1
|
Guo J, Liu F, Yang X, Wang M, Yang S, Lan K, Yan L, Cao R, Chen X, Zhong W. TRPM4 channels contribute to Adriamycin chemoresistance in breast cancer cells. Eur J Pharmacol 2025; 998:177637. [PMID: 40250821 DOI: 10.1016/j.ejphar.2025.177637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Chemoresistance presents a critical challenge in breast cancer treatment. Here, we report that transient receptor potential melastatin 4 (TRPM4) plays a role in modulating doxorubicin (ADR) resistance in breast cancer cells. TRPM4 expression was significantly upregulated at both the mRNA and protein levels in MCF-7/ADR cells, a human breast cancer cell line resistant to the chemotherapy drug ADR. Pharmacological inhibition or knockdown of TRPM4 restored ADR sensitivity, while its overexpression in non-resistant MCF-7 cells diminished drug response, confirming the regulatory role of TRPM4 in resistance mechanisms. Western blot analyses confirmed that elevated TRPM4 expression drives P-glycoprotein (P-gp) upregulation in both MCF-7/ADR and KBv200 cells (KB vinblastine 200 resistant cell line), as well as in Huh7 (human hepatocellular carcinoma cell line) and HCT116 (human colorectal cancer cell line). In addition, we demonstrate that TRPM4 inhibition suppresses the level of NF-κB, a pivotal transcription factor regulating P-gp expression. Furthermore, we found that TRPM4-mediated cellular swelling, rather than membrane depolarization, is the primary driver of P-gp overexpression. Drug-resistant MCF-7/ADR cells exhibited significantly larger cell sizes compared to non-resistant MCF-7 cells, and this effect was reversed following TRPM4 inhibition. The swelling was induced by hypotonic stress rather than changes in membrane potential, further confirming the role of TRPM4 in P-gp regulation through volume changes. Analysis of the TCGA (The Cancer Genome Atlas) database revealed that elevated TRPM4 expression correlates with reduced patient survival, suggesting that TRPM4 plays a role in both drug resistance and tumor progression. Our findings provide new insights into the role of TRPM4 in resistance mechanisms and propose that targeting TRPM4 could represent an innovative therapeutic strategy to overcome chemoresistance and enhance drug efficacy in breast cancer.
Collapse
Affiliation(s)
- Juan Guo
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China; National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Fang Liu
- Department of Pathology, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Xinyue Yang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Mengyuan Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Shangze Yang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Kun Lan
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Li Yan
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ruiyuan Cao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| | - Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China; National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| |
Collapse
|
2
|
Zhang Y, Yi Y, Shu Y, Ru X, He S. TRP channels and breast cancer: the role of TRPs in the pathophysiological development. Front Mol Biosci 2025; 12:1528663. [PMID: 40078961 PMCID: PMC11896876 DOI: 10.3389/fmolb.2025.1528663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/27/2025] [Indexed: 03/14/2025] Open
Abstract
TRP channels play important roles in regulating various physiological and pathological processes, including the progression of cancer. Several TRP channels mediate tumour development. This review focuses on the role of TRP channels in the development of breast cancer, including their involvement in proliferation, apoptosis, autophagy, metastasis, and angiogenesis. TRP channels are associated with breast carcinogenesis and their role as potential therapeutic targets and prognostic biomarkers is under investigation. This review summarizes the reported effects of inhibiting or agonizing various TRP channel in breast cancer cells. Although there are relatively mature protocols for the treatment of breast cancer, its treatment is not currently a breakthrough, and therapies targeting TRP channels may be a developable strategy for it.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| | - Yanfeng Yi
- Department of Life Sciences and Health, School of Science and Engineering, Huzhou College, Huzhou, China
| | - Yinghao Shu
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| | - Xiaochen Ru
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| | - Shuaibing He
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| |
Collapse
|
3
|
Demaree IS, Kumar S, Tennessen K, Hoang QQ, White FA, Obukhov AG. Effects of TRPC1's Lysines on Heteromeric TRPC5-TRPC1 Channel Function. Cells 2024; 13:2019. [PMID: 39682767 PMCID: PMC11640535 DOI: 10.3390/cells13232019] [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: 10/20/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND TRPC5 proteins form plasma membrane cation channels and are expressed in the nervous and cardiovascular systems. TRPC5 activation leads to cell depolarization and increases neuronal excitability, whereas a homologous TRPC1 inhibits TRPC5 function via heteromerization. The mechanism underlying the inhibitory effect of TRPC1 in TRPC5/TRPC1 heteromers remains unknown. METHODS We used electrophysiological techniques to examine the roles of subunit stoichiometry and positively charged luminal residues of TRPC1 on TRPC5/TRPC1 function. We also performed molecular dynamics simulations. RESULTS We found that increasing the relative amount of TRPC1 in TRPC5/TRPC1 heteromers reduced histamine-induced cation influx through the heteromeric channels. Consistently, histamine-induced cation influx was small in cells co-expressing TRPC5-TRPC1 concatemers and TRPC1, and large in cells co-expressing TRPC5-TRPC1 concatemers and TRPC5. Molecular dynamics simulations revealed that the TRPC1 protein has two positively charged lysine residues that are facing the heteromeric channel pore lumen. Substitution of these lysines with asparagines decreased TRPC1's inhibitory effect on TRPC5/TRPC1 function, indicating that these lysines may regulate cation influx through TRPC5/TRPC1 heteromers. Additionally, we established that extracellular Mg2+ inhibits cation influx through TRPC5/TRPC1, contributing to channel regulation. CONCLUSIONS We revealed that the inhibitory effect of TRPC1 on heteromeric TRPC5/TRPC1 function likely involves luminal lysines of TRPC1.
Collapse
Affiliation(s)
- Isaac S. Demaree
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (I.S.D.); (S.K.)
| | - Sanjay Kumar
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (I.S.D.); (S.K.)
- Department of Life Science, School of Earth, Biological, and Environmental Sciences, Central University of South Bihar, Gaya 824236, India
| | - Kayla Tennessen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (K.T.); (Q.Q.H.)
| | - Quyen Q. Hoang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (K.T.); (Q.Q.H.)
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Fletcher A. White
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander G. Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (I.S.D.); (S.K.)
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| |
Collapse
|
4
|
Sánchez JC, Alemán A, Henao JF, Olaya JC, Ehrlich BE. NCS-1 protein regulates TRPA1 channel through the PI3K pathway in breast cancer and neuronal cells. J Physiol Biochem 2024; 80:451-463. [PMID: 38564162 PMCID: PMC11074019 DOI: 10.1007/s13105-024-01016-z] [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: 11/29/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024]
Abstract
The physical and functional interaction between transient receptor potential channel ankyrin 1 (TRPA1) and neuronal calcium sensor 1 (NCS-1) was assessed. NCS-1 is a calcium (Ca2+) sensor found in many tissues, primarily neurons, and TRPA1 is a Ca2+ channel involved not only in thermal and pain sensation but also in conditions such as cancer and chemotherapy-induced peripheral neuropathy, in which NCS-1 is also a regulatory component.We explored the interactions between these two proteins by employing western blot, qRT-PCR, co-immunoprecipitation, Ca2+ transient monitoring with Fura-2 spectrophotometry, and electrophysiology assays in breast cancer cells (MDA-MB-231) with different levels of NCS-1 expression and neuroblastoma cells (SH-SY5Y).Our findings showed that the expression of TRPA1 was directly correlated with NCS-1 levels at both the protein and mRNA levels. Additionally, we found a physical and functional association between these two proteins. Physically, the NCS-1 and TRPA1 co-immunoprecipitate. Functionally, NCS-1 enhanced TRPA1-dependent Ca2+ influx, current density, open probability, and conductance, where the functional effects depended on PI3K. Conclusion: NCS-1 appears to act not only as a Ca2+ sensor but also modulates TRPA1 protein expression and channel function in a direct fashion through the PI3K pathway. These results contribute to understanding how Ca2+ homeostasis is regulated and provides a mechanism underlying conditions where Ca2+ dynamics are compromised, including breast cancer. With a cellular pathway identified, targeted treatments can be developed for breast cancer and neuropathy, among other related diseases.
Collapse
Affiliation(s)
- Julio C Sánchez
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia.
| | - Alexander Alemán
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia
| | - Juan F Henao
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia
| | - Juan C Olaya
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia
| | - Barbara E Ehrlich
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, CT, 06520, USA
| |
Collapse
|
5
|
Chen S, Kim JK. The Role of Cannabidiol in Liver Disease: A Systemic Review. Int J Mol Sci 2024; 25:2370. [PMID: 38397045 PMCID: PMC10888697 DOI: 10.3390/ijms25042370] [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: 01/22/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Cannabidiol (CBD), a non-psychoactive phytocannabinoid abundant in Cannabis sativa, has gained considerable attention for its anti-inflammatory, antioxidant, analgesic, and neuroprotective properties. It exhibits the potential to prevent or slow the progression of various diseases, ranging from malignant tumors and viral infections to neurodegenerative disorders and ischemic diseases. Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease, and viral hepatitis stand as prominent causes of morbidity and mortality in chronic liver diseases globally. The literature has substantiated CBD's potential therapeutic effects across diverse liver diseases in in vivo and in vitro models. However, the precise mechanism of action remains elusive, and an absence of evidence hinders its translation into clinical practice. This comprehensive review emphasizes the wealth of data linking CBD to liver diseases. Importantly, we delve into a detailed discussion of the receptors through which CBD might exert its effects, including cannabinoid receptors, CB1 and CB2, peroxisome proliferator-activated receptors (PPARs), G protein-coupled receptor 55 (GPR55), transient receptor potential channels (TRPs), and their intricate connections with liver diseases. In conclusion, we address new questions that warrant further investigation in this evolving field.
Collapse
Affiliation(s)
- Si Chen
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea;
| | - Jeon-Kyung Kim
- Institute of New Drug Development, School of Pharmacy, Jeonbuk National University, Jeonju 54896, Jeonbuk, Republic of Korea
| |
Collapse
|
6
|
Zhang Y, Guan Y, Zheng X, Li C. Hypoxia-induced miR-181a-5p up-regulation reduces epirubicin sensitivity in breast cancer cells through inhibiting EPDR1/TRPC1 to activate PI3K/AKT signaling pathway. BMC Cancer 2024; 24:167. [PMID: 38308220 PMCID: PMC10835859 DOI: 10.1186/s12885-024-11906-6] [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: 03/16/2023] [Accepted: 01/21/2024] [Indexed: 02/04/2024] Open
Abstract
Breast carcinoma (BC) ranks as a predominant malignancy and constitutes the second principal cause of mortality among women globally. Epirubicin stands as the drug of choice for BC therapeutics. Nevertheless, the emergence of chemoresistance has significantly curtailed its therapeutic efficacy. The resistance mechanisms to Epirubicin remain not entirely elucidated, yet they are conjectured to stem from diminished tumor vascular perfusion and resultant hypoxia consequent to Epirubicin administration. In our investigation, we meticulously scrutinized the Gene Expression Omnibus database for EPDR1, a gene implicated in hypoxia and Epirubicin resistance in BC. Subsequently, we delineated the impact of EPDR1 on cellular proliferation, motility, invasive capabilities, and interstitial-related proteins in BC cells, employing methodologies such as the CCK-8 assay, Transwell assay, and western blot analysis. Our research further unveiled that hypoxia-induced miR-181a-5p orchestrates the regulation of BC cell duplication, migration, invasion, and interstitial-related protein expression via modulation of EPDR1. In addition, we identified TRPC1, a gene associated with EPDR1 expression in BC, and substantiated that EPDR1 influences BC cellular dynamics through TRPC1-mediated modulation of the PI3K/AKT signaling cascade. Our findings underscore the pivotal role of EPDR1 in the development of BC. EPDR1 was found to be expressed at subdued levels in BC tissues, Epirubicin-resistant BC cells, and hypoxic BC cells. The overexpression of EPDR1 curtailed BC cell proliferation, motility, invasiveness, and the expression of interstitial-related proteins. At a mechanistic level, the overexpression of hypoxia-induced miR-181a-5p was observed to inhibit the EPDR1/TRPC1 axis, thereby activating the PI3K/AKT signaling pathway and diminishing the sensitivity to Epirubicin in BC cells. In summation, our study demonstrates that the augmentation of hypoxia-induced miR-181a-5p diminishes Epirubicin sensitivity in BC cells by attenuating EPDR1/TRPC1 expression, thereby invigorating the PI3K/AKT signaling pathway. This exposition offers a theoretical foundation for the application of Epirubicin in BC therapy, marking a significant contribution to the existing body of oncological literature.
Collapse
Affiliation(s)
- Yunwei Zhang
- Department of Breast Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China
- Department of Breast Clinic, Shenyang Maternity and Child Health Hosital, No. 20, Yuanjiang Street, Shenyang, Liaoning, China
| | - Yunping Guan
- Department of Breast Clinic, Shenyang Maternity and Child Health Hosital, No. 20, Yuanjiang Street, Shenyang, Liaoning, China
| | - Xinyu Zheng
- Department of Breast Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China.
- Lab 1, Cancer Institute, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China.
| | - Chenyang Li
- Department of Breast Clinic, Shenyang Maternity and Child Health Hosital, No. 20, Yuanjiang Street, Shenyang, Liaoning, China.
| |
Collapse
|
7
|
Chai XX, Liu J, Yu TY, Zhang G, Sun WJ, Zhou Y, Ren L, Cao HL, Yin DC, Zhang CY. Recent progress of mechanosensitive mechanism on breast cancer. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 185:1-16. [PMID: 37793504 DOI: 10.1016/j.pbiomolbio.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/10/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
The mechanical environment is important for tumorigenesis and progression. Tumor cells can sense mechanical signals by mechanosensitive receptors, and these mechanical signals can be converted to biochemical signals to regulate cell behaviors, such as cell differentiation, proliferation, migration, apoptosis, and drug resistance. Here, we summarized the effects of the mechanical microenvironment on breast cancer cell activity, and mechanotransduction mechanism from cellular microenvironment to cell membrane, and finally to the nucleus, and also relative mechanosensitive proteins, ion channels, and signaling pathways were elaborated, therefore the mechanical signal could be transduced to biochemical or molecular signal. Meanwhile, the mechanical models commonly used for biomechanics study in vitro and some quantitative descriptions were listed. It provided an essential theoretical basis for the occurrence and development of mechanosensitive breast cancer, and also some potential drug targets were proposed to treat such disease.
Collapse
Affiliation(s)
- Xiao-Xia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Jie Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Tong-Yao Yu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Wen-Jun Sun
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Yan Zhou
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Li Ren
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China; Key Laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, Ningbo, 315103, Zhejiang, PR China
| | - Hui-Ling Cao
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, School of Pharmacy, Xi'an Medical University, Xi'an, 710021, Shaanxi, PR China.
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China.
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China.
| |
Collapse
|
8
|
Tobiasz J, Polanska J. Proteomic Profile Distinguishes New Subpopulations of Breast Cancer Patients with Different Survival Outcomes. Cancers (Basel) 2023; 15:4230. [PMID: 37686507 PMCID: PMC10486506 DOI: 10.3390/cancers15174230] [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: 07/25/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
As a highly heterogeneous disease, breast cancer (BRCA) demonstrates a diverse molecular portrait. The well-established molecular classification (PAM50) relies on gene expression profiling. It insufficiently explains the observed clinical and histopathological diversity of BRCAs. This study aims to demographically and clinically characterize the six BRCA subpopulations (basal, HER2-enriched, and four luminal ones) revealed by their proteomic portraits. GMM-based high variate protein selection combined with PCA/UMAP was used for dimensionality reduction, while the k-means algorithm allowed patient clustering. The statistical analysis (log-rank and Gehan-Wilcoxon tests, hazard ratio HR as the effect size ES) showed significant differences across identified subpopulations in Disease-Specific Survival (p = 0.0160) and Progression-Free Interval (p = 0.0264). Luminal subpopulations vary in prognosis (Disease-Free Interval, p = 0.0277). The A2 subpopulation is of the poorest, comparable to the HER2-enriched subpopulation, prognoses (HR = 1.748, referenced to Luminal B, small ES), while A3 is of the best (HR = 0.250, large ES). Similar to PAM50 subtypes, no substantial dependency on demographic and clinical factors was detected across Luminal subpopulations, as measured by χ2 test and Cramér's V for ES, and ANOVA with appropriate post hocs combined with η2 or Cohen's d-type ES, respectively. Progesterone receptors can serve as the potential A2 biomarker within Luminal patients. Further investigation of molecular differences is required to examine the potential prognostic or clinical applications.
Collapse
Affiliation(s)
- Joanna Tobiasz
- Department of Data Science and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland;
- Department of Computer Graphics, Vision and Digital Systems, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Joanna Polanska
- Department of Data Science and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland;
| |
Collapse
|
9
|
Liu Y, Lyu Y, Zhu L, Wang H. Role of TRP Channels in Liver-Related Diseases. Int J Mol Sci 2023; 24:12509. [PMID: 37569884 PMCID: PMC10420300 DOI: 10.3390/ijms241512509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
The liver plays a crucial role in preserving the homeostasis of an entire organism by metabolizing both endogenous and exogenous substances, a process that relies on the harmonious interactions of hepatocytes, hepatic stellate cells (HSCs), Kupffer cells (KCs), and vascular endothelial cells (ECs). The disruption of the liver's normal structure and function by diverse pathogenic factors imposes a significant healthcare burden. At present, most of the treatments for liver disease are palliative in nature, rather than curative or restorative. Transient receptor potential (TRP) channels, which are extensively expressed in the liver, play a crucial role in regulating intracellular cation concentration and serve as the origin or intermediary stage of certain signaling pathways that contribute to liver diseases. This review provides an overview of recent developments in liver disease research, as well as an examination of the expression and function of TRP channels in various liver cell types. Furthermore, we elucidate the molecular mechanism by which TRP channels mediate liver injury, liver fibrosis, and hepatocellular carcinoma (HCC). Ultimately, the present discourse delves into the current state of research and extant issues pertaining to the targeting of TRP channels in the treatment of liver diseases and other ailments. Despite the numerous obstacles encountered, TRP channels persist as an extremely important target for forthcoming clinical interventions aimed at treating liver diseases.
Collapse
Affiliation(s)
- Yusheng Liu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China; (Y.L.); (Y.L.)
| | - Yihan Lyu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China; (Y.L.); (Y.L.)
| | - Lijuan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing 210009, China;
| | - Hongmei Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China; (Y.L.); (Y.L.)
| |
Collapse
|
10
|
Saldías MP, Cruz P, Silva I, Orellana-Serradell O, Lavanderos B, Maureira D, Pinto R, Cerda O. The Cytoplasmic Region of SARAF Reduces Triple-Negative Breast Cancer Metastasis through the Regulation of Store-Operated Calcium Entry. Int J Mol Sci 2023; 24:ijms24065306. [PMID: 36982380 PMCID: PMC10049260 DOI: 10.3390/ijms24065306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Triple-negative breast cancer has a poor prognosis and is non-responsive to first-line therapies; hence, new therapeutic strategies are needed. Enhanced store-operated Ca2+ entry (SOCE) has been widely described as a contributing factor to tumorigenic behavior in several tumor types, particularly in breast cancer cells. SOCE-associated regulatory factor (SARAF) acts as an inhibitor of the SOCE response and, therefore, can be a potential antitumor factor. Herein, we generated a C-terminal SARAF fragment to evaluate the effect of overexpression of this peptide on the malignancy of triple-negative breast cancer cell lines. Using both in vitro and in vivo approaches, we showed that overexpression of the C-terminal SARAF fragment reduced proliferation, cell migration, and the invasion of murine and human breast cancer cells by decreasing the SOCE response. Our data suggest that regulating the activity of the SOCE response via SARAF activity might constitute the basis for further alternative therapeutic strategies for triple-negative breast cancer.
Collapse
Affiliation(s)
- María Paz Saldías
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Pablo Cruz
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Ian Silva
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Octavio Orellana-Serradell
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Boris Lavanderos
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Diego Maureira
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Raquel Pinto
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Oscar Cerda
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
- Correspondence: ; Tel.: +56-2-29786909
| |
Collapse
|
11
|
Qi H, Wu F, Wang H. Function of TRPC1 in modulating hepatocellular carcinoma progression. Med Oncol 2023; 40:97. [PMID: 36797544 DOI: 10.1007/s12032-023-01964-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023]
Abstract
The liver is the main organ of metabolism in the human body, and it is easy to suffer from hepatitis, cirrhosis, liver cancer, and other diseases, the most serious of which is liver cancer. Worldwide, liver cancer is the most common and deadly malignant tumor, the third leading cause of cancer death in the world. Based on TCGA and ICGC databases, our research discovered the important role of TRPC1 in liver cancer through bioinformatics. The results showed that TRPC1 was over-expressed in hepatocellular carcinoma, and the higher the expression level of TRPC1, the worse the OS and the lower the survival rate. TRPC1 was a risk factor affecting the overall survival probability of hepatocellular carcinoma patients. By analyzing the function of the TRP family in liver cancer, TRPC1 might promote the occurrence of liver cancer by up-regulating common signal pathways in tumors such as tumor proliferation signature, and down-regulating important metabolic reactions such as retinol metabolism. In addition, TRPC1 could promote the development of liver cancer by up-regulating the expression of ABI2, MAPRE1, YEATS2, MTA3, TMEM237, MTMR2, CCDC6, AC069544.2, and NCBP2 genes. These results illustrate that TRPC1 is very valuable in the study of liver cancer.
Collapse
Affiliation(s)
- Huimin Qi
- School of Basic Medicine, Weifang Medical University, Weifang, 261053, China
| | - Fengming Wu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Hongmei Wang
- School of Medicine, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
12
|
Acid Adaptation Promotes TRPC1 Plasma Membrane Localization Leading to Pancreatic Ductal Adenocarcinoma Cell Proliferation and Migration through Ca 2+ Entry and Interaction with PI3K/CaM. Cancers (Basel) 2022; 14:cancers14194946. [PMID: 36230869 PMCID: PMC9563726 DOI: 10.3390/cancers14194946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers globally, with a 5-year overall survival of less than 10%. The development and progression of PDAC are linked to its fluctuating acidic tumor microenvironment. Ion channels act as important sensors of this acidic tumor microenvironment. They transduce extracellular signals and regulate signaling pathways involved in all hallmarks of cancer. In this study, we evaluated the interplay between a pH-sensitive ion channel, the calcium (Ca2+) channel transient receptor potential C1 (TRPC1), and three different stages of the tumor microenvironment, normal pH, acid adaptation, and acid recovery, and its impact on PDAC cell migration, proliferation, and cell cycle progression. In acid adaptation and recovery conditions, TRPC1 localizes to the plasma membrane, where it interacts with PI3K and calmodulin, and permits Ca2+ entry, which results in downstream signaling, leading to proliferation and migration. Thus, TRPC1 exerts a more aggressive role after adaptation to the acidic tumor microenvironment. Abstract Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a low overall survival rate of less than 10% and limited therapeutic options. Fluctuations in tumor microenvironment pH are a hallmark of PDAC development and progression. Many ion channels are bona fide cellular sensors of changes in pH. Yet, the interplay between the acidic tumor microenvironment and ion channel regulation in PDAC is poorly understood. In this study, we show that acid adaption increases PANC-1 cell migration but attenuates proliferation and spheroid growth, which are restored upon recovery. Moreover, acid adaptation and recovery conditions favor the plasma membrane localization of the pH-sensitive calcium (Ca2+) channel transient receptor potential C1 (TRPC1), TRPC1-mediated Ca2+ influx, channel interaction with the PI3K p85α subunit and calmodulin (CaM), and AKT and ERK1/2 activation. Knockdown (KD) of TRPC1 suppresses cell migration, proliferation, and spheroid growth, notably in acid-recovered cells. KD of TRPC1 causes the accumulation of cells in G0/G1 and G2/M phases, along with reduced expression of CDK6, −2, and −1, and cyclin A, and increased expression of p21CIP1. TRPC1 silencing decreases the basal Ca2+ influx in acid-adapted and -recovered cells, but not in normal pH conditions, and Ca2+ chelation reduces cell migration and proliferation solely in acid adaptation and recovery conditions. In conclusion, acid adaptation and recovery reinforce the involvement of TRPC1 in migration, proliferation, and cell cycle progression by permitting Ca2+ entry and forming a complex with the PI3K p85α subunit and CaM.
Collapse
|
13
|
Sun Y, Zboril EK, De La Chapa JJ, Chai X, Da Conceicao VN, Valdez MC, McHardy SF, Gonzales CB, Singh BB. Inhibition of Ca 2+ entry by capsazepine analog CIDD-99 prevents oral squamous carcinoma cell proliferation. Front Physiol 2022; 13:969000. [PMID: 36187775 PMCID: PMC9521718 DOI: 10.3389/fphys.2022.969000] [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: 06/15/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Oral cancer patients have a poor prognosis, with approximately 66% of patients surviving 5-years after diagnosis. Treatments for oral cancer are limited and have many adverse side effects; thus, further studies are needed to develop drugs that are more efficacious. To achieve this objective, we developed CIDD-99, which produces cytotoxic effects in multiple oral squamous cell carcinoma (OSCC) cell lines. While we demonstrated that CIDD-99 induces ER stress and apoptosis in OSCC, the mechanism was unclear. Investigation of the Bcl-family of proteins showed that OSCC cells treated with CIDD-99 undergo downregulation of Bcl-XL and Bcl-2 anti-apoptotic proteins and upregulation of Bax (pro-apoptotic). Importantly, OSCC cells treated with CIDD-99 displayed decreased calcium signaling in a dose and time-dependent manner, suggesting that blockage of calcium signaling is the key mechanism that induces cell death in OSCC. Indeed, CIDD-99 anti-proliferative effects were reversed by the addition of exogenous calcium. Moreover, electrophysiological properties further established that calcium entry was via the non-selective TRPC1 channel and prolonged CIDD-99 incubation inhibited STIM1 expression. CIDD-99 inhibition of calcium signaling also led to ER stress and inhibited mitochondrial complexes II and V in vitro. Taken together, these findings suggest that inhibition of TRPC mediates induction of ER stress and mitochondrial dysfunction as a part of the cellular response to CIDD-99 in OSCC.
Collapse
Affiliation(s)
- Yuyang Sun
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Emily K. Zboril
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Jorge J. De La Chapa
- Department of Comprehensive Dentistry, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Xiufang Chai
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | | | - Matthew C. Valdez
- Department of Chemistry and the Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX, United States
| | - Stanton F. McHardy
- Department of Chemistry and the Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX, United States
| | - Cara B. Gonzales
- Department of Comprehensive Dentistry, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Brij B. Singh
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
14
|
Chen W, Zhang Y, Li R, Huang W, Wei X, Zeng D, Liang Y, Zeng Y, Chen M, Zhang L, Gao W, Zhu Y, Li Y, Zhang G. Notch3 Transactivates Glycogen Synthase Kinase-3-Beta and Inhibits Epithelial-to-Mesenchymal Transition in Breast Cancer Cells. Cells 2022; 11:cells11182872. [PMID: 36139447 PMCID: PMC9497076 DOI: 10.3390/cells11182872] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/10/2022] [Accepted: 08/19/2022] [Indexed: 02/05/2023] Open
Abstract
As a critical transformational process in the attributes of epithelial cells, epithelial-to-mesenchymal transition (EMT) is involved in tumor invasion, metastasis, and resistance to treatment, which contributes to the ultimate death of some patients with breast cancer. Glycogen synthase kinase-3-beta (GSK3β) is thought to be an EMT suppressor that down-regulates the protein, snail, a zinc finger transcription inhibitor, and regulates E-cadherin expression and the Wnt signaling pathway. Our previous studies have shown that Notch3 also inhibits EMT in breast cancer. In mammary gland cells, GSK3β physically bound and phosphorylated the intracellular domain of two Notch paralogs: N1ICD was positively regulated, but N2ICD was negatively regulated; however, the relationship between Notch3, GSK3β, and EMT in breast cancer is still unclear and crosstalk between Notch3 and GSK3β has not been widely investigated. In this study, we revealed that Notch3 was an essential antagonist of EMT in breast cancer cells by transcriptionally upregulating GSK3β. In breast cancer, MCF-7 and MDA-MB-231 cell lines, the silencing of Notch3 reduced GSK3β expression, which is sufficient to induce EMT. Conversely, ectopic Notch3 expression re-activated GSK3β and E-cadherin. Mechanistically, Notch3 can bind to the GSK3β promoter directly and activate GSK3β transcription. In human breast cancer samples, Notch3 expression is positively associated with GSK3β (r = 0.416, p = 0.001); moreover, high expressions of Notch3 and GSK3β mRNA are correlated to better relapse-free survival in all breast cancer patients via analysis in "the Kaplan-Meier plotter" database. In summary, our preliminary results suggested that Notch3 might inhibit EMT by trans-activating GSK3β in breast cancer cells. The suppression of Notch3 expression may contribute to EMT by transcriptionally downregulating GSK3β in breast cancer.
Collapse
Affiliation(s)
- Weiling Chen
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Yongqu Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Ronghui Li
- Department of Medical Oncology, Xiang’an Hospital of Xiamen University, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Wenhe Huang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Xiaolong Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515041, China
| | - De Zeng
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515041, China
| | - Yuanke Liang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou 515041, China
| | - Yunzhu Zeng
- Department of Pathology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515041, China
| | - Min Chen
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Lixin Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Wenliang Gao
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Yuanyuan Zhu
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Yaochen Li
- Department of Central Lab, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515041, China
- Correspondence: (Y.L.); (G.Z.)
| | - Guojun Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Correspondence: (Y.L.); (G.Z.)
| |
Collapse
|
15
|
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
|
16
|
The TRPC1 Channel Forms a PI3K/CaM Complex and Regulates Pancreatic Ductal Adenocarcinoma Cell Proliferation in a Ca2+-Independent Manner. Int J Mol Sci 2022; 23:ijms23147923. [PMID: 35887266 PMCID: PMC9323718 DOI: 10.3390/ijms23147923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 02/04/2023] Open
Abstract
Dysregulation of the transient receptor canonical ion channel (TRPC1) has been found in several cancer types, yet the underlying molecular mechanisms through which TRPC1 impacts pancreatic ductal adenocarcinoma (PDAC) cell proliferation are incompletely understood. Here, we found that TRPC1 is upregulated in human PDAC tissue compared to adjacent pancreatic tissue and this higher expression correlates with low overall survival. TRPC1 is, as well, upregulated in the aggressive PDAC cell line PANC-1, compared to a duct-like cell line, and its knockdown (KD) reduced cell proliferation along with PANC-1 3D spheroid growth by arresting cells in the G1/S phase whilst decreasing cyclin A, CDK2, CDK6, and increasing p21CIP1 expression. In addition, the KD of TRPC1 neither affected Ca2+ influx nor store-operated Ca2+ entry (SOCE) and reduced cell proliferation independently of extracellular calcium. Interestingly, TRPC1 interacted with the PI3K-p85α subunit and calmodulin (CaM); both the CaM protein level and AKT phosphorylation were reduced upon TRPC1 KD. In conclusion, our results show that TRPC1 regulates PDAC cell proliferation and cell cycle progression by interacting with PI3K-p85α and CaM through a Ca2+-independent pathway.
Collapse
|
17
|
Shi G, Cui W, Liu Y, Li L, Sun Y, Zhang X, Jiao J. TRPC1 correlates with poor tumor features, radiotherapy efficacy and survival in tongue squamous cell carcinoma. Biomark Med 2022; 16:867-877. [PMID: 35833829 DOI: 10.2217/bmm-2021-0208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The present study aimed to explore the clinical association of TRPC1 with tongue squamous cell carcinoma (TSCC) tumor features and prognosis. Methods: A total of 246 TSCC patients who underwent surgical resection were retrospectively analyzed, and their tissue specimens were acquired for TRPC1 protein and mRNA detection. Results: TRPC1 protein immunohistochemistry score and mRNA expression were of good value in differentiating TSCC tissue from tumor-adjacent tissue and were positively correlated with pathological grade and tumor node metastasis stage. A high TRPC1 protein score was negatively correlated with overall survival, and this correlation was dramatically obvious in patients who received adjuvant radiotherapy. Conclusion: TRPC1 correlates with poor tumor features and unfavorable survival in TSCC patients.
Collapse
Affiliation(s)
- Guang Shi
- Department of Stomatology, HanDan Central Hospital, Hebei Handan, 056000, China
| | - Weiwei Cui
- Department of Stomatology, HanDan Central Hospital, Hebei Handan, 056000, China
| | - Yingqi Liu
- Department of Stomatology, HanDan Central Hospital, Hebei Handan, 056000, China
| | - Lei Li
- Department of Stomatology, HanDan Central Hospital, Hebei Handan, 056000, China
| | - Ying Sun
- Department of Stomatology, HanDan Central Hospital, Hebei Handan, 056000, China
| | - Xueqiang Zhang
- Department of Stomatology, HanDan Central Hospital, Hebei Handan, 056000, China
| | - Jianjun Jiao
- Department of Stomatology, HanDan Central Hospital, Hebei Handan, 056000, China
| |
Collapse
|
18
|
Wang Y, Zhang C. Aberrant TRPC1 expression reflects stromal cervical invasion, lymphovascular invasion, elevated FIGO stage, and poor survival in resectable endometrial carcinoma patients. J Clin Lab Anal 2022; 36:e24560. [PMID: 35754147 PMCID: PMC9396166 DOI: 10.1002/jcla.24560] [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: 05/09/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 11/12/2022] Open
Abstract
Background Transient receptor potential channel 1 (TRPC1) promotes tumor growth and metastasis in endometrial carcinoma (EC) cell lines, whereas its clinical role in EC management remains unclear. Therefore, this study aimed to investigate the association of TRPC1 protein expression with the clinical features and survival of EC patients, then was further validated by TRPC1 mRNA measurement and data from The Human Protein Atlas. Methods TRPC1 protein expression in tumor tissues and normal endometria of 176 resectable EC patients was determined using immunohistochemistry. Besides, TRPC1 mRNA expression of partial patients (n = 80) was detected using RT‐qPCR. Additionally, survival data from The Human Protein Atlas (derived from The Cancer Genome Atlas [TCGA]) was analyzed. Results TRPC1 protein expression was up‐regulated in tumor tissue compared with normal endometrium (p < 0.001). Up‐regulated TRPC1 protein expression was associated with stromal cervical invasion (p = 0.044), lymphovascular invasion (p = 0.032), and increased federation of gynecology and obstetrics (FIGO) stage (p = 0.005). Tumor TRPC1 protein high was linked with shortened accumulating disease‐free survival (DFS) (p = 0.009) and overall survival (OS) (p = 0.026), which were also confirmed by multivariate Cox's regression analysis (both p < 0.050). Further, TRPC1 mRNA validation disclosed that TRPC1 mRNA high was related to shortened accumulating DFS (p = 0.038) and exhibited a correlating trend with declined OS (lacked statistical significance) (p = 0.162). Meanwhile, survival analysis on the data from The Human Protein Atlas (derived from TCGA) also exhibited that TRPC1 mRNA high was correlated with reduced accumulating OS (p < 0.001). Conclusion Our findings support TRPC1 as a prognostic biomarker in resectable EC patients.
Collapse
Affiliation(s)
- Yi Wang
- Gynecology Department, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun Zhang
- Gynecology Department, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Fallah HP, Ahuja E, Lin H, Qi J, He Q, Gao S, An H, Zhang J, Xie Y, Liang D. A Review on the Role of TRP Channels and Their Potential as Drug Targets_An Insight Into the TRP Channel Drug Discovery Methodologies. Front Pharmacol 2022; 13:914499. [PMID: 35685622 PMCID: PMC9170958 DOI: 10.3389/fphar.2022.914499] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/27/2022] [Indexed: 01/13/2023] Open
Abstract
Transient receptor potential (TRP) proteins are a large group of ion channels that control many physiological functions in our body. These channels are considered potential therapeutic drug targets for various diseases such as neurological disorders, cancers, cardiovascular disease, and many more. The Nobel Prize in Physiology/Medicine in the year 2021 was awarded to two scientists for the discovery of TRP and PIEZO ion channels. Improving our knowledge of technologies for their study is essential. In the present study, we reviewed the role of TRP channel types in the control of normal physiological functions as well as disease conditions. Also, we discussed the current and novel technologies that can be used to study these channels successfully. As such, Flux assays for detecting ionic flux through ion channels are among the core and widely used tools for screening drug compounds. Technologies based on these assays are available in fully automated high throughput set-ups and help detect changes in radiolabeled or non-radiolabeled ionic flux. Aurora's Ion Channel Reader (ICR), which works based on label-free technology of flux assay, offers sensitive, accurate, and reproducible measurements to perform drug ranking matching with patch-clamp (gold standard) data. The non-radiolabeled trace-based flux assay coupled with the ICR detects changes in various ion types, including potassium, calcium, sodium, and chloride channels, by using appropriate tracer ions. This technology is now considered one of the very successful approaches for analyzing ion channel activity in modern drug discovery. It could be a successful approach for studying various ion channels and transporters, including the different members of the TRP family of ion channels.
Collapse
Affiliation(s)
| | - Ekta Ahuja
- Aurora Biomed Inc., Vancouver, BC, Canada
| | | | - Jinlong Qi
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Qian He
- Aurora Discovery Inc., Foshan, China
| | - Shan Gao
- Aurora Discovery Inc., Foshan, China
| | | | | | | | - Dong Liang
- Aurora Biomed Inc., Vancouver, BC, Canada
- Aurora Discovery Inc., Foshan, China
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
20
|
Chen L, Shan G, Ge M, Qian H, Xia Y. Transient Receptor Potential Channel 1 Potentially Serves as a Biomarker Indicating T/TNM Stages and Predicting Long-Term Prognosis in Patients With Renal Cell Carcinoma. Front Surg 2022; 9:853310. [PMID: 35548183 PMCID: PMC9081676 DOI: 10.3389/fsurg.2022.853310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/18/2022] [Indexed: 12/27/2022] Open
Abstract
Background Transient receptor potential channel 1 (TRPC1) regulates the progression of several cancers, but its clinical implication in renal cell carcinoma (RCC) has not been explored yet. This study aimed to investigate the correlation of TRPC1 with clinical characteristics and prognosis in patients with RCC. Methods Totally, 177 patients with primary RCC who received surgical resection were retrospectively screened. Their tumor and paired adjacent tissue specimens were retrieved to assess TRPC1 mRNA expression using RT-qPCR and TRPC1 protein expression using immunohistochemistry (IHC). Results Both TRPC1 IHC score and TRPC1 mRNA expression were elevated in RCC tissue than in adjacent tissue (both P < 0.001). Meanwhile, both TRPC1 IHC score and TRPC1 mRNA expression in tumor were associated with higher T stage (both P = 0.02) and TNM stage (P = 0.009, P = 0.003, respectively). However, no correlation was found in tumor TRPC1 IHC score or TRPC1 mRNA expression with other tumor properties (all P > 0.05). Besides, the 3-, 5-, and 7-year overall survival (OS) were 81.4, 68.6, and 60.2%, respectively in patients with high tumor TRPC1 protein, while they were 89.3, 82.7, and 76.7%, respectively in patients with low tumor TRPC1 protein. High (vs. low) TRPC1 protein in the tumor was associated with shorter OS (P = 0.017), while high (vs. low) TRPC1 mRNA in the tumor was not correlated with OS (P = 0.144). By the forward stepwise method, TRPC1 protein expression independently predicted poor OS (P = 0.01, hazard ratio = 2.052). Conclusion TRPC1 serves as a potential biomarker reflecting tumor features and long-term survival profile in patients with RCC.
Collapse
Affiliation(s)
- Liang Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guang Shan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Minghuan Ge
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huijun Qian
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yue Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
21
|
Ke C, Long S. Dysregulated transient receptor potential channel 1 expression and its correlation with clinical features and survival profile in surgical non-small-cell lung cancer patients. J Clin Lab Anal 2022; 36:e24229. [PMID: 35106847 PMCID: PMC8906054 DOI: 10.1002/jcla.24229] [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: 11/19/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Transient receptor potential channel 1 (TRPC1) facilitates the tumor growth, metastasis, and chemoresistance in a series of neoplasms, while its correlation with clinical features and survival profile in NSCLC patients remains elusive. Hence, this study aimed to explore this topic. METHODS Totally, 192 NSCLC patients were enrolled. Protein and mRNA expression of TRPC1 in carcinoma tissue and para-carcinoma tissue were evaluated by immunohistochemistry (IHC) assay and reverse transcription quantitative polymerase chain reaction (RT-qPCR) assay, respectively. RESULTS Immunohistochemistry score and mRNA expression of TRPC1 were higher in carcinoma tissue compared with para-carcinoma tissue (both p < 0.001). Besides, increased TRPC1 IHC score (p = 0.004) and elevated TRPC1 mRNA overexpression (p = 0.016) were linked with occurrence of LYN metastasis; meanwhile, increased TRPC1 IHC score (p = 0.015) and raised TRPC1 mRNA expression (p = 0.009) were also linked with advanced TNM stage, whereas TRPC1 IHC score and TRPC1 mRNA expression were not correlated with other clinical features (all p > 0.05). Additionally, TRPC1 protein high (p = 0.007) and TRPC1 mRNA high (p = 0.015) were correlated with poor disease-free survival (DFS) but not correlated with overall survival (OS). Moreover, multivariate Cox's proportional hazards regression analysis showed that high TRPC1 protein expression (p = 0.046) and advanced TNM stage (p < 0.001) were independently correlated with poor DFS. However, TRPC1 protein and mRNA expression were not linked with OS (both p > 0.05), while poor differentiation (p = 0.003) and advanced TNM stage (p < 0.001) were independently associated with worse OS. CONCLUSIONS TRPC1 is unregulated in NSCLC tissue with its overexpression relating to the occurrence of LYN metastasis and worse DFS in NSCLC patients.
Collapse
Affiliation(s)
- Changjiang Ke
- Department of Respiratory and Critical Care Medicine (Respiratory Medicine), Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Hubei, China.,Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Hubei, China
| | - Shenghua Long
- Department of Respiratory and Critical Care Medicine (Respiratory Medicine), Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Hubei, China.,Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Hubei, China
| |
Collapse
|
22
|
Tai YK, Chan KKW, Fong CHH, Ramanan S, Yap JLY, Yin JN, Yip YS, Tan WR, Koh APF, Tan NS, Chan CW, Huang RYJ, Li JZ, Fröhlich J, Franco-Obregón A. Modulated TRPC1 Expression Predicts Sensitivity of Breast Cancer to Doxorubicin and Magnetic Field Therapy: Segue Towards a Precision Medicine Approach. Front Oncol 2022; 11:783803. [PMID: 35141145 PMCID: PMC8818958 DOI: 10.3389/fonc.2021.783803] [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: 09/27/2021] [Accepted: 12/21/2021] [Indexed: 12/19/2022] Open
Abstract
Chemotherapy is the mainstream treatment modality for invasive breast cancer. Unfortunately, chemotherapy-associated adverse events can result in early termination of treatment. Paradoxical effects of chemotherapy are also sometimes observed, whereby prolonged exposure to high doses of chemotherapeutic agents results in malignant states resistant to chemotherapy. In this study, potential synergism between doxorubicin (DOX) and pulsed electromagnetic field (PEMF) therapy was investigated in: 1) MCF-7 and MDA-MB-231 cells in vitro; 2) MCF-7 tumors implanted onto a chicken chorioallantoic membrane (CAM) and; 3) human patient-derived and MCF-7 and MDA-MB-231 breast cancer xenografts implanted into NOD-SCID gamma (NSG) mice. In vivo, synergism was observed in patient-derived and breast cancer cell line xenograft mouse models, wherein PEMF exposure and DOX administration individually reduced tumor size and increased apoptosis and could be augmented by combined treatments. In the CAM xenograft model, DOX and PEMF exposure also synergistically reduced tumor size as well as reduced Transient Receptor Potential Canonical 1 (TRPC1) channel expression. In vitro, PEMF exposure alone impaired the survival of MCF-7 and MDA-MB-231 cells, but not that of non-malignant MCF10A breast cells; the selective vulnerability of breast cancer cells to PEMF exposure was corroborated in human tumor biopsy samples. Stable overexpression of TRPC1 enhanced the vulnerability of MCF-7 cells to both DOX and PEMF exposure and promoted proliferation, whereas TRPC1 genetic silencing reduced sensitivity to both DOX and PEMF treatments and mitigated proliferation. Chronic exposure to DOX depressed TRPC1 expression, proliferation, and responses to both PEMF exposure and DOX in a manner that was reversible upon removal of DOX. TRPC1 channel overexpression and silencing positively correlated with markers of epithelial-mesenchymal transition (EMT), including SLUG, SNAIL, VIMENTIN, and E-CADHERIN, indicating increased and decreased EMT, respectively. Finally, PEMF exposure was shown to attenuate the invasiveness of MCF-7 cells in correlation with TRPC1 expression. We thus demonstrate that the expression levels of TRPC1 consistently predicted breast cancer sensitivity to DOX and PEMF interventions and positively correlated to EMT status, providing an initial rationale for the use of PEMF-based therapies as an adjuvant to DOX chemotherapy for the treatment of breast cancers characterized by elevated TRPC1 expression levels.
Collapse
Affiliation(s)
- Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore, Singapore
| | - Karen Ka Wing Chan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore, Singapore
| | - Charlene Hui Hua Fong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore, Singapore
| | - Sharanya Ramanan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore, Singapore
| | - Jasmine Lye Yee Yap
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore, Singapore
| | - Jocelyn Naixin Yin
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore, Singapore
| | - Yun Sheng Yip
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Wei Ren Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Angele Pei Fern Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Ching Wan Chan
- Division of General Surgery (Breast Surgery), Department of Surgery, National University Hospital, Singapore, Singapore
- Division of Surgical Oncology, National University Cancer Institute, Singapore, Singapore
| | - Ruby Yun Ju Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jing Ze Li
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jürg Fröhlich
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Fields at Work GmbH, Zürich, Switzerland
- Institute of Electromagnetic Fields , ETH Zürich (Swiss Federal Institute of Technology in Zürich), Zürich, Switzerland
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore, Singapore
- Institute for Health Innovation & Technology (iHealthtech), National University of Singapore, Singapore, Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zürich, Zürich, Switzerland
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- *Correspondence: Alfredo Franco-Obregón,
| |
Collapse
|
23
|
Canales Coutiño B, Mayor R. Reprint of: Mechanosensitive ion channels in cell migration. Cells Dev 2021; 168:203730. [PMID: 34456177 DOI: 10.1016/j.cdev.2021.203730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 11/18/2022]
Abstract
Cellular processes are initiated and regulated by different stimuli, including mechanical forces. Cell membrane mechanosensors represent the first step towards the conversion of mechanical stimuli to a biochemical or electrical response. Mechanosensitive (MS) ion channels form a growing family of ion gating channels that respond to direct physical force or plasma membrane deformations. A number of calcium (Ca2+) permeable MS channels are known to regulate the initiation, direction, and persistence of cell migration during development and tumour progression. While the evidence that links individual MS ion channels to cell migration is growing, a unified analysis of the molecular mechanisms regulated downstream of MS ion channel activation is lacking. In this review, we describe the MS ion channel families known to regulate cell migration. We discuss the molecular mechanisms that act downstream of MS ion channels with an emphasis on Ca2+ mediated processes. Finally, we propose the future directions and impact of MS ion channel activity in the field of cell migration.
Collapse
Affiliation(s)
- Brenda Canales Coutiño
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
24
|
BEKTUR AYKANAT NE, ŞAHİN E, KAÇAR S, BAĞCI R, KARAKAYA Ş, BURUKOĞLU DÖNMEZ D, ŞAHİNTÜRK V. Investigation of the effect of hyperthyroidism on endoplasmic reticulum stress and tran- sient receptor potential canonical 1 channel in the kidney. Turk J Med Sci 2021; 51:1554-1563. [PMID: 33754657 PMCID: PMC8283502 DOI: 10.3906/sag-2007-109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 03/20/2021] [Indexed: 11/07/2022] Open
Abstract
Background/aim Hyperthyroidism is associated with results in increased glomerular filtration rate as well as increased renin-angio- tensin-aldosterone activation. The disturbance of Ca2+ homeostasis in the endoplasmic reticulum (ER) is associated with many diseases, including diabetic nephropathy and hyperthyroidism. Transient receptor potential canonical 1 (TRPC1) channel is the first cloned TRPC family protein. Although it is expressed in many places in the kidney, its function is uncertain. TRPC1 is involved in regulating Ca2+ homeostasis, and its upregulation increases ER Ca2+ level, activates the unfolded protein response, which leads to cellular damage in the kidney. This study investigated the role of TRPC1 in the kidneys of hyperthyroid rats in terms of ER stress markers that are gluco- se-regulated protein 78 (GRP78), activating transcription factor 6 (ATF6), (protein kinase R (PKR)-like endoplasmic reticulum kinase) (PERK), Inositol-requiring enzyme 1 (IRE1). Materials and methods Twenty male rats were assigned into control and hyperthyroid groups (n = 10). Hyperthyroidism was induced by adding 12 mg/L thyroxine into the drinking water of rats for 4 weeks. The serum-free T3 and T4 (fT3, fT4), TSH, blood urea nitrogen (BUN), and creatinine levels were measured. The histochemical analysis of kidney sections for morphological changes and also im- munohistochemical and western blot analysis of kidney sections were performed for GRP78, ATF6, PERK, IRE1, TRPC1 antibodies. Results TSH, BUN, and creatinine levels decreased while fT3 and fT4 levels increased in the hyperthyroid rat. The morphologic analy- sis resulted in the capillary basal membrane thickening in glomeruli and also western blot, and immunohistochemical results showed an increase in TRPC1, GRP78, and ATF6 in the hyperthyroid rat (p < 0.05). Conclusion In conclusion, in our study, we showed for the first time that the relationship between ER stress and TRPC1, and their increased expression caused renal damage in hyperthyroid rats.
Collapse
Affiliation(s)
| | - Erhan ŞAHİN
- Department of Histology and Embryology, Faculty of Medicine, Osmangazi University, AnkaraTurkey
| | - Sedat KAÇAR
- Department of Histology and Embryology, Faculty of Medicine, Osmangazi University, AnkaraTurkey
| | - Rıdvan BAĞCI
- Department of IVF Unit Andrology Laboratory, Adana City Education and Research Hospital, AdanaTurkey
| | - Şerife KARAKAYA
- Department of Histology and Embryology, Faculty of Medicine, Osmangazi University, AnkaraTurkey
| | - Dilek BURUKOĞLU DÖNMEZ
- Department of Histology and Embryology, Faculty of Medicine, Osmangazi University, AnkaraTurkey
| | - Varol ŞAHİNTÜRK
- Department of Histology and Embryology, Faculty of Medicine, Osmangazi University, AnkaraTurkey
| |
Collapse
|
25
|
Cui C, Zhang Y, Liu G, Zhang S, Zhang J, Wang X. Advances in the study of cancer metastasis and calcium signaling as potential therapeutic targets. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:266-291. [PMID: 36046433 PMCID: PMC9400724 DOI: 10.37349/etat.2021.00046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/21/2021] [Indexed: 11/19/2022] Open
Abstract
Metastasis is still the primary cause of cancer-related mortality. However, the underlying mechanisms of cancer metastasis are not yet fully understood. Currently, the epithelial-mesenchymal transition, metabolic remodeling, cancer cell intercommunication and the tumor microenvironment including diverse stromal cells, are reported to affect the metastatic process of cancer cells. Calcium ions (Ca2+) are ubiquitous second messengers that manipulate cancer metastasis by affecting signaling pathways. Diverse transporter/pump/channel-mediated Ca2+ currents form Ca2+ oscillations that can be decoded by Ca2+-binding proteins, which are promising prognostic biomarkers and therapeutic targets of cancer metastasis. This paper presents a review of the advances in research on the mechanisms underlying cancer metastasis and the roles of Ca2+-related signals in these events.
Collapse
Affiliation(s)
- Chaochu Cui
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Gang Liu
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Shuhong Zhang
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Jinghang Zhang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| |
Collapse
|
26
|
Canales Coutiño B, Mayor R. Mechanosensitive ion channels in cell migration. Cells Dev 2021; 166:203683. [PMID: 33994356 PMCID: PMC8240554 DOI: 10.1016/j.cdev.2021.203683] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 01/05/2023]
Abstract
Cellular processes are initiated and regulated by different stimuli, including mechanical forces. Cell membrane mechanosensors represent the first step towards the conversion of mechanical stimuli to a biochemical or electrical response. Mechanosensitive (MS) ion channels form a growing family of ion gating channels that respond to direct physical force or plasma membrane deformations. A number of calcium (Ca2+) permeable MS channels are known to regulate the initiation, direction, and persistence of cell migration during development and tumour progression. While the evidence that links individual MS ion channels to cell migration is growing, a unified analysis of the molecular mechanisms regulated downstream of MS ion channel activation is lacking. In this review, we describe the MS ion channel families known to regulate cell migration. We discuss the molecular mechanisms that act downstream of MS ion channels with an emphasis on Ca2+ mediated processes. Finally, we propose the future directions and impact of MS ion channel activity in the field of cell migration.
Collapse
Affiliation(s)
- Brenda Canales Coutiño
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
27
|
Zhang YQ, Zhang F, Zeng YZ, Chen M, Huang WH, Wu JD, Chen WL, Gao WL, Bai JW, Yang RQ, Zeng HC, Wei XL, Zhang GJ. Mutant p53 and Twist1 Co-Expression Predicts Poor Prognosis and Is an Independent Prognostic Factor in Breast Cancer. Front Oncol 2021; 11:628814. [PMID: 34249678 PMCID: PMC8263931 DOI: 10.3389/fonc.2021.628814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 06/04/2021] [Indexed: 02/05/2023] Open
Abstract
PURPOSE The basic helix-loop-helix transcription factor (bHLH) transcription factor Twist1 plays a key role in embryonic development and tumorigenesis. p53 is a frequently mutated tumor suppressor in cancer. Both proteins play a key and significant role in breast cancer tumorigenesis. However, the regulatory mechanism and clinical significance of their co-expression in this disease remain unclear. The purpose of this study was to analyze the expression patterns of p53 and Twist1 and determine their association with patient prognosis in breast cancer. We also investigated whether their co-expression could be a potential marker for predicting patient prognosis in this disease. METHODS Twist1 and mutant p53 expression in 408 breast cancer patient samples were evaluated by immunohistochemistry. Kaplan-Meier Plotter was used to analyze the correlation between co-expression of Twist1 and wild-type or mutant p53 and prognosis for recurrence-free survival (RFS) and overall survival (OS). Univariate analysis, multivariate analysis, and nomograms were used to explore the independent prognostic factors in disease-free survival (DFS) and OS in this cohort. RESULTS Of the 408 patients enrolled, 237 (58%) had high mutant p53 expression. Two-hundred twenty patients (53.9%) stained positive for Twist1, and 188 cases were Twist1-negative. Furthermore, patients that co-expressed Twist1 and mutant p53 (T+P+) had significantly advanced-stage breast cancer [stage III, 61/89 T+P+ (68.5%) vs. 28/89 T-P- (31.5%); stage II, 63/104 T+P+ (60.6%)vs. 41/104 T-P- (39.4%)]. Co-expression was negatively related to early clinical stage (i.e., stages 0 and I; P = 0.039). T+P+ breast cancer patients also had worse DFS (95% CI = 1.217-7.499, P = 0.017) and OS (95% CI = 1.009-9.272, P = 0.048). Elevated Twist1 and mutant p53 expression predicted shorter RFS in basal-like patients. Univariate and multivariate analysis identified three variables (i.e., lymph node involvement, larger tumor, and T+P+) as independent prognostic factors for DFS. Lymph node involvement and T+P+ were also independent factors for OS in this cohort. The total risk scores and nomograms were reliable for predicting DFS and OS in breast cancer patients. CONCLUSIONS Our results revealed that co-expression of mutant p53 and Twist1 was associated with advanced clinical stage, triple negative breast cancer (TNBC) subtype, distant metastasis, and shorter DFS and OS in breast cancer patients. Furthermore, lymph nodes status and co-expression of Twist1 and mutant p53 were classified as independent factors for DFS and OS in this cohort. Co-evaluation of mutant p53 and Twist1 might be an appropriate tool for predicting breast cancer patient outcome.
Collapse
Affiliation(s)
- Yong-Qu Zhang
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
- Clinical Central Research Core, School of Medicine, Xiang’an Hospital of Xiamen University, Xiamen, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, Xiang’an Hospital of Xiamen University, Xiamen, China
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Fan Zhang
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yun-Zhu Zeng
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Min Chen
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
- Clinical Central Research Core, School of Medicine, Xiang’an Hospital of Xiamen University, Xiamen, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, Xiang’an Hospital of Xiamen University, Xiamen, China
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Wen-He Huang
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Jun-Dong Wu
- Department of Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Wei-Ling Chen
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Wen-Liang Gao
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Jing-Wen Bai
- Department of Medical Oncology, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Rui-Qin Yang
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Huan-Cheng Zeng
- Department of Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xiao-Long Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Guo-Jun Zhang, ; Xiao-Long Wei,
| | - Guo-Jun Zhang
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
- Clinical Central Research Core, School of Medicine, Xiang’an Hospital of Xiamen University, Xiamen, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, Xiang’an Hospital of Xiamen University, Xiamen, China
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Guo-Jun Zhang, ; Xiao-Long Wei,
| |
Collapse
|
28
|
Zeng YZ, Zhang YQ, Chen JY, Zhang LY, Gao WL, Lin XQ, Huang SM, Zhang F, Wei XL. TRPC1 Inhibits Cell Proliferation/Invasion and Is Predictive of a Better Prognosis of Esophageal Squamous Cell Carcinoma. Front Oncol 2021; 11:627713. [PMID: 33854967 PMCID: PMC8039442 DOI: 10.3389/fonc.2021.627713] [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: 11/10/2020] [Accepted: 03/08/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVES In China, over 90% of esophageal cancer (EC) cases are esophageal squamous cell carcinoma (ESCC). ESCC is a frequently malignant tumor with poor prognosis despite the development of comprehensive therapeutic strategies, for which there is still a lack of effective prognostic factors. Previous studies found that the abnormal expression of TRPC1 is closely related to the proliferation, invasion, metastasis, and differentiation of various tumors. However, the relationship between TRPC1 and ESCC is currently unclear. The present study aimed to clarify the clinical significance of TRPC1 and to preliminarily assess the molecular mechanism by which TRPC1 regulates cell proliferation, migration, and invasion in ESCC. MATERIALS AND METHODS Immunohistochemistry (IHC) was used to determine the expression of TRPC1 and Ki-67 in 165 cases of ESCC. The correlations between TRPC1 expression and clinicopathological characteristics were determined, and both univariate and multivariate analyses were utilized to quantify the impact of TRPC1 expression on patient survival. Cell Counting Kit-8, scratch wound healing, and transwell assays were used to determine the effects of TRPC1 on proliferation, migration, and invasion in ESCC in vitro, respectively. RESULTS The positive expression rate of TRPC1 showed significantly decreased in ESCC (45.50%) compared with the levels in normal esophageal mucosa (NEM; 80.80%) and high-grade intraepithelial neoplasia (HGIEN; 63.20%) (P<0.001). Higher expression rate of TRPC1 was associated with low lymph node metastasis (P<0.001), high differentiation (rs = 0.232, P=0.003), and low Ki-67 (rs = -0.492, P<0.001). We further revealed that low expression of TRPC1 was associated with poor prognosis (Disease-free survival, DFS: 95% CI=0.545-0.845, P=0.001; Overall survival, OS: 95% CI=0.553-0.891, P=0.004). Furthermore, we showed that downregulation of TRPC1 promoted the proliferation, migration, and invasion of human esophageal squamous cell carcinoma cell line EC9706 in vitro. In contrast, overexpression of TRPC1 inhibited the proliferation, migration, and invasion of human esophageal squamous cell carcinoma cell line KYSE150 (P<0.01), in a manner at least in part mediated through the AKT/p27 pathway. CONCLUSION TRPC1 inhibited the proliferation, migration, and invasion of EC9706 and KYSE150 cells, at least, in part mediated through the AKT/p27 pathway in vitro. The downregulation of TRPC1 may be one of the most important molecular events in the malignant progression of ESCC. TRPC1 could be a new candidate tumor suppressor gene and a new prognostic factor of ESCC.
Collapse
Affiliation(s)
- Yun-Zhu Zeng
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yong-Qu Zhang
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Jiong-Yu Chen
- Oncological Research Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Li-Ying Zhang
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Wen-Liang Gao
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Xue-Qiong Lin
- Clinical Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Shao-Min Huang
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Fan Zhang
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xiao-Long Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Xiao-Long Wei,
| |
Collapse
|