the ER stress condition. In this regard, calcium levels play a pivotal role in ER homeostasis and therefore in cell fate regulation since calcium signaling is implicated in a plethora of physiological processes, but also in disease conditions such as neurodegeneration, cancer and metabolic disorders. A large body of emerging evidence highlighted the functional role of TRP channels and their ability to promote cell survival or death depending on endoplasmic reticulum stress resolution, making them an attractive target. Thus, in this review we focused on the TRP channels' correlation to UPR-mediated ER stress in disease pathogenesis, providing an overview of their implication in the activation of this cellular response.
Collapse
Affiliation(s)
- Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Giorgio Di Dona
- Pineta Grande Hospital, Via Domiziana, km 30/00, 81030 Castel Volturno, CE, Italy
| | - Ornella Moltedo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | | | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
- European Biomedical Research Institute of Salerno, Via S. De Renzi 50, 84125 Salerno, SA, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| |
Collapse
|
7
|
Grolez GP, Chinigò G, Barras A, Hammadi M, Noyer L, Kondratska K, Bulk E, Oullier T, Marionneau-Lambot S, Le Mée M, Rétif S, Lerondel S, Bongiovanni A, Genova T, Roger S, Boukherroub R, Schwab A, Fiorio Pla A, Gkika D. TRPM8 as an Anti-Tumoral Target in Prostate Cancer Growth and Metastasis Dissemination. Int J Mol Sci 2022; 23:ijms23126672. [PMID: 35743115 PMCID: PMC9224463 DOI: 10.3390/ijms23126672] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/05/2022] [Accepted: 06/12/2022] [Indexed: 02/04/2023] Open
Abstract
In the fight against prostate cancer (PCa), TRPM8 is one of the most promising clinical targets. Indeed, several studies have highlighted that TRPM8 involvement is key in PCa progression because of its impact on cell proliferation, viability, and migration. However, data from the literature are somewhat contradictory regarding the precise role of TRPM8 in prostatic carcinogenesis and are mostly based on in vitro studies. The purpose of this study was to clarify the role played by TRPM8 in PCa progression. We used a prostate orthotopic xenograft mouse model to show that TRPM8 overexpression dramatically limited tumor growth and metastasis dissemination in vivo. Mechanistically, our in vitro data revealed that TRPM8 inhibited tumor growth by affecting the cell proliferation and clonogenic properties of PCa cells. Moreover, TRPM8 impacted metastatic dissemination mainly by impairing cytoskeleton dynamics and focal adhesion formation through the inhibition of the Cdc42, Rac1, ERK, and FAK pathways. Lastly, we proved the in vivo efficiency of a new tool based on lipid nanocapsules containing WS12 in limiting the TRPM8-positive cells' dissemination at metastatic sites. Our work strongly supports the protective role of TRPM8 on PCa progression, providing new insights into the potential application of TRPM8 as a therapeutic target in PCa treatment.
Collapse
Affiliation(s)
- Guillaume P. Grolez
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille, 59000 Villeneuve d’Ascq, France; (G.P.G.); (G.C.); (L.N.); (K.K.); (A.F.P.)
| | - Giorgia Chinigò
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille, 59000 Villeneuve d’Ascq, France; (G.P.G.); (G.C.); (L.N.); (K.K.); (A.F.P.)
- Department of Life Science and Systems Biology, University of Turin, 10123 Turin, Italy;
| | - Alexandre Barras
- CNRS, Centrale Lille, Univ. Lille, Univ. Polytechnique Hauts-de-France, UMR 8520—IEMN, 59000 Lille, France; (A.B.); (M.H.); (R.B.)
| | - Mehdi Hammadi
- CNRS, Centrale Lille, Univ. Lille, Univ. Polytechnique Hauts-de-France, UMR 8520—IEMN, 59000 Lille, France; (A.B.); (M.H.); (R.B.)
| | - Lucile Noyer
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille, 59000 Villeneuve d’Ascq, France; (G.P.G.); (G.C.); (L.N.); (K.K.); (A.F.P.)
| | - Kateryna Kondratska
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille, 59000 Villeneuve d’Ascq, France; (G.P.G.); (G.C.); (L.N.); (K.K.); (A.F.P.)
| | - Etmar Bulk
- Institute of Physiology II, University of Münster, 48149 Münster, Germany; (E.B.); (A.S.)
| | - Thibauld Oullier
- Cancéropôle du Grand Ouest, Plateforme In Vivo, 44000 Nantes, France; (T.O.); (S.M.-L.)
| | | | - Marilyne Le Mée
- CNRS UAR44, PHENOMIN-TAAM, 45071 Orléans, France; (M.L.M.); (S.R.); (S.L.)
| | - Stéphanie Rétif
- CNRS UAR44, PHENOMIN-TAAM, 45071 Orléans, France; (M.L.M.); (S.R.); (S.L.)
| | - Stéphanie Lerondel
- CNRS UAR44, PHENOMIN-TAAM, 45071 Orléans, France; (M.L.M.); (S.R.); (S.L.)
| | - Antonino Bongiovanni
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41—UMS 2014—PLBS, University of Lille, 59000 Lille, France;
| | - Tullio Genova
- Department of Life Science and Systems Biology, University of Turin, 10123 Turin, Italy;
- Nanostructured Interfaces and Surfaces Centre of Excellence (NIS), University of Turin, 10123 Turin, Italy
| | - Sébastien Roger
- Transplantation, Immunologie et Inflammation T2I-EA 4245, Université de Tours, 37044 Tours, France;
| | - Rabah Boukherroub
- CNRS, Centrale Lille, Univ. Lille, Univ. Polytechnique Hauts-de-France, UMR 8520—IEMN, 59000 Lille, France; (A.B.); (M.H.); (R.B.)
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, 48149 Münster, Germany; (E.B.); (A.S.)
| | - Alessandra Fiorio Pla
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille, 59000 Villeneuve d’Ascq, France; (G.P.G.); (G.C.); (L.N.); (K.K.); (A.F.P.)
- Department of Life Science and Systems Biology, University of Turin, 10123 Turin, Italy;
- CNRS UAR44, PHENOMIN-TAAM, 45071 Orléans, France; (M.L.M.); (S.R.); (S.L.)
| | - Dimitra Gkika
- CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020-UMR 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, University Lille, 59000 Villeneuve d’Ascq, France
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
- Institut Universitaire de France (IUF), 75231 Paris, France
- Correspondence:
| |
Collapse
|
8
|
Zhong T, Zhang W, Guo H, Pan X, Chen X, He Q, Yang B, Ding L. The regulatory and modulatory roles of TRP family channels in malignant tumors and relevant therapeutic strategies. Acta Pharm Sin B 2022; 12:1761-1780. [PMID: 35847486 PMCID: PMC9279634 DOI: 10.1016/j.apsb.2021.11.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/11/2021] [Accepted: 10/19/2021] [Indexed: 02/08/2023] Open
Abstract
Transient receptor potential (TRP) channels are one primary type of calcium (Ca2+) permeable channels, and those relevant transmembrane and intracellular TRP channels were previously thought to be mainly associated with the regulation of cardiovascular and neuronal systems. Nowadays, however, accumulating evidence shows that those TRP channels are also responsible for tumorigenesis and progression, inducing tumor invasion and metastasis. However, the overall underlying mechanisms and possible signaling transduction pathways that TRP channels in malignant tumors might still remain elusive. Therefore, in this review, we focus on the linkage between TRP channels and the significant characteristics of tumors such as multi-drug resistance (MDR), metastasis, apoptosis, proliferation, immune surveillance evasion, and the alterations of relevant tumor micro-environment. Moreover, we also have discussed the expression of relevant TRP channels in various forms of cancer and the relevant inhibitors' efficacy. The chemo-sensitivity of the anti-cancer drugs of various acting mechanisms and the potential clinical applications are also presented. Furthermore, it would be enlightening to provide possible novel therapeutic approaches to counteract malignant tumors regarding the intervention of calcium channels of this type.
Collapse
Key Words
- 4α-PDD, 4α-phorbol-12,13-didecanoate
- ABCB, ATP-binding cassette B1
- AKT, protein kinase B
- ALA, alpha lipoic acid
- AMPK, AMP-activated protein kinase
- APB, aminoethoxydiphenyl borate
- ATP, adenosine triphosphate
- CBD, cannabidiol
- CRAC, Ca2+ release-activated Ca2+ channel
- CaR, calcium-sensing receptor
- CaSR, calcium sensing receptor
- Cancer progression
- DAG, diacylglycerol
- DBTRG, Denver Brain Tumor Research Group
- ECFC, endothelial colony-forming cells
- ECM, enhanced extracellular matrix
- EGF, epidermal growth factor
- EMT, epithelial–mesenchymal transition
- ER, endoplasmic reticulum
- ERK, extracellular signal-regulated kinase
- ETS, erythroblastosis virus E26 oncogene homolog
- FAK, focal adhesion kinase
- GADD, growth arrest and DNA damage-inducible gene
- GC, gastric cancer
- GPCR, G-protein coupled receptor
- GSC, glioma stem-like cells
- GSK, glycogen synthase kinase
- HCC, hepatocellular carcinoma
- HIF, hypoxia-induced factor
- HSC, hematopoietic stem cells
- IP3R, inositol triphosphate receptor
- Intracellular mechanism
- KO, knockout
- LOX, lipoxygenase
- LPS, lipopolysaccharide
- LRP, lipoprotein receptor-related protein
- MAPK, mitogen-activated protein kinase
- MLKL, mixed lineage kinase domain-like protein
- MMP, matrix metalloproteinases
- NEDD4, neural precursor cell expressed, developmentally down-regulated 4
- NFAT, nuclear factor of activated T-cells
- NLRP3, NLR family pyrin domain containing 3
- NO, nitro oxide
- NSCLC, non-small cell lung cancer
- Nrf2, nuclear factor erythroid 2-related factor 2
- P-gp, P-glycoprotein
- PCa, prostate cancer
- PDAC, pancreatic ductal adenocarcinoma
- PHD, prolyl hydroxylases
- PI3K, phosphoinositide 3-kinase
- PKC, protein kinase C
- PKD, polycystic kidney disease
- PLC, phospholipase C
- Programmed cancer cell death
- RNS/ROS, reactive nitrogen species/reactive oxygen species
- RTX, resiniferatoxin
- SMAD, Caenorhabditis elegans protein (Sma) and mothers against decapentaplegic (Mad)
- SOCE, store operated calcium entry
- SOR, soricimed
- STIM1, stromal interaction molecules 1
- TEC, tumor endothelial cells
- TGF, transforming growth factor-β
- TNF-α, tumor necrosis factor-α
- TRP channels
- TRPA/C/M/ML/N/P/V, transient receptor potential ankyrin/canonical/melastatin/mucolipon/NOMPC/polycystin/vanilloid
- Targeted tumor therapy
- Tumor microenvironment
- Tumor-associated immunocytes
- UPR, unfolded protein response
- VEGF, vascular endothelial growth factor
- VIP, vasoactive intestinal peptide
- VPAC, vasoactive intestinal peptide receptor subtype
- mTOR, mammalian target of rapamycin
- pFRG/RTN, parafacial respiratory group/retrotrapezoid nucleus
Collapse
|
9
|
Ji D, Fleig A, Horgen FD, Feng ZP, Sun HS. Modulators of TRPM7 and its potential as a drug target for brain tumours. Cell Calcium 2021; 101:102521. [PMID: 34953296 DOI: 10.1016/j.ceca.2021.102521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022]
Abstract
TRPM7 is a non-selective divalent cation channel with an alpha-kinase domain. Corresponding with its broad expression, TRPM7 has a role in a wide range of cell functions, including proliferation, migration, and survival. Growing evidence shows that TRPM7 is also aberrantly expressed in various cancers, including brain cancers. Because ion channels have widespread tissue distribution and result in extensive physiological consequences when dysfunctional, these proteins can be compelling drug targets. In fact, ion channels comprise the third-largest drug target type, following enzymes and receptors. Literature has shown that suppression of TRPM7 results in inhibition of migration, invasion, and proliferation in several human brain tumours. Therefore, TRPM7 presents a potential target for therapeutic brain tumour interventions. This article reviews current literature on TRPM7 as a potential drug target in the context of brain tumours and provides an overview of various selective and non-selective modulators of the channel relevant to pharmacology, oncology, and ion channel function.
Collapse
Affiliation(s)
- Delphine Ji
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine and Cancer Center at the University of Hawaii, Honolulu, Hawaii 96813, USA
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, Hawaii 96744, USA
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8.
| | - Hong-Shuo Sun
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8; Department of Pharmacology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8; Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario, Canada M5S 3M2.
| |
Collapse
|
10
|
Panda S, Chatterjee O, Roy L, Chatterjee S. Targeting Ca 2+ signaling: A new arsenal against cancer. Drug Discov Today 2021; 27:923-934. [PMID: 34793973 DOI: 10.1016/j.drudis.2021.11.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/24/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023]
Abstract
The drug resistance of cancer cells is a major concern in medical oncology, resulting in the failure of chemotherapy. Ca2+ plays a pivotal role in inducing multidrug resistance in cancer cells. Calcium signaling is a critical regulator of many cancer hallmarks, such as angiogenesis, invasiveness, and migration. In this review, we describe the involvement of Ca2+ signaling and associated proteins in cancer progression and in the development of multidrug resistance in cancer cells. We also highlight the possibilities and challenges of targeting the Ca2+ channels, transporters, and pumps involved in Ca2+ signaling in cancer cells through structure-based drug design. This work will open a new therapeutic window to be used against cancer in upcoming years.
Collapse
Affiliation(s)
- Suman Panda
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India
| | - Oishika Chatterjee
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India
| | - Laboni Roy
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India
| | - Subhrangsu Chatterjee
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India.
| |
Collapse
|
11
|
Izquierdo C, Martín-Martínez M, Gómez-Monterrey I, González-Muñiz R. TRPM8 Channels: Advances in Structural Studies and Pharmacological Modulation. Int J Mol Sci 2021; 22:ijms22168502. [PMID: 34445208 PMCID: PMC8395166 DOI: 10.3390/ijms22168502] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
The transient receptor potential melastatin subtype 8 (TRPM8) is a cold sensor in humans, activated by low temperatures (>10, <28 °C), but also a polymodal ion channel, stimulated by voltage, pressure, cooling compounds (menthol, icilin), and hyperosmolarity. An increased number of experimental results indicate the implication of TRPM8 channels in cold thermal transduction and pain detection, transmission, and maintenance in different tissues and organs. These channels also have a repercussion on different kinds of life-threatening tumors and other pathologies, which include urinary and respiratory tract dysfunctions, dry eye disease, and obesity. This compendium firstly covers newly described papers on the expression of TRPM8 channels and their correlation with pathological states. An overview on the structural knowledge, after cryo-electron microscopy success in solving different TRPM8 structures, as well as some insights obtained from mutagenesis studies, will follow. Most recently described families of TRPM8 modulators are also covered, along with a section of molecules that have reached clinical trials. To finalize, authors provide an outline of the potential prospects in the TRPM8 field.
Collapse
Affiliation(s)
- Carolina Izquierdo
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
- Programa de Doctorado en Química Orgánica, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Mercedes Martín-Martínez
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
| | - Isabel Gómez-Monterrey
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
- Correspondence: (I.G.-M.); (R.G.-M.)
| | - Rosario González-Muñiz
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
- Correspondence: (I.G.-M.); (R.G.-M.)
| |
Collapse
|
12
|
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. [PMID: 34175300 DOI: 10.1016/j.bcp.2021.114664] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
Transient receptor potential melastatin (TRPM) channels are members of the transient receptor potential (TRP) channels, a family of evolutionarily conserved integral membrane proteins. TRPM channels are nonselective cation channels, mediating the influx of various ions including Ca2+, Na+ and Zn2+. The function of TRPM channels is vital for cell proliferation, cell development and cell death. Cell death is a key procedure during embryonic development, organism homeostasis, aging and disease. The category of cell death modalities, beyond the traditionally defined concepts of necrosis, autophagy, and apoptosis, were extended with the discovery of pyroptosis, necroptosis and ferroptosis. As upstream signaling regulators of cell death, TRPM channels have been involved inrelevant pathologies. In this review, we introduced several cell death modalities, then summarized the contribution of TRPM channels (especially TRPM2 and TRPM7) to different cell death modalities and discussed the underlying regulatory mechanisms. Our work highlighted the possibility of TRPM channels as potential therapeutic targets in cell death-related diseases.
Collapse
Affiliation(s)
- Ruixue Shi
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yu Fu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Dongyi Zhao
- The University of Tokyo, Department of Pharmaceutical Science, 1130033, Japan
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, 92215, Poland.
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
13
|
Nomura D, Abe R, Tsukimoto M. Involvement of TRPM8 Channel in Radiation-Induced DNA Damage Repair Mechanism Contributing to Radioresistance of B16 Melanoma. Biol Pharm Bull 2021; 44:642-652. [PMID: 33658452 DOI: 10.1248/bpb.b20-00934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Radiation is an effective cancer treatment, but cancer cells can acquire radioresistance, which is associated with increased DNA damage response and enhanced proliferative capacity, and therefore, it is important to understand the intracellular biochemical responses to γ-irradiation. The transient receptor potential melastatin 8 (TRPM8) channel plays roles in the development and progression of tumors, but it is unclear whether it is involved in the DNA damage response induced by γ-irradiation. Here, we show that a TRPM8 channel inhibitor suppresses the DNA damage response (phosphorylated histone variant H2AX-p53-binding protein 1 (γH2AX-53BP1) focus formation) and colony formation of B16 melanoma cells. Furthermore, the TRPM8 channel-specific agonist WS-12 enhanced the DNA damage response and increased the survival fraction after γ-irradiation. We found that the TRPM8 channel inhibitor enhanced G2/M phase arrest after γ-irradiation. Phosphorylation of ataxia telangiectasia mutated and p53, which both contribute to the DNA damage response was also suppressed after γ-irradiation. In addition, the TRPM8 channel inhibitor enhanced the γ-irradiation-induced suppression of tumor growth in vivo. We conclude that the TRPM8 channel is involved in radiation-induced DNA damage repair and contributes to the radioresistance of B16 melanoma cells. TRPM8 channel inhibitors might be clinically useful as radiosensitizers to enhance radiation therapy of melanoma.
Collapse
Affiliation(s)
- Daichi Nomura
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Ryo Abe
- Research Institute for Biomedical Sciences, Tokyo University of Science.,Strategic Innovation and Research Center, Teikyo University
| | - Mitsutoshi Tsukimoto
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| |
Collapse
|
14
|
Lässig F, Klann A, Bekeschus S, Lendeckel U, Wolke C. Expression of canonical transient receptor potential channels in U-2 OS and MNNG-HOS osteosarcoma cell lines. Oncol Lett 2021; 21:307. [PMID: 33732383 DOI: 10.3892/ol.2021.12568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/04/2021] [Indexed: 11/06/2022] Open
Abstract
In U-2 OS and MNNG-HOS osteosarcoma cells, small interfering RNA-mediated knockdown of the angiotensin-(1-7) receptor, Mas, increases cell proliferation. Whether alterations in canonical transient receptor potential channels (TRPC) expression contribute to this effect is not clear. In the present study, a basic description of TRPC subtype expression in osteosarcoma cell lines was provided. The pharmacological modulators of the angiotensin-(1-7) receptor, Mas, AVE0991 (agonist), or D-Ala7-Ang-(1-7) (antagonist) were applied to elucidate a possible role of Mas in the regulation of TRPC mRNA levels. The contribution of other G-protein coupled receptors (GPCR) or receptor tyrosine kinases to TRCP expression was studied by applying the selective pharmacological blockers of either PI3 kinase or MEK/Erk1/2 signaling, Ly294002 and PD98059. AVE0991 and D-Ala7-Ang-(1-7) exhibited no or marginal effects on TRPC mRNA expression. Ly294002 provoked a 9.6- and 5.9-fold increase in the amounts of TRPC5 mRNA in MNNG-HOS and U-2 OS cells, respectively. Additionally, Ly294002 increased TRPC6 mRNA levels; however, it had no effect on TRPCs 1, 3 and 4. Administration of PD98059 increased the amounts of TRPC6 and TRPC4 ~2-fold. In conclusion, the present study demonstrated that Mas-dependent alterations in osteosarcoma cell line proliferation were not mediated by any changes in TRPC subtype gene expression. The data shows in principle, and consistent with the literature, that the signaling pathways examined can regulate the expression of TRPCs at the mRNA level. Therefore, direct and signaling pathway-specific pharmacological targeting of TRPC subtypes may represent an option for improving the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Florian Lässig
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Anja Klann
- Institute of Forensic Medicine, University Medicine Greifswald, D-17489 Greifswald, Germany
| | - Sander Bekeschus
- Zentrum für Innovationskompetenz (ZIK) plasmatis, Leibniz Institute for Plasma Science and Technology (INP), D-17489 Greifswald, Germany
| | - Uwe Lendeckel
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Carmen Wolke
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, D-17475 Greifswald, Germany
| |
Collapse
|
15
|
Xu Q, Kong N, Zhang J, Bai N, Bi J, Li W. Expression of transient receptor potential cation channel subfamily M member 8 in gastric cancer and its clinical significance. Exp Ther Med 2021; 21:377. [PMID: 33680099 PMCID: PMC7918222 DOI: 10.3892/etm.2021.9808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 06/02/2020] [Indexed: 11/29/2022] Open
Abstract
Transient receptor potential cation channel subfamily M member (TRPM8) is abnormally expressed in many malignant tumors, such as breast cancer and pancreatic cancer, but its expression in gastric cancer (GC) has remained unclear. The present study aimed to detect TRPM8 expression and to explore its clinical significance in GC. Western blotting and immunohistochemistry were used to detect the protein expression of TRPM8 in 134 pairs of GC and adjacent healthy tissues. The association of TRMP8 with the 5-year overall survival rate of patients with GC was assessed using a Cox regression model. TRPM8 protein expression was significantly elevated (P<0.05) in gastric tumor cells (SUN-1, AGS, SNU-5 and NCI-N87) and was significantly associated with tumor diameter (P=0.003), Tumor-Node-Metastasis stage (P=0.003), lymph node metastasis (P=0.001) and cancer cell remote metastasis (P=0.010) in patients with GC. The expression of TRPM8 protein was significantly higher in GC patients with a tumor diameter of ≥2.5 cm. Additionally, TRPM8 protein expression in patients with metastases was significantly higher compared with patients without metastasis. Cox regression analysis revealed that TRPM8 protein expression was an independent risk factor for prognosis (odds ratio, 1.625; 95% CI=0.552-3.128) in patients with GC. In addition, the 5-year overall survival rate of patients with high expression of TRPM8 protein (64.44%) in GC was significantly lower compared with patients with low expression (12.36%). TRPM8 was highly expressed in GC tissues and may promote GC cell proliferation and metastasis in vivo.
Collapse
Affiliation(s)
- Qiqi Xu
- Department of General Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Na Kong
- Department of General Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Jun Zhang
- Department of General Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Nan Bai
- Department of General Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Jingtao Bi
- Department of General Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Wendong Li
- Department of General Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| |
Collapse
|
16
|
Liu JJ, Li LZ, Xu P. Upregulation of TRPM8 can promote the colon cancer liver metastasis through mediating Akt/GSK-3 signal pathway. Biotechnol Appl Biochem 2021; 69:230-239. [PMID: 33432591 DOI: 10.1002/bab.2102] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/06/2021] [Indexed: 01/06/2023]
Abstract
This study aims to clarify the function of transient receptor potential melastatin 8 (TRPM8) in colon cancer liver metastasis. First, TRPM8 expression was determined by Western blotting in colon cancer patients with/without liver metastasis. Second, colon cancer cells were grouped into Mock, siCON, and siTRPM8 groups. Then, a series of in vitro experiments were conducted. Last, CT26 cells were used to construct colon cancer liver metastasis models on mice in vivo, followed by comparison of liver metastasis and determination of AKT/glycogen synthase kinase-3β (GSK-3β) pathway. Consequently, TRPM8 was upregulated in both colon cancer patients with/without liver metastasis, especially in those with metastasis. Compared with Mock and siCON groups, cells in siTRPM8 group demonstrated significant decreases in clone numbers, cell invasion, and migration; and obvious downregulations of p-AKT/AKT, p-GSK3β/GSK3β, Snail, and Vimentin, with an upregulation of E-cadherin. For in vivo experiments, a sharp decrease was observed in metastatic liver of mice in siTRPM8 group, with significant downregulations of p-AKT/AKT, p-GSK3β/GSK3β, Snail, and Vimentin and an upregulation of E-cadherin, as compared with Mock and siCON groups. Thus, TRPM8 was upregulated in colon cancer patients with liver metastasis, and silencing TRPM8 may suppress the progression and epithelial-mesenchymal transition of colon cancer cells to block its liver metastasis possibly by inhibiting AKT/GSK-3β pathway.
Collapse
Affiliation(s)
- Jia-Jun Liu
- Department of Gastroenterology, Shiyan Hospital of Traditional Chinese Medicine, Shiyan, Hubei Province, People's Republic of China
| | - Long-Zhu Li
- Department of Gastroenterology, Shiyan Hospital of Traditional Chinese Medicine, Shiyan, Hubei Province, People's Republic of China
| | - Peng Xu
- Department of Gastroenterology, Shiyan Hospital of Traditional Chinese Medicine, Shiyan, Hubei Province, People's Republic of China
| |
Collapse
|
17
|
Alharbi A, Zhang Y, Parrington J. Deciphering the Role of Ca 2+ Signalling in Cancer Metastasis: From the Bench to the Bedside. Cancers (Basel) 2021; 13:E179. [PMID: 33430230 PMCID: PMC7825727 DOI: 10.3390/cancers13020179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/01/2021] [Accepted: 01/03/2021] [Indexed: 01/03/2023] Open
Abstract
Metastatic cancer is one of the major causes of cancer-related mortalities. Metastasis is a complex, multi-process phenomenon, and a hallmark of cancer. Calcium (Ca2+) is a ubiquitous secondary messenger, and it has become evident that Ca2+ signalling plays a vital role in cancer. Ca2+ homeostasis is dysregulated in physiological processes related to tumour metastasis and progression-including cellular adhesion, epithelial-mesenchymal transition, cell migration, motility, and invasion. In this review, we looked at the role of intracellular and extracellular Ca2+ signalling pathways in processes that contribute to metastasis at the local level and also their effects on cancer metastasis globally, as well as at underlying molecular mechanisms and clinical applications. Spatiotemporal Ca2+ homeostasis, in terms of oscillations or waves, is crucial for hindering tumour progression and metastasis. They are a limited number of clinical trials investigating treating patients with advanced stages of various cancer types. Ca2+ signalling may serve as a novel hallmark of cancer due to the versatility of Ca2+ signals in cells, which suggests that the modulation of specific upstream/downstream targets may be a therapeutic approach to treat cancer, particularly in patients with metastatic cancers.
Collapse
Affiliation(s)
- Abeer Alharbi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
- Pharmaceutical Sciences Department, College of Pharmacy, King Saud Bin Abdul-Aziz University for Health Sciences, Riyadh 11426, Saudi Arabia
| | - Yuxuan Zhang
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
| |
Collapse
|
18
|
Expression regulation of cold-inducible protein RBM3 by FAK/Src signaling for neuroprotection against rotenone under mild hypothermia. Biochem Biophys Res Commun 2020; 534:240-247. [PMID: 33272569 DOI: 10.1016/j.bbrc.2020.11.105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/22/2022]
Abstract
Mild hypothermia is a well-established technique for alleviating neurological injuries in clinical surgery. RNA-binding protein motif 3 (RBM3) has been identified as a crucial factor in mediating hypothermic neuroprotection, providing its induction as a promising strategy for mimicking therapeutic hypothermia. However, little is known about molecular control of RBM3 and signaling pathways affected by hypothermia. In the present study, human SH-SY5Y neuroblastoma cells were used as a neural cell model. Screening of signaling pathways showed that cold exposure led to inactivation of ERK and AMPK pathways, and activation of FAK and PLCγ pathways, with activities of p38, JNK and AKT pathways moderately changed. Next, various small molecule inhibitors specific to these signaling pathways were applied. Interestingly, only FAK-specific inhibitor exhibited a significant inhibitory effect on hypothermia-induced RBM3 gene transcription and protein expression. Likewise, FAK silencing using siRNA technique significantly abrogated the induction of RBM3 by hypothermia. Moreover, FAK inhibition accounted for an inactivation of Src, a known kinase downstream of FAK. Next, either the silencing of Src by siRNA or its inactivation by a chemical inhibitor, strongly blocked the induction of RBM3 by cooling. Notably, in HEK293 and PC12 cells, FAK/Src activation was also shown to be indispensable for hypothermia-stimulated RBM3 expression. Lastly, the CCK8 and Western blot assays showed that both FAK/Src inacitivation and their knockdown substantially abrogate the neuroprotective effects of mild hypothermia against rotenone in SH-SY5Y cells. These data suggest that FAK/Src signaling axis regulates the transcription of Rbm3 gene and mediates neuroprotective effects of mild hypothermia.
Collapse
|
19
|
Capatina AL, Lagos D, Brackenbury WJ. Targeting Ion Channels for Cancer Treatment: Current Progress and Future Challenges. Rev Physiol Biochem Pharmacol 2020; 183:1-43. [PMID: 32865696 DOI: 10.1007/112_2020_46] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ion channels are key regulators of cancer cell pathophysiology. They contribute to a variety of processes such as maintenance of cellular osmolarity and membrane potential, motility (via interactions with the cytoskeleton), invasion, signal transduction, transcriptional activity and cell cycle progression, leading to tumour progression and metastasis. Ion channels thus represent promising targets for cancer therapy. Ion channels are attractive targets because many of them are expressed at the plasma membrane and a broad range of existing inhibitors are already in clinical use for other indications. However, many of the ion channels identified in cancer cells are also active in healthy normal cells, so there is a risk that certain blockers may have off-target effects on normal physiological function. This review describes recent research advances into ion channel inhibitors as anticancer therapeutics. A growing body of evidence suggests that a range of existing and novel Na+, K+, Ca2+ and Cl- channel inhibitors may be effective for suppressing cancer cell proliferation, migration and invasion, as well as enhancing apoptosis, leading to suppression of tumour growth and metastasis, either alone or in combination with standard-of-care therapies. The majority of evidence to date is based on preclinical in vitro and in vivo studies, although there are several examples of ion channel-targeting strategies now reaching early phase clinical trials. Given the strong links between ion channel function and regulation of tumour growth, metastasis and chemotherapy resistance, it is likely that further work in this area will facilitate the development of new therapeutic approaches which will reach the clinic in the future.
Collapse
Affiliation(s)
| | - Dimitris Lagos
- Hull York Medical School, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - William J Brackenbury
- Department of Biology, University of York, York, UK.
- York Biomedical Research Institute, University of York, York, UK.
| |
Collapse
|
20
|
Liu Y, Mikrani R, He Y, Faran Ashraf Baig MM, Abbas M, Naveed M, Tang M, Zhang Q, Li C, Zhou X. TRPM8 channels: A review of distribution and clinical role. Eur J Pharmacol 2020; 882:173312. [PMID: 32610057 DOI: 10.1016/j.ejphar.2020.173312] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/10/2020] [Accepted: 06/23/2020] [Indexed: 12/15/2022]
Abstract
Ion channels are important therapeutic targets due to their plethoric involvement in physiological and pathological consequences. The transient receptor potential cation channel subfamily M member 8 (TRPM8) is a nonselective cation channel that controls Ca2+ homeostasis. It has been proposed to be the predominant thermoreceptor for cellular and behavioral responses to cold stimuli in the transient receptor potential (TRP) channel subfamilies and exploited so far to reach the clinical-stage of drug development. TRPM8 channels can be found in multiple organs and tissues, regulating several important processes such as cell proliferation, migration and apoptosis, inflammatory reactions, immunomodulatory effects, pain, and vascular muscle tension. The related disorders have been expanded to new fields ranging from cancer and migraine to dry eye disease, pruritus, irritable bowel syndrome (IBS), and chronic cough. This review is aimed to summarize the distribution of TRPM8 and disorders related to it from a clinical perspective, so as to broaden the scope of knowledge of researchers to conduct more studies on this subject.
Collapse
Affiliation(s)
- Yuqian Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, 211198, PR China
| | - Reyaj Mikrani
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, 211198, PR China
| | - Yanjun He
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, 211198, PR China
| | - Mirza Muhammad Faran Ashraf Baig
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Muhammad Abbas
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Muhammad Naveed
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Meng Tang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, 211198, PR China
| | - Qin Zhang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, 211198, PR China
| | - Cuican Li
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, 211198, PR China
| | - Xiaohui Zhou
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, 211198, PR China; Department of Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu Province, 210017, PR China; Department of Surgery, Nanjing Shuiximen Hospital, Jiangsu Province, 210017, PR China.
| |
Collapse
|
21
|
Almasi S, El Hiani Y. Exploring the Therapeutic Potential of Membrane Transport Proteins: Focus on Cancer and Chemoresistance. Cancers (Basel) 2020; 12:cancers12061624. [PMID: 32575381 PMCID: PMC7353007 DOI: 10.3390/cancers12061624] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Improving the therapeutic efficacy of conventional anticancer drugs represents the best hope for cancer treatment. However, the shortage of druggable targets and the increasing development of anticancer drug resistance remain significant problems. Recently, membrane transport proteins have emerged as novel therapeutic targets for cancer treatment. These proteins are essential for a plethora of cell functions ranging from cell homeostasis to clinical drug toxicity. Furthermore, their association with carcinogenesis and chemoresistance has opened new vistas for pharmacology-based cancer research. This review provides a comprehensive update of our current knowledge on the functional expression profile of membrane transport proteins in cancer and chemoresistant tumours that may form the basis for new cancer treatment strategies.
Collapse
Affiliation(s)
- Shekoufeh Almasi
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON KIH 8M5, Canada;
| | - Yassine El Hiani
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Correspondence:
| |
Collapse
|
22
|
Effects of curcumin complexes on MDA‑MB‑231 breast cancer cell proliferation. Int J Oncol 2020; 57:445-455. [PMID: 32626932 PMCID: PMC7307592 DOI: 10.3892/ijo.2020.5065] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Curcumin displays anticancer properties; however, some issues with the drug delivery mode limit its therapeutic use. Although reformulation and derivatization of curcumin have improved its bioavailability, curcumin derivatives may not retain the same anticancer properties as the parent compound. The present study investigated the anticancer properties of two curcumin complexes, the iron‑curcumin [Fe(Cur)3] and boron‑curcumin [B(Cur)2] complexes, in the MDA‑MB‑231 breast cancer cell line. The cellular localization of curcumin, B(Cur)2 and Fe(Cur)3 was determined by fluorescence microscopy. Cell proliferation, migration and invasion were also analysed. Furthermore, apoptosis‑associated proteins were detected by using a proteome profiler array, and ion channel gene expression was analysed by reverse transcription‑quantitative PCR. The results demonstrated that the three compounds were localized in the perinuclear and cytoplasmic regions of the cell, and displayed cytotoxicity with IC50 values of 25, 35 and 8 µM for curcumin, B(Cur)2 and Fe(Cur)3, respectively. In addition, the three compounds inhibited cell invasion, whereas only curcumin and B(Cur)2 inhibited cell migration. Furthermore, cell exposure to curcumin resulted in an increase in the relative expression of the two key proapoptotic proteins, cytochrome c and cleaved caspase‑3, as well as the antiapoptotic protein haem oxygenase‑1. In addition, curcumin increased the expression levels of the voltage‑gated potassium channels Kv2.1 and Kv3.2. Similarly, the expression levels of the chloride channel bestrophin‑1 and the calcium channel coding gene calcium voltage‑gated channel auxiliary subunit γ4 were increased following exposure to curcumin. Taken together, these results indicated that Fe(Cur)3 and B(Cur)2 may display similar anticancer properties as curcumin, suggesting that chemical complexation may be considered as a strategy for improving the potency of curcumin in the treatment of breast cancer.
Collapse
|
23
|
Santoni G, Morelli MB, Marinelli O, Nabissi M, Santoni M, Amantini C. Calcium Signaling and the Regulation of Chemosensitivity in Cancer Cells: Role of the Transient Receptor Potential Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:505-517. [PMID: 31646523 DOI: 10.1007/978-3-030-12457-1_20] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer cells acquire the ability to modify the calcium signaling network by altering the expression and functions of cation channels, pumps or transporters. Calcium signaling pathways are involved in proliferation, angiogenesis, invasion, immune evasion, disruption of cell death pathways, ECM remodelling, epithelial-mesenchymal transition (EMT) and drug resistance. Among cation channels, a pivotal role is played by the Transient Receptor Potential non-selective cation-permeable receptors localized in plasma membrane, endoplasmic reticulum, mitochondria and lysosomes. Several findings indicate that the dysregulation in calcium signaling induced by TRP channels is responsible for cancer growth, metastasis and chemoresistance. Drug resistance represents a major limitation in the application of current therapeutic regimens and several efforts are spent to overcome it. Here we describe the ability of Transient Receptor Potential Channels to modify, by altering the intracellular calcium influx, the cancer cell sensitivity to chemotherapeutic drugs.
Collapse
Affiliation(s)
- Giorgio Santoni
- School of Pharmacy, Immunopathology and Molecular Medicine Laboratory, University of Camerino, Camerino, Italy
| | - Maria Beatrice Morelli
- School of Pharmacy, Immunopathology and Molecular Medicine Laboratory, University of Camerino, Camerino, Italy.,School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Oliviero Marinelli
- School of Pharmacy, Immunopathology and Molecular Medicine Laboratory, University of Camerino, Camerino, Italy.,School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Massimo Nabissi
- School of Pharmacy, Immunopathology and Molecular Medicine Laboratory, University of Camerino, Camerino, Italy
| | - Matteo Santoni
- Clinic and Oncology Unit, Macerata Hospital, Macerata, Italy
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy.
| |
Collapse
|
24
|
Transient Receptor Potential Cation Channels in Cancer Therapy. Med Sci (Basel) 2019; 7:medsci7120108. [PMID: 31801263 PMCID: PMC6950741 DOI: 10.3390/medsci7120108] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/08/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023] Open
Abstract
In mammals, the transient receptor potential (TRP) channels family consists of six different families, namely TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPML (mucolipin), TRPP (polycystin), and TRPA (ankyrin), that are strictly connected with cancer cell proliferation, differentiation, cell death, angiogenesis, migration, and invasion. Changes in TRP channels' expression and function have been found to regulate cell proliferation and resistance or sensitivity of cancer cells to apoptotic-induced cell death, resulting in cancer-promoting effects or resistance to chemotherapy treatments. This review summarizes the data reported so far on the effect of targeting TRP channels in different types of cancer by using multiple TRP-specific agonists, antagonists alone, or in combination with classic chemotherapeutic agents, microRNA specifically targeting the TRP channels, and so forth, and the in vitro and in vivo feasibility evaluated in experimental models and in cancer patients. Considerable efforts have been made to fight cancer cells, and therapies targeting TRP channels seem to be the most promising strategy. However, more in-depth investigations are required to completely understand the role of TRP channels in cancer in order to design new, more specific, and valuable pharmacological tools.
Collapse
|
25
|
Aung T, Asam C, Haerteis S. Ion channels in sarcoma: pathophysiology and treatment options. Pflugers Arch 2019; 471:1163-1171. [PMID: 31377822 DOI: 10.1007/s00424-019-02299-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 12/21/2022]
Abstract
Sarcomas are characterized by aggressive growth and a high metastasis potentially leading in most cases to a lethal outcome. These malignant tumors of the connective tissue have a high heterogeneity with numerous genetic mutations resulting in more than 100 types of sarcoma that can be grouped into two main kinds: soft tissue sarcoma and bone sarcoma. Sarcomas are often diagnosed at late disease stage, whereas a guaranteed diagnosis of the sarcoma type is fundamental for successful therapy. However, there is no appropriate therapy available. Therefore, the need for new therapies, which prolong survival and improve quality of life, is high. In the last two decades, the role of ion channels in cancer has emerged. Ion channels seem to be an ideal target for anti-tumor therapies. However, different cancer types have their own altered ion channel pattern, and the knowledge about the tumor-associated ion channel expression is fundamental. Here, we focus on the role of different ion channels in sarcoma, their pathophysiology, and possible treatment options.
Collapse
Affiliation(s)
- Thiha Aung
- Abteilung für Plastische, Hand- und Wiederherstellungschirurgie, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Claudia Asam
- Lehrstuhl für Molekulare und Zelluläre Anatomie, Universität Regensburg, 93053, Regensburg, Germany
| | - Silke Haerteis
- Lehrstuhl für Molekulare und Zelluläre Anatomie, Universität Regensburg, 93053, Regensburg, Germany.
| |
Collapse
|
26
|
GSK3 regulates hair cell fate in the developing mammalian cochlea. Dev Biol 2019; 453:191-205. [PMID: 31185200 DOI: 10.1016/j.ydbio.2019.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 06/07/2019] [Accepted: 06/07/2019] [Indexed: 12/27/2022]
Abstract
The development of asymmetric patterns along biologically relevant axes is a hallmark of many vertebrate organs or structures. One example is the sensory epithelium of the mammalian auditory system. Two distinct types of mechanosensory hair cells (inner and outer) and at least six types of associated supporting cells are precisely and asymmetrically arrayed along the radial (medial-lateral) axis of the cochlear spiral. Immunolabeling of developing cochleae indicates differential expression of Glycogen synthase kinase 3β (GSK3β) along the same axis. To determine whether GSK3β plays a role in specification of cell fates along the medial-lateral axis, GSK3 activity was blocked pharmacologically in cochlear explants. Results indicate significant changes in both the number of hair cells and in the specification of hair cell phenotypes. The overall number of inner hair cells increased as a result of both a shift in the medial boundary between sensory and non-sensory regions of the cochlea and a change in the specification of inner and outer hair cell phenotypes. Previous studies have inhibited GSK3 as a method to examine effects of canonical Wnt signaling. However, quantification of changes in Wnt pathway target genes in GSK3-inhibited cochleae, and treatment with more specific Wnt agonists, indicated that the Wnt pathway is not activated. Instead, expression of Bmp4 in a population of GSK3β-expressing cells was shown to be down-regulated. Finally, addition of BMP4 to GSK3-inhibited cochleae achieved a partial rescue of the hair cell phenotype. These results demonstrate a role for GSK3β in the specification of cellular identities along the medial-lateral axis of the cochlea and provide evidence for a positive role for GSK3β in the expression of Bmp4.
Collapse
|
27
|
Ion Channels: New Actors Playing in Chemotherapeutic Resistance. Cancers (Basel) 2019; 11:cancers11030376. [PMID: 30884858 PMCID: PMC6468599 DOI: 10.3390/cancers11030376] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/07/2019] [Accepted: 03/12/2019] [Indexed: 01/23/2023] Open
Abstract
In the battle against cancer cells, therapeutic modalities are drastically limited by intrinsic or acquired drug resistance. Resistance to therapy is not only common, but expected: if systemic agents used for cancer treatment are usually active at the beginning of therapy (i.e., 90% of primary breast cancers and 50% of metastases), about 30% of patients with early-stage breast cancer will have recurrent disease. Altered expression of ion channels is now considered as one of the hallmarks of cancer, and several ion channels have been linked to cancer cell resistance. While ion channels have been associated with cell death, apoptosis and even chemoresistance since the late 80s, the molecular mechanisms linking ion channel expression and/or function with chemotherapy have mostly emerged in the last ten years. In this review, we will highlight the relationships between ion channels and resistance to chemotherapy, with a special emphasis on the underlying molecular mechanisms.
Collapse
|
28
|
Hantute-Ghesquier A, Haustrate A, Prevarskaya N, Lehen'kyi V. TRPM Family Channels in Cancer. Pharmaceuticals (Basel) 2018; 11:ph11020058. [PMID: 29875336 PMCID: PMC6027338 DOI: 10.3390/ph11020058] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/28/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022] Open
Abstract
Members of the TRPM ("Melastatin") family fall into the subclass of the TRP channels having varying permeability to Ca2+ and Mg2+, with three members of the TRPM family being chanzymes, which contain C-terminal enzyme domains. The role of different TRPM members has been shown in various cancers such as prostate cancer for mostly TRPM8 and TRPM2, breast cancer for mostly TRPM2 and TRPM7, and pancreatic cancer for TRPM2/7/8 channels. The role of TRPM5 channels has been shown in lung cancer, TRPM1 in melanoma, and TRPM4 channel in prostate cancer as well. Thus, the TRPM family of channels may represent an appealing target for the anticancer therapy.
Collapse
Affiliation(s)
- Aline Hantute-Ghesquier
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d'Ascq, France.
| | - Aurélien Haustrate
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d'Ascq, France.
- FONDATION ARC, 9 rue Guy Môquet 94830 Villejuif, France.
| | - Natalia Prevarskaya
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d'Ascq, France.
| | - V'yacheslav Lehen'kyi
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d'Ascq, France.
- FONDATION ARC, 9 rue Guy Môquet 94830 Villejuif, France.
| |
Collapse
|
29
|
Du JD, Zheng X, Chen YL, Huang ZQ, Cai SW, Jiao HB, Zhu ZM, Hu B. Elevated Transient Receptor Potential Melastatin 8 (TRPM8) Expression Is Correlated with Poor Prognosis in Pancreatic Cancer. Med Sci Monit 2018; 24:3720-3725. [PMID: 29860264 PMCID: PMC6015479 DOI: 10.12659/msm.909968] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The transient receptor potential melastatin 8 (TRPM8) was found to be expressed abnormally in a variety of tumors and is associated with unfavorable prognosis in human cancers. However, its clinical significance in pancreatic cancer (PC) is mostly unknown. MATERIAL AND METHODS qRT-PCR was performed to measure the expression of TRPM8 in 110 pairs of PC tissues and the adjacent non-cancerous tissues. The association of TRPM8 expression with the clinical characters of PC patients was analyzed using the chi-square test. Furthermore, the prognostic value of TRPM8 was determined with Kaplan-Meier survival curve and Cox regression analysis. RESULTS We found that the expression level of TRPM8 was significantly elevated in PC tissues compared to the non-cancerous controls (P<0.001). In addition, a close relationship was observed between elevated TRPM8 expression with large tumor size (P=0.001), advanced TNM (P=0.013), and distant metastasis (P=0.034). Survival analysis suggested that patients with high TRPM8 expression has worse OS (P=0.001) and DFS (P<0.001) than those with low TRPM8 expression. Moreover, TRPM8 was confirmed as a valuable prognostic biomarker for OS (HR=1.913; 95% CI: 1.020-3.589; P=0.043) or DFS (HR=2.374; 95% CI: 1.269-4.443; P=0.007) of PC patients. CONCLUSIONS This study shows that TRPM8 expression is significantly up-regulated in PC and it might be a useful prognostic factor for patients with PC.
Collapse
Affiliation(s)
- Jun-Dong Du
- Department of Heptapobiliary Surgery, First Affiliated Hospital to General Hospital of the PLA, Beijing, China (mainland)
| | - Xi Zheng
- Department of Division Three for Senior Officers, First Affiliated Hospital to General Hospital of the PLA, Beijing, China (mainland)
| | - Yong-Liang Chen
- Department of Heptapobiliary Surgery, General Hospital of PLA, Beijing, China (mainland)
| | - Zhi-Qiang Huang
- Department of Heptapobiliary Surgery, General Hospital of PLA, Beijing, China (mainland)
| | - Shou-Wang Cai
- Department of Heptapobiliary Surgery, General Hospital of PLA, Beijing , China (mainland)
| | - Hua-Bo Jiao
- Department of Heptapobiliary Surgery, First Affiliated Hospital to General Hospital of the PLA, Beijing, China (mainland)
| | - Zi-Man Zhu
- Department of Heptapobiliary Surgery, First Affiliated Hospital to General Hospital of the PLA, Beijing, China (mainland)
| | - Bin Hu
- Department of Heptapobiliary Surgery, First Affiliated Hospital to General Hospital of the PLA, Beijing, China (mainland)
| |
Collapse
|
30
|
Decreased TRPM7 inhibits activities and induces apoptosis of bladder cancer cells via ERK1/2 pathway. Oncotarget 2018; 7:72941-72960. [PMID: 27662662 PMCID: PMC5341955 DOI: 10.18632/oncotarget.12146] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 09/13/2016] [Indexed: 12/15/2022] Open
Abstract
Transient receptor potential melastatin 7 (TRPM7) functions as a Mg2+/Ca2+-permeable channel fused with a kinase domain and regulates various physical processes and diseases. However, its effects on pathogenesis of human bladder cancer (BCa) has not been clarified yet. Our microarray analysis has suggested that calcium signaling pathway is connected with bladder cancer via MAPK pathway. Therefore, we aim to investigate the mechanism of TRPM7 in BCa tumorigenesis by using BCa tissues compared with normal bladder epithelium tissues, as well as using distinct BCa cell lines (EJ, 5637 and T24). We observed increased TRPM7 expression and dysregulation of proteins involved in Epithelial-Mesenchymal Transition (EMT) in BCa tissues. Moreover, knockdown of TRPM7 in BCa cells reversed the EMT status, accompanied by increase of reactive oxygen species (ROS). Furthermore, TRPM7 deficiency could inhibit BCa cell proliferation, migration and invasion, as well as induce p-ERK1/2 and suppress PI3K/AKT at the protein level. Downregulation of TRPM7 promoted cell cycle arrest at G0/G1 phase and apoptosis in vitro, which could be recovered by pre-treatment with U0126 to deactivate ERK1/2, suggesting a close correlation between TRPM7 and the MAPK signaling pathway. Furthermore, a NOD/SCID mouse model transplanted using the BCa cells was established, revealing delayed tumor growth by reduced protein activity and mRNA transcription of TRPM7 in vivo. Our results suggested TRPM7 might be essential for BCa tumorigenesis by interfering BCa cell proliferation, motility and apoptosis.
Collapse
|
31
|
Fliniaux I, Germain E, Farfariello V, Prevarskaya N. TRPs and Ca2+ in cell death and survival. Cell Calcium 2018; 69:4-18. [DOI: 10.1016/j.ceca.2017.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 10/19/2022]
|
32
|
Li C, Li J, Xiong X, Liu Y, Lv Y, Qin S, Liu D, Wei R, Ruan X, Zhang J, Xu L, Wang X, Chen J, Zhang Y, Zheng L. TRPM8 activation improves energy expenditure in skeletal muscle and exercise endurance in mice. Gene 2018; 641:111-116. [DOI: 10.1016/j.gene.2017.10.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/06/2017] [Accepted: 10/16/2017] [Indexed: 10/18/2022]
|
33
|
TRPM8 is required for survival and radioresistance of glioblastoma cells. Oncotarget 2017; 8:95896-95913. [PMID: 29221175 PMCID: PMC5707069 DOI: 10.18632/oncotarget.21436] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 08/25/2017] [Indexed: 01/29/2023] Open
Abstract
TRPM8 is a Ca2+-permeable nonselective cation channel belonging to the melastatin sub-group of the transient receptor potential (TRP) family. TRPM8 is aberrantly overexpressed in a variety of tumor entities including glioblastoma multiforme where it reportedly contributes to tumor invasion. The present study aimed to disclose further functions of TRPM8 in glioma biology in particular upon cell injury by ionizing radiation. To this end, TCGA data base was queried to expose the TRPM8 mRNA abundance in human glioblastoma specimens and immunoblotting was performed to analyze the TRPM8 protein abundance in primary cultures of human glioblastoma. Moreover, human glioblastoma cell lines were irradiated with 6 MV photons and TRPM8 channels were targeted pharmacologically or by RNA interference. TRPM8 abundance, Ca2+ signaling and resulting K+ channel activity, chemotaxis, cell migration, clonogenic survival, DNA repair, apoptotic cell death, and cell cycle control were determined by qRT-PCR, fura-2 Ca2+ imaging, patch-clamp recording, transfilter migration assay, wound healing assay, colony formation assay, immunohistology, flow cytometry, and immunoblotting. As a result, human glioblastoma upregulates TRPM8 channels to variable extent. TRPM8 inhibition or knockdown slowed down cell migration and chemotaxis, attenuated DNA repair and clonogenic survival, triggered apoptotic cell death, impaired cell cycle and radiosensitized glioblastoma cells. Mechanistically, ionizing radiation activated and upregulated TRPM8-mediated Ca2+ signaling that interfered with cell cycle control probably via CaMKII, cdc25C and cdc2. Combined, our data suggest that TRPM8 channels contribute to spreading, survival and radioresistance of human glioblastoma and, therefore, might represent a promising target in future anti-glioblastoma therapy.
Collapse
|
34
|
Chen L, Cao R, Wang G, Yuan L, Qian G, Guo Z, Wu CL, Wang X, Xiao Y. Downregulation of TRPM7 suppressed migration and invasion by regulating epithelial–mesenchymal transition in prostate cancer cells. Med Oncol 2017; 34:127. [DOI: 10.1007/s12032-017-0987-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/26/2017] [Indexed: 12/24/2022]
|
35
|
Peruzzo R, Biasutto L, Szabò I, Leanza L. Impact of intracellular ion channels on cancer development and progression. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2016; 45:685-707. [PMID: 27289382 PMCID: PMC5045486 DOI: 10.1007/s00249-016-1143-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 12/13/2022]
Abstract
Cancer research is nowadays focused on the identification of possible new targets in order to try to develop new drugs for curing untreatable tumors. Ion channels have emerged as "oncogenic" proteins, since they have an aberrant expression in cancers compared to normal tissues and contribute to several hallmarks of cancer, such as metabolic re-programming, limitless proliferative potential, apoptosis-resistance, stimulation of neo-angiogenesis as well as cell migration and invasiveness. In recent years, not only the plasma membrane but also intracellular channels and transporters have arisen as oncological targets and were proposed to be associated with tumorigenesis. Therefore, the research is currently focusing on understanding the possible role of intracellular ion channels in cancer development and progression on one hand and, on the other, on developing new possible drugs able to modulate the expression and/or activity of these channels. In a few cases, the efficacy of channel-targeting drugs in reducing tumors has already been demonstrated in vivo in preclinical mouse models.
Collapse
Affiliation(s)
| | - Lucia Biasutto
- CNR Institute of Neuroscience, Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padua, Padua, Italy
- CNR Institute of Neuroscience, Padua, Italy
| | - Luigi Leanza
- Department of Biology, University of Padua, Padua, Italy.
| |
Collapse
|
36
|
High expression of TRPM8 predicts poor prognosis in patients with osteosarcoma. Oncol Lett 2016; 12:1373-1379. [PMID: 27446440 PMCID: PMC4950156 DOI: 10.3892/ol.2016.4764] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 02/24/2016] [Indexed: 12/30/2022] Open
Abstract
The transient receptor potential melastatin member 8 (TRPM8) is a newly characterized oncoprotein involved in various malignant tumors. However, its expression pattern and biological function in osteosarcoma remain unclear. The present study aimed to explore the expression and prognostic significance of TRPM8 in osteosarcoma (OS). The results revealed that the expression of TRPM8 mRNA and protein in OS tissue was significantly higher than that in paired normal bone tissue (P<0.05). Additionally, the level of TRPM8 mRNA and protein in patients with a higher clinical stage and with distant metastasis was markedly higher than in those with a lower clinical stage and no metastasis (P<0.05). Furthermore, a high TRPM8 level was closely associated with clinical stage and distant metastasis (P=0.007 and 0.030), but not associated with the patient age (P=0.481), gender (P=0.905), tumor size (P=0.429), histological subtype (P=0.207) or anatomical location (P=0.369). In addition, OS patients with high TRPM8 expression had significantly shorter overall survival (P=0.008) and disease-free survival times (P=0.008) when compared with patients with low expression of TRPM8. In Cox multivariate analysis, TRPM8 overexpression was identified to be an independent and significant prognostic factor for overall survival (P=0.040), but not for disease-free survival (P=0.051). Collectively, the present data suggest that TRPM8 may play a crucial role in the development and progression of OS, and thus may be considered as a novel molecular target for therapy in patients with OS.
Collapse
|
37
|
Lv YF, Dai H, Yan GN, Meng G, Zhang X, Guo QN. Downregulation of tumor suppressing STF cDNA 3 promotes epithelial-mesenchymal transition and tumor metastasis of osteosarcoma by the Wnt/GSK-3β/β-catenin/Snail signaling pathway. Cancer Lett 2016; 373:164-73. [PMID: 26845447 DOI: 10.1016/j.canlet.2016.01.046] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/30/2015] [Accepted: 01/26/2016] [Indexed: 01/11/2023]
Abstract
Epithelial to mesenchymal transition (EMT) has received considerable attention as a conceptual paradigm for explaining the invasive and metastatic behavior of cells during cancer progression. Our previous study showed that loss of expression of TSSC3 is positively associated with osteosarcoma malignancy and progression. However, whether TSSC3 mediates EMT in osteosarcoma is poorly understood. In the present study, we determined that TSSC3 downregulation induced cell migration and invasion ability and promoted mesenchymal transition of osteosarcoma cells by upregulating mesenchymal markers and inhibiting the epithelial markers. Furthermore, TSSC3 downregulation elicited a signaling cascade that included increased levels of Wnt3a and LRP5, inactivation of GSK-3β, accumulation of nuclear β-catenin and Snail, the augmented binding of β-catenin to TCF-4, and accordingly increased the expression of Wnt target genes (CD44, MMP7). The gene knockdown of these signaling proteins could inhibit TSSC3 downregulation-promoted EMT, migration, and invasion in osteosarcoma. Finally, TSSC3 overexpression obviously inhibited cell migration, invasion, and repressed mesenchymal phenotypes, reducing lung metastasis through GSK-3β activation. Collectively, TSSC3 downregulation promotes the EMT of osteosarcoma cells by regulating EMT markers via a signal transduction pathway that involves Snail, Wnt-β-catenin/TCF, and GSK-3β.
Collapse
Affiliation(s)
- Yang-fan Lv
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, China
| | - Huanzi Dai
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, China; Department of Nephrology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Guang-ning Yan
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, China
| | - Gang Meng
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, China; Department of Pathology, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Xi Zhang
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, China; Department of Pathology, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Qiao-nan Guo
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
38
|
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; 142:1871-81. [PMID: 26803314 DOI: 10.1007/s00432-015-2112-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 12/28/2015] [Indexed: 01/09/2023]
Abstract
Transient receptor potential (TRP) cation channel superfamily plays critical roles in variety of processes, including temperature perception, pain transduction, vasorelaxation, male fertility, and tumorigenesis. One of seven families within the TRP superfamily of ion channels, the melastatin, or TRPM family comprises a group of eight structurally and functionally diverse channels. Of all the members of TRPM subfamily, TRPM8 is the most notable one. A lot of literatures have demonstrated that transient receptor potential melastatin 8 (TRPM8) could perform a myriad of functions in vertebrates and invertebrates alike. In addition to its well-known function in cold sensation, TRPM8 has an emerging role in a variety of biological systems, including thermoregulation, cancer, bladder function, and asthma. Recent studies have shown that TRPM8 is necessary to the initiation and progression of tumors, and the aberrant expression of TRPM8 was found in varieties of tumors, such as prostate tumor, melanoma, breast adenocarcinoma, bladder cancer, and colorectal cancer, making it a novel molecular target potentially useful in the diagnosis and treatment of cancer. This review outlines our current understanding on the role of TRPM8 in occurrence and development of different kinds of tumor and also includes discussion about the regulation of TRPM8 during carcinogenesis as well as therapeutic potential of targeting TRPM8 in tumor, which may be utilized for a potential pharmacological use as a target for anti-cancer therapy.
Collapse
Affiliation(s)
- Zhaoguo Liu
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Hongyan Wu
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China.,Department of Pharmacy, Yancheng Health Vocational and Technical College, Yancheng, 224005, Jiangsu Province, China
| | - Zhonghong Wei
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Xu Wang
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Peiliang Shen
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Siliang Wang
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Aiyun Wang
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Wenxing Chen
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Yin Lu
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China. .,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
39
|
Yee NS. TRPM8 Ion Channels as Potential Cancer Biomarker and Target in Pancreatic Cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 104:127-155. [DOI: 10.1016/bs.apcsb.2016.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
40
|
Roles of TRPM8 Ion Channels in Cancer: Proliferation, Survival, and Invasion. Cancers (Basel) 2015; 7:2134-46. [PMID: 26512697 PMCID: PMC4695882 DOI: 10.3390/cancers7040882] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 11/24/2022] Open
Abstract
The goal of this article is to provide a critical review of the transient receptor potential melastatin-subfamily member 8 (TRPM8) in cancers, with an emphasis on its roles in cellular proliferation, survival, and invasion. The TRPM8 ion channels regulate Ca2+ homeostasis and function as a cellular sensor and transducer of cold temperature. Accumulating evidence has demonstrated that TRPM8 is aberrantly expressed in a variety of malignant solid tumors. Clinicopathological analysis has shown that over-expression of TRPM8 correlates with tumor progression. Experimental data have revealed important roles of TRPM8 channels in cancer cells proliferation, survival, and invasion, which appear to be dependent on the cancer type. Recent reports have begun to reveal the signaling mechanisms that mediate the biological roles of TRPM8 in tumor growth and metastasis. Determining the mechanistic roles of TRPM8 in cancer is expected to elucidate the impact of thermal and chemical stimuli on the formation and progression of neoplasms. Translational research and clinical investigation of TRPM8 in malignant diseases will help exploit these ion channels as molecular biomarkers and therapeutic targets for developing precision cancer medicine.
Collapse
|
41
|
Liu S, Chen Z. The Functional Role of PMP22 Gene in the Proliferation and Invasion of Osteosarcoma. Med Sci Monit 2015; 21:1976-82. [PMID: 26154129 PMCID: PMC4501650 DOI: 10.12659/msm.893430] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background As the most common primary bone tumor, osteosarcoma has an improved survival rates with advancement of treatment methods. A higher rate of metastasis, however, leads to the aggravation of the disease. Studies have shown that some genes, namely osteosarcoma metastasis-related genes, participate in the process of tumor metastasis. The peripheral myelin protein 22 (PMP22) gene has recently been found to be abundantly expressed in the oncogenesis of osteosarcoma. Its detailed role and function in the tumor metastasis, however, remains unknown. Material/Methods The recombinant retroviral plasmid pcDNA3.1-PMP22 was constructed and used to transfect osteosarcoma cells SOSP-M, whose cell proliferation was measured by MTT method. The formation of tumor cell colony, the cell migration and invasion were also measured. The signal transduction pathway MAPK was further analyzed by Western blotting. Results The pcDNA3.1-PMP22 plasmid was confirmed to have a 305bp PMP22 fragment by EcoRI-XhoI dual digestion. Compared to the control group, osteosarcoma cell invasion was significantly facilitated by the transfection of pcDNA3.1-PMP22 plasmid (p<0.05). The recombinant plasmid also significantly potentiated the formation of tumor cell colony and increased the migration and invasion ability of tumor cells (p<0.05 in all cases). Phosphorylated p-ERK and p-P38 were also up-regulated by vector transfection (p<0.05). Conclusions Osteosarcoma metastasis-related gene PMP22 participates in the proliferation, invasion, migration and colony formation of osteosarcoma cells possibly via the MAPK signal transduction pathway, providing evidences for further investigation of metastatic mechanism of osteosarcoma.
Collapse
Affiliation(s)
- Shuyong Liu
- Department of Hand and Foot Surgery, Jining No. 1 People's Hospital, Jining, Shandong, China (mainland)
| | - Zhiping Chen
- Department of Pediatrics, Jining No. 1 People's Hospital, Jining, Shandong, China (mainland)
| |
Collapse
|
42
|
Kondratskyi A, Kondratska K, Skryma R, Prevarskaya N. Ion channels in the regulation of apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2532-46. [PMID: 25450339 DOI: 10.1016/j.bbamem.2014.10.030] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 10/08/2014] [Accepted: 10/20/2014] [Indexed: 02/07/2023]
Abstract
Apoptosis, a type of genetically controlled cell death, is a fundamental cellular mechanism utilized by multicellular organisms for disposal of cells that are no longer needed or potentially detrimental. Given the crucial role of apoptosis in physiology, deregulation of apoptotic machinery is associated with various diseases as well as abnormalities in development. Acquired resistance to apoptosis represents the common feature of most and perhaps all types of cancer. Therefore, repairing and reactivating apoptosis represents a promising strategy to fight cancer. Accumulated evidence identifies ion channels as essential regulators of apoptosis. However, the contribution of specific ion channels to apoptosis varies greatly depending on cell type, ion channel type and intracellular localization, pathology as well as intracellular signaling pathways involved. Here we discuss the involvement of major types of ion channels in apoptosis regulation. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Artem Kondratskyi
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Kateryna Kondratska
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Roman Skryma
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Natalia Prevarskaya
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France.
| |
Collapse
|