1
|
Liu Q, Hu M, Li S, Zhang X, Zhang R, Lyu H, Xiao S, Guo D, Chen XZ, Tang J, Zhou C. TRPM channels in human cancers: regulatory mechanism and therapeutic prospects. Biomark Res 2024; 12:152. [PMID: 39633507 PMCID: PMC11616203 DOI: 10.1186/s40364-024-00699-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
The transient receptor potential melastatin (TRPM) channel family has been previously implicated in various diseases, including those related to temperature sensing, cardiovascular health, and neurodegeneration. Nowadays, increasing evidence indicates that TRPM family members also play significant roles in various types of cancers, exhibiting both pro- and anti-tumorigenic functions. They are involved in tumor cell proliferation, survival, invasion, and metastasis, serving as potential diagnostic and prognostic biomarkers for cancer. This paper begins by describing the structure and physiological functions of the TRPM family members. It then outlines their roles in several common malignancies, including pancreatic, prostate, colorectal, breast, brain cancer, and melanoma. Subsequently, we focused on investigating the specific mechanisms by which TRPM family members are involved in tumorigenesis and development from both the tumor microenvironment (TME) and intracellular signaling. TRPM channels not only transmit signals from the TME to regulate tumor cell functions, but also mediate extracellular matrix remodeling, which is conducive to the malignant transformation of tumor cells. Importantly, TRPM channels depend on the regulation of the inflow of various ions in cells, and participate in key signaling pathways involved in tumor progression, such as Wnt/β-catenin, MAPK, PI3K/AKT, p53, and autophagy. Finally, we summarize the current strategies and challenges of targeting TRPM channels in tumor treatment, and discuss the feasibility of combining targeted TRPM channel drugs with cancer immunotherapy.
Collapse
Affiliation(s)
- Qinfeng Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Mengyu Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Shi Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Xin Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Dong Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China.
| |
Collapse
|
2
|
Song C, Wang G, Liu M, Xu Z, Liang X, Ding K, Chen Y, Wang W, Lou W, Liu L. Identification of methylation driver genes for predicting the prognosis of pancreatic cancer patients based on whole-genome DNA methylation sequencing technology. Heliyon 2024; 10:e29914. [PMID: 38737285 PMCID: PMC11088258 DOI: 10.1016/j.heliyon.2024.e29914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/14/2024] Open
Abstract
This study was based on the use of whole-genome DNA methylation sequencing technology to identify DNA methylation biomarkers in tumor tissue that can predict the prognosis of patients with pancreatic cancer (PCa). TCGA database was used to download PCa-related DNA methylation and transcriptome atlas data. Methylation driver genes (MDGs) were obtained using the MethylMix package. Candidate genes in the MDGs were screened for prognostic relevance to PCa patients by univariate Cox analysis, and a prognostic risk score model was constructed based on the key MDGs. ROC curve analysis was performed to assess the accuracy of the prognostic risk score model. The effects of PIK3C2B knockdown on malignant phenotypes of PCa cells were investigated in vitro. A total of 2737 differentially expressed genes were identified, with 649 upregulated and 2088 downregulated, using 178 PCa samples and 171 normal samples. MethylMix was employed to identify 71 methylation-driven genes (47 hypermethylated and 24 hypomethylated) from 185 TCGA PCa samples. Cox regression analyses identified eight key MDGs (LEF1, ZIC3, VAV3, TBC1D4, FABP4, MAP3K5, PIK3C2B, IGF1R) associated with prognosis in PCa. Seven of them were hypermethylated, while PIK3C2B was hypomethylated. A prognostic risk prediction model was constructed based on the eight key MDGs, which was found to accurately predict the prognosis of PCa patients. In addition, the malignant phenotypes of PANC-1 cells were decreased after the knockdown of PIK3C2B. Therefore, the prognostic risk prediction model based on the eight key MDGs could accurately predict the prognosis of PCa patients.
Collapse
Affiliation(s)
- Chao Song
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200000, China
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital of Fudan University, Shanghai, 200000, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Fudan University, Qingpu Branch, Shanghai, 200000, China
| | - Ganggang Wang
- Department of Hepatobiliary Surgery, Pudong Hospital, Fudan University, Shanghai, 200000, China
| | - Mengmeng Liu
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Fudan University, Qingpu Branch, Shanghai, 200000, China
| | - Zijin Xu
- Department of General Surgery, Affiliated Zhongshan Hospital of Fudan University, Qingpu Branch, Shanghai, 200000, China
| | - Xin Liang
- CAS Key Laboratory of Nutrition, University of Chinese Academy of Sciences, Shanghai, 200000, China
| | - Kai Ding
- CAS Key Laboratory of Nutrition, University of Chinese Academy of Sciences, Shanghai, 200000, China
| | - Yu Chen
- CAS Key Laboratory of Nutrition, University of Chinese Academy of Sciences, Shanghai, 200000, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital of Fudan University, Shanghai, 200000, China
| | - Wenhui Lou
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital of Fudan University, Shanghai, 200000, China
| | - Liang Liu
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200000, China
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital of Fudan University, Shanghai, 200000, China
| |
Collapse
|
3
|
Mao J, Tian Y, Luo N. An ion channel-based prognostic model identified TRPV2 and GJB3 as immunotherapy determinants in pancreatic cancer. Heliyon 2024; 10:e27301. [PMID: 38560261 PMCID: PMC10979059 DOI: 10.1016/j.heliyon.2024.e27301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Background Less than 10% of people who have pancreatic ductal adenocarcinoma (PDAC) will survive the malignancy for five years. The ion channel genes-related biomarker and predictive model were needed for exploitation. Methods Differentially expressed ion channel genes (DEICGs) were detected in PDAC patients. GO and KEGG enrichment analysis was conducted on DEICGs. The prognostic genes were found using Cox regression analysis. After that, a risk model was created and examined. A nomogram was created based on independent predictive analysis. The molecular functions of two risk groups were explored. Immune checkpoint molecule expression was compared in two risk groups. We evaluated the possible cancer immunotherapy response in two risk groups using the TIDE method. We further examined how TRPV2 functions in PDAC as a potent oncogene and regulates the activity of macrophages by in vitro validation, including CCK8, EdU, and Transwell assays. Results A total of twenty-four DEICGs were found. Next, we discovered that two DEICGs (TRPV2 and GJB3) were connected to PDAC patients' overall survival (OS). The risk model was created and validated, and a nomogram was used to forecast the overall survival of PDAC patients. The high-risk group considerably accumulated oncogenic pathways. Furthermore, we discovered a correlation between the expression of critical immunological checkpoints and the risk score. Furthermore, patients in the high-risk category had a lower chance of benefiting from immune therapy. The HPA database confirmed that TRPV2 is expressed as a protein. Lastly, TRPV2 controls macrophage activity and acts as a potent oncogene in PDAC. Conclusion Altogether, this study suggested that two ion channel genes, TRPV2 and GJB3, were potential biomarkers for the prognosis of PDAC and immunotherapy targets, and the research will be crucial for creating novel PDAC treatment targets and predictive molecular indicators.
Collapse
Affiliation(s)
- Jiakai Mao
- Department of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yu Tian
- Department of Vascular Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Nan Luo
- Department of Infection, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
4
|
Xiao P, Li C, Liu Y, Gao Y, Liang X, Liu C, Yang W. The role of metal ions in the occurrence, progression, drug resistance, and biological characteristics of gastric cancer. Front Pharmacol 2024; 15:1333543. [PMID: 38370477 PMCID: PMC10869614 DOI: 10.3389/fphar.2024.1333543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/22/2024] [Indexed: 02/20/2024] Open
Abstract
Metal ions exert pivotal functions within the human body, encompassing essential roles in upholding cell structure, gene expression regulation, and catalytic enzyme activity. Additionally, they significantly influence various pathways implicated in divergent mechanisms of cell death. Among the prevailing malignant tumors of the digestive tract worldwide, gastric cancer stands prominent, exhibiting persistent high mortality rates. A compelling body of evidence reveals conspicuous ion irregularities in tumor tissues, encompassing gastric cancer. Notably, metal ions have been observed to elicit distinct contributions to the progression, drug resistance, and biological attributes of gastric cancer. This review consolidates pertinent literature on the involvement of metal ions in the etiology and advancement of gastric cancer. Particular attention is directed towards metal ions, namely, Na, K, Mg, Ca, Fe, Cu, Zn, and Mn, elucidating their roles in the initiation and progression of gastric cancer, cellular demise processes, drug resistance phenomena, and therapeutic approaches.
Collapse
Affiliation(s)
- Pengtuo Xiao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuanda Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yan Gao
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaojing Liang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chang Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
5
|
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: 1.5] [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
|
6
|
Bao L, Festa F, Hirschler-Laszkiewicz I, Keefer K, Wang HG, Cheung JY, Miller BA. The human ion channel TRPM2 modulates migration and invasion in neuroblastoma through regulation of integrin expression. Sci Rep 2022; 12:20544. [PMID: 36446940 PMCID: PMC9709080 DOI: 10.1038/s41598-022-25138-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
Transient receptor potential channel TRPM2 is highly expressed in many cancers and involved in regulation of key physiological processes including mitochondrial function, bioenergetics, and oxidative stress. In Stage 4 non-MYCN amplified neuroblastoma patients, high TRPM2 expression is associated with worse outcome. Here, neuroblastoma cells with high TRPM2 expression demonstrated increased migration and invasion capability. RNA sequencing, RT-qPCR, and Western blotting demonstrated that the mechanism involved significantly greater expression of integrins α1, αv, β1, and β5 in cells with high TRPM2 expression. Transcription factors HIF-1α, E2F1, and FOXM1, which bind promoter/enhancer regions of these integrins, were increased in cells with high TRPM2 expression. Subcellular fractionation confirmed high levels of α1, αv, and β1 membrane localization and co-immunoprecipitation confirmed the presence of α1β1, αvβ1, and αvβ5 complexes. Inhibitors of α1β1, αvβ1, and αvβ5 complexes significantly reduced migration and invasion in cells highly expressing TRPM2, confirming their functional role. Increased pAktSer473 and pERKThr202/Tyr204, which promote migration through mechanisms including integrin activation, were found in cells highly expressing TRPM2. TRPM2 promotes migration and invasion in neuroblastoma cells with high TRPM2 expression through modulation of integrins together with enhancing cell survival, negatively affecting patient outcome and providing rationale for TRPM2 inhibition in anti-neoplastic therapy.
Collapse
Affiliation(s)
- Lei Bao
- grid.29857.310000 0001 2097 4281Departments of Pediatrics, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033 USA
| | - Fernanda Festa
- grid.29857.310000 0001 2097 4281Departments of Pediatrics, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033 USA ,grid.29857.310000 0001 2097 4281Departments of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033 USA
| | - Iwona Hirschler-Laszkiewicz
- grid.29857.310000 0001 2097 4281Departments of Pediatrics, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033 USA
| | - Kerry Keefer
- grid.29857.310000 0001 2097 4281Departments of Pediatrics, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033 USA
| | - Hong-Gang Wang
- grid.29857.310000 0001 2097 4281Departments of Pediatrics, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033 USA ,grid.29857.310000 0001 2097 4281Departments of Pharmacology, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033 USA
| | - Joseph Y. Cheung
- grid.62560.370000 0004 0378 8294Renal Medicine, Brigham and Women’s Hospital, Boston, MA 02115 USA
| | - Barbara A. Miller
- grid.29857.310000 0001 2097 4281Departments of Pediatrics, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033 USA ,grid.29857.310000 0001 2097 4281Departments of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033 USA
| |
Collapse
|
7
|
Li L, Xiao Z, He P, Zou W, Deng Z, Zhang G, Liu R. Molecular subtyping based on TRP family and prognostic assessment for TRP-associated lncRNAs in pancreatic adenocarcinoma. BMC Gastroenterol 2022; 22:454. [PMID: 36371178 PMCID: PMC9652922 DOI: 10.1186/s12876-022-02552-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/17/2022] [Indexed: 11/15/2022] Open
Abstract
Background Transient receptor potential (TRP) channels have high permeability to Ca2+ ions because they are non-selective ion channels. TRP channels have been implicated in tumor onset and progression, proliferation, and migration in recent years. However, the prognostic value of genes related to TRP and their specific mechanism in pancreatic adenocarcinoma (PAAD) are yet to be understood. Methods Public databases such as TCGA and GEO were used to retrieve data on gene expression and clinical information of patients with pancreatic adenocarcinoma for our study. ConsensusClusterPlus package was used for unsupervised clustering analysis. The microenvironment cell population (MCP)-counter approach was employed to measure the immune cells infiltration status. The Pearson correlation was performed to identify TRP-associated lncRNAs. Results Initially, we separated PAAD patients into three clusters depending on TRP-related genes, and of the three clusters, cluster B showed the least immune cell infiltration, which was correlated with poor prognosis. Moreover, GSVA enrichment analysis further revealed that cluster A was subjected to a considerable enrichment in carcinogenic signaling pathways, whereas cluster C was enriched in immune-related pathways. Then, using TRP-associated lncRNAs as a starting point, we constructed a prognostic risk model for PAAD patients that could efficiently predict their prognosis. Further, GSEA revealed that cancer-related pathways, for instance, the cell cycle, p53 signaling pathway, etc. were considerably enriched in the high-risk group. In addition, we looked into the link between the prognostic model and the immunological microenvironment. Lower cytotoxic lymphocytes, NK cells, CD8 T cells, and endothelial cells infiltration were found to be associated with high risk using the MCP-counter algorithm. The expression of CD274, POLE2, MCM6, and LOXL2 was also found to be higher in the high-risk group. TMB was also considerably greater in high-risk individuals, indicating that immune checkpoint inhibitors (ICIs) therapy may benefit them more. Lastly, qRT-PCR further confirmed the differential expression of these prognostic TRP-associated lncRNAs, indicating that these lncRNAs play an imperative role in PAAD tumorigenesis. Conclusion TRP family genes may represent a new class of candidate molecular markers of the occurrence and progression of PAAD. Risk models based on TRP-associated lncRNAs could provide important new references for immunotargeted therapy of pancreatic adenocarcinoma. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02552-y.
Collapse
|
8
|
Bera K, Kiepas A, Zhang Y, Sun SX, Konstantopoulos K. The interplay between physical cues and mechanosensitive ion channels in cancer metastasis. Front Cell Dev Biol 2022; 10:954099. [PMID: 36158191 PMCID: PMC9490090 DOI: 10.3389/fcell.2022.954099] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Physical cues have emerged as critical influencers of cell function during physiological processes, like development and organogenesis, and throughout pathological abnormalities, including cancer progression and fibrosis. While ion channels have been implicated in maintaining cellular homeostasis, their cell surface localization often places them among the first few molecules to sense external cues. Mechanosensitive ion channels (MICs) are especially important transducers of physical stimuli into biochemical signals. In this review, we describe how physical cues in the tumor microenvironment are sensed by MICs and contribute to cancer metastasis. First, we highlight mechanical perturbations, by both solid and fluid surroundings typically found in the tumor microenvironment and during critical stages of cancer cell dissemination from the primary tumor. Next, we describe how Piezo1/2 and transient receptor potential (TRP) channels respond to these physical cues to regulate cancer cell behavior during different stages of metastasis. We conclude by proposing alternative mechanisms of MIC activation that work in tandem with cytoskeletal components and other ion channels to bestow cells with the capacity to sense, respond and navigate through the surrounding microenvironment. Collectively, this review provides a perspective for devising treatment strategies against cancer by targeting MICs that sense aberrant physical characteristics during metastasis, the most lethal aspect of cancer.
Collapse
Affiliation(s)
- Kaustav Bera
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Alexander Kiepas, ; Konstantinos Konstantopoulos,
| | - Yuqi Zhang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
| | - Sean X. Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, The Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Alexander Kiepas, ; Konstantinos Konstantopoulos,
| |
Collapse
|
9
|
Cutliffe AL, McKenna SL, Chandrashekar DS, Ng A, Devonshire G, Fitzgerald RC, O’Donovan TR, Mackrill JJ. Alterations in the Ca2+ toolkit in oesophageal adenocarcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:543-575. [PMID: 36046118 PMCID: PMC9400700 DOI: 10.37349/etat.2021.00063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/08/2021] [Indexed: 11/24/2022] Open
Abstract
Aim: To investigate alterations in transcription of genes, encoding Ca2+ toolkit proteins, in oesophageal adenocarcinoma (OAC) and to assess associations between gene expression, tumor grade, nodal-metastatic stage, and patient survival. Methods: The expression of 275 transcripts, encoding components of the Ca2+ toolkit, was analyzed in two OAC datasets: the Cancer Genome Atlas [via the University of Alabama Cancer (UALCAN) portal] and the oesophageal-cancer, clinical, and molecular stratification [Oesophageal Cancer Clinical and Molecular Stratification (OCCAMS)] dataset. Effects of differential expression of these genes on patient survival were determined using Kaplan-Meier log-rank tests. OAC grade- and metastatic-stage status was investigated for a subset of genes. Adjustment for the multiplicity of testing was made throughout. Results: Of the 275 Ca2+-toolkit genes analyzed, 75 displayed consistent changes in expression between OAC and normal tissue in both datasets. The channel-encoding genes, N-methyl-D-aspartate receptor 2D (GRIN2D), transient receptor potential (TRP) ion channel classical or canonical 4 (TRPC4), and TRP ion channel melastatin 2 (TRPM2) demonstrated the greatest increase in expression in OAC in both datasets. Nine genes were consistently upregulated in both datasets and were also associated with improved survival outcomes. The 6 top-ranking genes for the weighted significance of altered expression and survival outcomes were selected for further analysis: voltage-gated Ca2+ channel subunit α 1D (CACNA1D), voltage-gated Ca2+ channel auxiliary subunit α2 δ4 (CACNA2D4), junctophilin 1 (JPH1), acid-sensing ion channel 4 (ACCN4), TRPM5, and secretory pathway Ca2+ ATPase 2 (ATP2C2). CACNA1D, JPH1, and ATP2C2 were also upregulated in advanced OAC tumor grades and nodal-metastatic stages in both datasets. Conclusions: This study has unveiled alterations of the Ca2+ toolkit in OAC, compared to normal tissue. Such Ca2+ signalling findings are consistent with those from studies on other cancers. Genes that were consistently upregulated in both datasets might represent useful markers for patient diagnosis. Genes that were consistently upregulated, and which were associated with improved survival, might be useful markers for patient outcome. These survival-associated genes may also represent targets for the development of novel chemotherapeutic agents.
Collapse
Affiliation(s)
- Alana L. Cutliffe
- Department of Physiology, University College Cork, BioSciences Institute, T12 YT20 Cork, Ireland
| | - Sharon L. McKenna
- Cancer Research, UCC, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
| | - Darshan S. Chandrashekar
- Department of Pathology, Molecular & Cellular, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Alvin Ng
- Cancer Research UK Cambridge Institute, University of Cambridge Li Ka Shing Centre, Robinson Way, CB2 0RE Cambridge, UK
| | - Ginny Devonshire
- Cancer Research UK Cambridge Institute, University of Cambridge Li Ka Shing Centre, Robinson Way, CB2 0RE Cambridge, UK
| | - Rebecca C. Fitzgerald
- Cancer Research UK Cambridge Institute, University of Cambridge Li Ka Shing Centre, Robinson Way, CB2 0RE Cambridge, UK
| | - Tracey R. O’Donovan
- Cancer Research, UCC, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
| | - John J. Mackrill
- Department of Physiology, University College Cork, BioSciences Institute, T12 YT20 Cork, Ireland
| |
Collapse
|
10
|
Hsu WL, Noda M, Yoshioka T, Ito E. A novel strategy for treating cancer: understanding the role of Ca2+ signaling from nociceptive TRP channels in regulating cancer progression. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:401-415. [PMID: 36045706 PMCID: PMC9400763 DOI: 10.37349/etat.2021.00053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022] Open
Abstract
Cancer is an aging-associated disease and caused by genomic instability that is driven by the accumulation of mutations and epimutations in the aging process. Although Ca2+ signaling, reactive oxygen species (ROS) accumulation, DNA damage response (DDR) and senescence inflammation response (SIR) are processed during genomic instability, the underlying mechanism for the cause of genomic instability and cancer development is still poorly understood and needs to be investigated. Nociceptive transient receptor potential (TRP) channels, which firstly respond to environmental stimuli, such as microbes, chemicals or physical injuries, potentiate regulation of the aging process by Ca2+ signaling. In this review, the authors provide an explanation of the dual role of nociceptive TRP channels in regulating cancer progression, initiating cancer progression by aging-induced genomic instability, and promoting malignancy by epigenetic regulation. Thus, therapeutically targeting nociceptive TRP channels seems to be a novel strategy for treating cancers.
Collapse
Affiliation(s)
- Wen-Li Hsu
- Department of Dermatology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80145, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tohru Yoshioka
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Etsuro Ito
- Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 162-8480, Japan; Department of Biology, Waseda University, Tokyo 162-8480, Japan
| |
Collapse
|
11
|
TRPM2 Oxidation Activates Two Distinct Potassium Channels in Melanoma Cells through Intracellular Calcium Increase. Int J Mol Sci 2021; 22:ijms22168359. [PMID: 34445066 PMCID: PMC8393965 DOI: 10.3390/ijms22168359] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor microenvironments are often characterized by an increase in oxidative stress levels. We studied the response to oxidative stimulation in human primary (IGR39) or metastatic (IGR37) cell lines obtained from the same patient, performing patch-clamp recordings, intracellular calcium ([Ca2+]i) imaging, and RT-qPCR gene expression analysis. In IGR39 cells, chloramine-T (Chl-T) activated large K+ currents (KROS) that were partially sensitive to tetraethylammonium (TEA). A large fraction of KROS was inhibited by paxilline—a specific inhibitor of large-conductance Ca2+-activated BK channels. The TEA-insensitive component was inhibited by senicapoc—a specific inhibitor of the Ca2+-activated KCa3.1 channel. Both BK and KCa3.1 activation were mediated by an increase in [Ca2+]i induced by Chl-T. Both KROS and [Ca2+]i increase were inhibited by ACA and clotrimazole—two different inhibitors of the calcium-permeable TRPM2 channel. Surprisingly, IGR37 cells did not exhibit current increase upon the application of Chl-T. Expression analysis confirmed that the genes encoding BK, KCa3.1, and TRPM2 are much more expressed in IGR39 than in IGR37. The potassium currents and [Ca2+]i increase observed in response to the oxidizing agent strongly suggest that these three molecular entities play a major role in the progression of melanoma. Pharmacological targeting of either of these ion channels could be a new strategy to reduce the metastatic potential of melanoma cells, and could complement classical radio- or chemotherapeutic treatments.
Collapse
|
12
|
Gao L, Liu Y, Du X, Ma S, Ge M, Tang H, Han C, Zhao X, Liu Y, Shao Y, Wu Z, Zhang L, Meng F, Xiao-Feng Qin F. The intrinsic role and mechanism of tumor expressed-CD38 on lung adenocarcinoma progression. Cell Death Dis 2021; 12:680. [PMID: 34226519 PMCID: PMC8256983 DOI: 10.1038/s41419-021-03968-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022]
Abstract
It has been recently reported that CD38 expressed on tumor cells of multiple murine and human origins could be upregulated in response to PD-L1 antibody therapy, which led to dysfunction of tumor-infiltrating CD8+ T immune cells due to increasing the production of adenosine. However, the role of tumor expressed-CD38 on neoplastic formation and progression remains elusive. In the present study, we aimed to delineate the molecular and biochemical function of the tumor-associated CD38 in lung adenocarcinoma progression. Our clinical data showed that the upregulation of tumor-originated CD38 was correlated with poor survival of lung cancer patients. Using multiple in vitro assays we found that the enzymatic activity of tumor expressed-CD38 facilitated lung cancer cell migration, proliferation, colony formation, and tumor development. Consistently, our in vivo results showed that inhibition of the enzymatic activity or antagonizing the enzymatic product of CD38 resulted in the similar inhibition of tumor proliferation and metastasis as CD38 gene knock-out or mutation. At biochemical level, we further identified that cADPR, the mainly hydrolytic product of CD38, was responsible for inducing the opening of TRPM2 iron channel leading to the influx of intracellular Ca2+ and then led to increasing levels of NRF2 while decreasing expression of KEAP1 in lung cancer cells. These findings suggested that malignant lung cancer cells were capable of using cADPR catalyzed by CD38 to facilitate tumor progression, and blocking the enzymatic activity of CD38 could be represented as an important strategy for preventing tumor progression.
Collapse
Affiliation(s)
- Long Gao
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Yuan Liu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Xiaohong Du
- Institute of Clinical Medicine Research, the Affiliated Suzhou Hospital of Nanjing Medical University; Suzhou Science and Technology Town Hospital, Suzhou, China
| | - Sai Ma
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Minmin Ge
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Haijun Tang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Chenfeng Han
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Xin Zhao
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Yanbin Liu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Yun Shao
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Zhao Wu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Lianjun Zhang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China
| | - Fang Meng
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China.
| | - F Xiao-Feng Qin
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
- Suzhou Institute of Systems Medicine, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
13
|
Bettaieb L, Brulé M, Chomy A, Diedro M, Fruit M, Happernegg E, Heni L, Horochowska A, Housseini M, Klouyovo K, Laratte A, Leroy A, Lewandowski P, Louvieaux J, Moitié A, Tellier R, Titah S, Vanauberg D, Woesteland F, Prevarskaya N, Lehen’kyi V. Ca 2+ Signaling and Its Potential Targeting in Pancreatic Ductal Carcinoma. Cancers (Basel) 2021; 13:3085. [PMID: 34205590 PMCID: PMC8235326 DOI: 10.3390/cancers13123085] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/02/2021] [Accepted: 06/13/2021] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer (PC) is a major cause of cancer-associated mortality in Western countries (and estimated to be the second cause of cancer deaths by 2030). The main form of PC is pancreatic adenocarcinoma, which is the fourth most common cause of cancer-related death, and this situation has remained virtually unchanged for several decades. Pancreatic ductal adenocarcinoma (PDAC) is inherently linked to the unique physiology and microenvironment of the exocrine pancreas, such as pH, mechanical stress, and hypoxia. Of them, calcium (Ca2+) signals, being pivotal molecular devices in sensing and integrating signals from the microenvironment, are emerging to be particularly relevant in cancer. Mutations or aberrant expression of key proteins that control Ca2+ levels can cause deregulation of Ca2+-dependent effectors that control signaling pathways determining the cells' behavior in a way that promotes pathophysiological cancer hallmarks, such as enhanced proliferation, survival and invasion. So far, it is essentially unknown how the cancer-associated Ca2+ signaling is regulated within the characteristic landscape of PDAC. This work provides a complete overview of the Ca2+ signaling and its main players in PDAC. Special consideration is given to the Ca2+ signaling as a potential target in PDAC treatment and its role in drug resistance.
Collapse
Affiliation(s)
- Louay Bettaieb
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Maxime Brulé
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Axel Chomy
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Mel Diedro
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Malory Fruit
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Eloise Happernegg
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Leila Heni
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Anaïs Horochowska
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Mahya Housseini
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Kekely Klouyovo
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Agathe Laratte
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Alice Leroy
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Paul Lewandowski
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Joséphine Louvieaux
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Amélie Moitié
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Rémi Tellier
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Sofia Titah
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Dimitri Vanauberg
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - Flavie Woesteland
- Option Biology, Master Biology-Health, Faculty of Medicine Henry Warembourg, University of Lille, 59120 Loos, France; (L.B.); (M.B.); (A.C.); (M.D.); (M.F.); (E.H.); (L.H.); (A.H.); (M.H.); (K.K.); (A.L.); (A.L.); (P.L.); (J.L.); (A.M.); (R.T.); (S.T.); (D.V.); (F.W.)
| | - 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;
- University Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, F-59000 Lille, 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;
- University Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, F-59000 Lille, France
| |
Collapse
|
14
|
Lin R, Bao X, Wang H, Zhu S, Liu Z, Chen Q, Ai K, Shi B. TRPM2 promotes pancreatic cancer by PKC/MAPK pathway. Cell Death Dis 2021; 12:585. [PMID: 34099637 PMCID: PMC8184946 DOI: 10.1038/s41419-021-03856-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022]
Abstract
The mechanism of pancreatic cancer (PA) is not fully understanded. In our last report, TRPM2 plays a promising role in pancreatic cancer. However, the mechanism of TRPM2 is still unknown in this dismal disease. This study was designed to investigate the role and mechanism of TRPM2 in pancreatic cancer. TRPM2 overexpressed and siRNA plasmid were created and transfected with pancreatic cancer cell line (BxPC-3) to construct the cell model. We employed CCK-8, Transwell, scratch wound, and nude mice tumor-bearing model to investigate the role of TRPM2 in pancreatic cancer. Besides, we collected the clinical data, tumor tissue sample (TT) and para-tumor sample (TP) from the pancreatic cancer patients treated in our hospital. We analyzed the mechanism of TRPM2 in pancreatic cancer by transcriptome analysis, western blot, and PCR. We blocked the downstream PKC/MEK pathway of TRPM2 to investigate the mechanism of TRPM2 in pancreatic cancer by CCK8, scratch wound healing, and transwell assays. Overexpressed TRPM2 could promote pancreatic cancer in proliferation, migration, and invasion ability in no matter the cell model or nude mice tumor-bearing model. TRPM2 level is highly negative correlated to the overall survival and progression-free survival time in PA patients, however, it is significantly increased in PA tissue as the tumor stage increases. The transcriptome analysis, GSEA analysis, western-blot, and PCR results indicate TRPM2 is highly correlated with PKC/MAPK pathways. The experiments of PKC/MEK inhibitors added to TRPM2 overexpressed BxPC-3 cell showed that significant inhibition of PA cells happened in CCK8, transwell, and wound-healing assay. TRPM2 may directly activate PKCα by calcium or indirectly activate PKCε and PKCδ by increased DAG in PA, which promote PA by downstream MAPK/MEK pathway activation.
Collapse
Affiliation(s)
- Rui Lin
- General Surgery Department, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Xunxia Bao
- School of Life Science, Anhui Medical University, Hefei, 230022, China
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Hui Wang
- General Surgery Department, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Sibo Zhu
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Zhongyan Liu
- General Surgery Department, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Quanning Chen
- General Surgery Department, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Kaixing Ai
- Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200082, China
| | - Baomin Shi
- General Surgery Department, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
15
|
Prasongsukarn K, Dechkhajorn W, Benjathummarak S, Maneerat Y. TRPM2, PDLIM5, BCL3, CD14, GBA Genes as Feasible Markers for Premature Coronary Heart Disease Risk. Front Genet 2021; 12:598296. [PMID: 34093636 PMCID: PMC8172979 DOI: 10.3389/fgene.2021.598296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 04/19/2021] [Indexed: 12/22/2022] Open
Abstract
Background: Beyond non-genetic risk factors, familial hypercholesterolemia (FH) plays a major role in the development of CHD. FH is a genetic disorder characterized by heritable and severely elevated levels of low-density lipoprotein (LDL) cholesterol, which can lead to premature cardiovascular disease, particularly familial coronary heart disease (FH-CHD). Method: To explore genes indicating a risk of familial (premature) coronary heart disease (FH-CHD) development in FH, 30 Thai male volunteers were enrolled: 7 healthy controls (N), 6 patients with hypercholesterolemia (H), 4 with FH, 10 with CHD, and 3 with FH-CHD. Transcriptome data were investigated using next-generation sequencing analysis in whole blood (n = 3). Genes that were significantly expressed in both FH and FH-CHD, but not in N, H, and CHD groups, were selected and functionally analyzed. Results: The findings revealed that 55 intersecting genes were differentially expressed between FH and FH-CHD groups. Ten of the 55 genes (MAPK14, TRPM2, STARD8, PDLIM5, BCL3, BLOC1S5, GBA, RBMS1, CD14, and CD36 were selected for validation. These 10 genes play potential roles in chronic inflammation and are involved in pathways related to pathogenesis of CHD. Using quantitative real-time PCR, we evaluated the mRNA expression of the selected genes in all 30 volunteers. TRPM2, PDLIM5, BCL3 were significantly upregulated and GBA was significantly downregulated in both FH and FH-CHD compared with the N, H, and CHD groups. Conclusion: our preliminary investigation reveals that the TRPM2, PDLIM5, BCL3, and GBA genes may have potential for further development as predictive markers for FH-CHD.
Collapse
Affiliation(s)
| | - Wilanee Dechkhajorn
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Surachet Benjathummarak
- Center of Excellence for Antibody Research, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yaowapa Maneerat
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
16
|
Mesquita G, Prevarskaya N, Schwab A, Lehen’kyi V. Role of the TRP Channels in Pancreatic Ductal Adenocarcinoma Development and Progression. Cells 2021; 10:cells10051021. [PMID: 33925979 PMCID: PMC8145744 DOI: 10.3390/cells10051021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 12/28/2022] Open
Abstract
The transient receptor potential channels (TRPs) have been related to several different physiologies that range from a role in sensory physiology (including thermo- and osmosensation) to a role in some pathologies like cancer. The great diversity of functions performed by these channels is represented by nine sub-families that constitute the TRP channel superfamily. From the mid-2000s, several reports have shown the potential role of the TRP channels in cancers of multiple origin. The pancreatic cancer is one of the deadliest cancers worldwide. Its prevalence is predicted to rise further. Disappointingly, the treatments currently used are ineffective. There is an urgency to find new ways to counter this disease and one of the answers may lie in the ion channels belonging to the superfamily of TRP channels. In this review, we analyse the existing knowledge on the role of TRP channels in the development and progression of pancreatic ductal adenocarcinoma (PDAC). The functions of these channels in other cancers are also considered. This might be of interest for an extrapolation to the pancreatic cancer in an attempt to identify potential therapeutic interventions.
Collapse
Affiliation(s)
- Gonçalo Mesquita
- 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; (G.M.); (N.P.)
- PHYCELL—Laboratoire de Physiologie Cellulaire, INSERM U1003, University of Lille, 59655 Villeneuve d’Ascq, France
- Institute of Physiology II, University Münster, 48149 Münster, Germany;
| | - 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; (G.M.); (N.P.)
- PHYCELL—Laboratoire de Physiologie Cellulaire, INSERM U1003, University of Lille, 59655 Villeneuve d’Ascq, France
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, 48149 Münster, Germany;
| | - 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; (G.M.); (N.P.)
- PHYCELL—Laboratoire de Physiologie Cellulaire, INSERM U1003, University of Lille, 59655 Villeneuve d’Ascq, France
- Correspondence: ; Tel.: +33-(0)-3-20-33-70-78; Fax: +33-(0)-3-20-43-40-66
| |
Collapse
|
17
|
Hofschröer V, Najder K, Rugi M, Bouazzi R, Cozzolino M, Arcangeli A, Panyi G, Schwab A. Ion Channels Orchestrate Pancreatic Ductal Adenocarcinoma Progression and Therapy. Front Pharmacol 2021; 11:586599. [PMID: 33841132 PMCID: PMC8025202 DOI: 10.3389/fphar.2020.586599] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is a devastating disease with a dismal prognosis. Therapeutic interventions are largely ineffective. A better understanding of the pathophysiology is required. Ion channels contribute substantially to the "hallmarks of cancer." Their expression is dysregulated in cancer, and they are "misused" to drive cancer progression, but the underlying mechanisms are unclear. Ion channels are located in the cell membrane at the interface between the intracellular and extracellular space. They sense and modify the tumor microenvironment which in itself is a driver of PDAC aggressiveness. Ion channels detect, for example, locally altered proton and electrolyte concentrations or mechanical stimuli and transduce signals triggered by these microenvironmental cues through association with intracellular signaling cascades. While these concepts have been firmly established for other cancers, evidence has emerged only recently that ion channels are drivers of PDAC aggressiveness. Particularly, they appear to contribute to two of the characteristic PDAC features: the massive fibrosis of the tumor stroma (desmoplasia) and the efficient immune evasion. Our critical review of the literature clearly shows that there is still a remarkable lack of knowledge with respect to the contribution of ion channels to these two typical PDAC properties. Yet, we can draw parallels from ion channel research in other fibrotic and inflammatory diseases. Evidence is accumulating that pancreatic stellate cells express the same "profibrotic" ion channels. Similarly, it is at least in part known which major ion channels are expressed in those innate and adaptive immune cells that populate the PDAC microenvironment. We explore potential therapeutic avenues derived thereof. Since drugs targeting PDAC-relevant ion channels are already in clinical use, we propose to repurpose those in PDAC. The quest for ion channel targets is both motivated and complicated by the fact that some of the relevant channels, for example, KCa3.1, are functionally expressed in the cancer, stroma, and immune cells. Only in vivo studies will reveal which arm of the balance we should put our weights on when developing channel-targeting PDAC therapies. The time is up to explore the efficacy of ion channel targeting in (transgenic) murine PDAC models before launching clinical trials with repurposed drugs.
Collapse
Affiliation(s)
| | - Karolina Najder
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Micol Rugi
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Rayhana Bouazzi
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Marco Cozzolino
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Münster, Germany
| |
Collapse
|
18
|
Yu P, Cai X, Liang Y, Wang M, Yang W. Roles of NAD + and Its Metabolites Regulated Calcium Channels in Cancer. Molecules 2020; 25:molecules25204826. [PMID: 33092205 PMCID: PMC7587972 DOI: 10.3390/molecules25204826] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/11/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential cofactor for redox enzymes, but also moonlights as a regulator for ion channels, the same as its metabolites. Ca2+ homeostasis is dysregulated in cancer cells and affects processes such as tumorigenesis, angiogenesis, autophagy, progression, and metastasis. Herein, we summarize the regulation of the most common calcium channels (TRPM2, TPCs, RyRs, and TRPML1) by NAD+ and its metabolites, with a particular focus on their roles in cancers. Although the mechanisms of NAD+ metabolites in these pathological processes are yet to be clearly elucidated, these ion channels are emerging as potential candidates of alternative targets for anticancer therapy.
Collapse
Affiliation(s)
- Peilin Yu
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; (P.Y.); (Y.L.)
| | - Xiaobo Cai
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China;
| | - Yan Liang
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; (P.Y.); (Y.L.)
| | - Mingxiang Wang
- BrioPryme Biologics, Inc., Hangzhou 310058, Zhejiang, China;
| | - Wei Yang
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China;
- Correspondence: ; Tel.: +86-571-8820-8713
| |
Collapse
|
19
|
Schnipper J, Dhennin-Duthille I, Ahidouch A, Ouadid-Ahidouch H. Ion Channel Signature in Healthy Pancreas and Pancreatic Ductal Adenocarcinoma. Front Pharmacol 2020; 11:568993. [PMID: 33178018 PMCID: PMC7596276 DOI: 10.3389/fphar.2020.568993] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/16/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth most common cause of cancer-related deaths in United States and Europe. It is predicted that PDAC will become the second leading cause of cancer-related deaths during the next decades. The development of PDAC is not well understood, however, studies have shown that dysregulated exocrine pancreatic fluid secretion can contribute to pathologies of exocrine pancreas, including PDAC. The major roles of healthy exocrine pancreatic tissue are secretion of enzymes and bicarbonate rich fluid, where ion channels participate to fine-tune these biological processes. It is well known that ion channels located in the plasma membrane regulate multiple cellular functions and are involved in the communication between extracellular events and intracellular signaling pathways and can function as signal transducers themselves. Hereby, they contribute to maintain resting membrane potential, electrical signaling in excitable cells, and ion homeostasis. Despite their contribution to basic cellular processes, ion channels are also involved in the malignant transformation from a normal to a malignant phenotype. Aberrant expression and activity of ion channels have an impact on essentially all hallmarks of cancer defined as; uncontrolled proliferation, evasion of apoptosis, sustained angiogenesis and promotion of invasion and migration. Research indicates that certain ion channels are involved in the aberrant tumor growth and metastatic processes of PDAC. The purpose of this review is to summarize the important expression, localization, and function of ion channels in normal exocrine pancreatic tissue and how they are involved in PDAC progression and development. As ion channels are suggested to be potential targets of treatment they are furthermore suggested to be biomarkers of different cancers. Therefore, we describe the importance of ion channels in PDAC as markers of diagnosis and clinical factors.
Collapse
Affiliation(s)
- Julie Schnipper
- Laboratory of Cellular and Molecular Physiology, UR-4667, University of Picardie Jules Verne, Amiens, France
| | - Isabelle Dhennin-Duthille
- Laboratory of Cellular and Molecular Physiology, UR-4667, University of Picardie Jules Verne, Amiens, France
| | - Ahmed Ahidouch
- Laboratory of Cellular and Molecular Physiology, UR-4667, University of Picardie Jules Verne, Amiens, France.,Department of Biology, Faculty of Sciences, Ibn Zohr University, Agadir, Morocco
| | - Halima Ouadid-Ahidouch
- Laboratory of Cellular and Molecular Physiology, UR-4667, University of Picardie Jules Verne, Amiens, France
| |
Collapse
|
20
|
Stokłosa P, Borgström A, Kappel S, Peinelt C. TRP Channels in Digestive Tract Cancers. Int J Mol Sci 2020; 21:E1877. [PMID: 32182937 PMCID: PMC7084354 DOI: 10.3390/ijms21051877] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/03/2020] [Accepted: 03/06/2020] [Indexed: 12/24/2022] Open
Abstract
Cancers of the digestive tract are among the most prevalent types of cancer. These types of cancers are often diagnosed at a late stage, which results in a poor prognosis. Currently, many biomedical studies focus on the role of ion channels, in particular transient receptor potential (TRP) channels, in cancer pathophysiology. TRP channels show mostly non-selective permeability to monovalent and divalent cations. TRP channels are often dysregulated in digestive tract cancers, which can result in alterations of cancer hallmark functions, such as enhanced proliferation, migration, invasion and the inability to induce apoptosis. Therefore, TRP channels could serve as potential diagnostic biomarkers. Moreover, TRP channels are mostly expressed on the cell surface and ion channel targeting drugs do not need to enter the cell, making them attractive candidate drug targets. In this review, we summarize the current knowledge about TRP channels in connection to digestive tract cancers (oral cancer, esophageal cancer, liver cancer, pancreatic cancer, gastric cancer and colorectal cancer) and give an outlook on the potential of TRP channels as cancer biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Paulina Stokłosa
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, 3012 Bern, Switzerland; (A.B.); (S.K.); (C.P.)
| | | | | | | |
Collapse
|
21
|
Anderson KJ, Cormier RT, Scott PM. Role of ion channels in gastrointestinal cancer. World J Gastroenterol 2019; 25:5732-5772. [PMID: 31636470 PMCID: PMC6801186 DOI: 10.3748/wjg.v25.i38.5732] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/26/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023] Open
Abstract
In their seminal papers Hanahan and Weinberg described oncogenic processes a normal cell undergoes to be transformed into a cancer cell. The functions of ion channels in the gastrointestinal (GI) tract influence a variety of cellular processes, many of which overlap with these hallmarks of cancer. In this review we focus on the roles of the calcium (Ca2+), sodium (Na+), potassium (K+), chloride (Cl-) and zinc (Zn2+) transporters in GI cancer, with a special emphasis on the roles of the KCNQ1 K+ channel and CFTR Cl- channel in colorectal cancer (CRC). Ca2+ is a ubiquitous second messenger, serving as a signaling molecule for a variety of cellular processes such as control of the cell cycle, apoptosis, and migration. Various members of the TRP superfamily, including TRPM8, TRPM7, TRPM6 and TRPM2, have been implicated in GI cancers, especially through overexpression in pancreatic adenocarcinomas and down-regulation in colon cancer. Voltage-gated sodium channels (VGSCs) are classically associated with the initiation and conduction of action potentials in electrically excitable cells such as neurons and muscle cells. The VGSC NaV1.5 is abundantly expressed in human colorectal CRC cell lines as well as being highly expressed in primary CRC samples. Studies have demonstrated that conductance through NaV1.5 contributes significantly to CRC cell invasiveness and cancer progression. Zn2+ transporters of the ZIP/SLC39A and ZnT/SLC30A families are dysregulated in all major GI organ cancers, in particular, ZIP4 up-regulation in pancreatic cancer (PC). More than 70 K+ channel genes, clustered in four families, are found expressed in the GI tract, where they regulate a range of cellular processes, including gastrin secretion in the stomach and anion secretion and fluid balance in the intestinal tract. Several distinct types of K+ channels are found dysregulated in the GI tract. Notable are hERG1 upregulation in PC, gastric cancer (GC) and CRC, leading to enhanced cancer angiogenesis and invasion, and KCNQ1 down-regulation in CRC, where KCNQ1 expression is associated with enhanced disease-free survival in stage II, III, and IV disease. Cl- channels are critical for a range of cellular and tissue processes in the GI tract, especially fluid balance in the colon. Most notable is CFTR, whose deficiency leads to mucus blockage, microbial dysbiosis and inflammation in the intestinal tract. CFTR is a tumor suppressor in several GI cancers. Cystic fibrosis patients are at a significant risk for CRC and low levels of CFTR expression are associated with poor overall disease-free survival in sporadic CRC. Two other classes of chloride channels that are dysregulated in GI cancers are the chloride intracellular channels (CLIC1, 3 & 4) and the chloride channel accessory proteins (CLCA1,2,4). CLIC1 & 4 are upregulated in PC, GC, gallbladder cancer, and CRC, while the CLCA proteins have been reported to be down-regulated in CRC. In summary, it is clear, from the diverse influences of ion channels, that their aberrant expression and/or activity can contribute to malignant transformation and tumor progression. Further, because ion channels are often localized to the plasma membrane and subject to multiple layers of regulation, they represent promising clinical targets for therapeutic intervention including the repurposing of current drugs.
Collapse
Affiliation(s)
- Kyle J Anderson
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Robert T Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Patricia M Scott
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| |
Collapse
|
22
|
Abstract
The TRP ion channel TRPM2 has an essential function in cell survival and protects the viability of a number of cell types after oxidative stress. It is highly expressed in many cancers including breast, prostate, and pancreatic cancer, melanoma, leukemia, and neuroblastoma, suggesting it promotes cancer cell survival. TRPM2 is activated by production of ADP-ribose (ADPR) following oxidative stress, which binds to the C-terminus of TRPM2, resulting in channel opening. In a number of cancers including neuroblastoma, TRPM2 has been shown to preserve viability and mechanisms have been identified. Activation of TRPM2 results in expression of transcription factors and kinases important in cell proliferation and survival including HIF-1/2α, CREB, nuclear factor (erythroid-derived 2)-related factor-2 (Nrf2), and Pyk2, and Src phosphorylation. Together, HIF-1/2α and CREB regulate expression of genes encoding proteins with roles in mitochondrial function including members of the electron transport complex involved in ATP production. These contribute to lower mitochondrial ROS production while expression of antioxidants regulated by HIF-1/2α, FOXO3a, CREB, and Nrf2 is maintained. CREB is also important in control of expression of key proteins involved in autophagy. When TRPM2-mediated calcium influx is inhibited, mitochondria are dysfunctional, cellular bioenergetics are reduced, production of ROS is increased, and autophagy and DNA repair are impaired, decreasing tumor growth and increasing chemotherapy sensitivity. Inhibition of TRPM2 expression or function results in decreased tumor proliferation and/or viability in many malignancies including breast, gastric, pancreatic, prostate, head and neck cancers, melanoma, neuroblastoma, and T-cell and acute myelogenous leukemia. However, in a small number of malignancies, activation of TRPM2 rather than inhibition has been reported to reduce tumor cell survival. Here, TRPM2-mediated Ca2+ signaling and mechanisms of regulation of cancer cell growth and survival are reviewed and controversies discussed. Evidence suggests that targeting TRPM2 may be a novel therapeutic approach in many cancers.
Collapse
Affiliation(s)
- Barbara A Miller
- Departments of Pediatrics, and Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033, USA.
| |
Collapse
|
23
|
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: 9.7] [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
|