1
|
Lin J, Wang X, Ma S, Yang D, Li K, Li D, Zeng X. Calcium channels as therapeutic targets in head and neck squamous cell carcinoma: current evidence and clinical trials. Front Oncol 2024; 14:1516357. [PMID: 39759147 PMCID: PMC11695298 DOI: 10.3389/fonc.2024.1516357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) originates from the mucosal epithelium of the oral cavity, pharynx, and larynx, and is marked by high rates of recurrence and metastasis. Calcium signaling is associated with the progression of HNSCC and the development of drug resistance. Changes in calcium ion flow can trigger severe pathophysiological processes, including malignant transformation, tumor proliferation, epithelial-mesenchymal transition, and apoptosis evasion. Calcium channels regulate and facilitate these processes. Remodeling of calcium signaling has become one of the most prevalent adaptive mechanisms in cancer cells. Preclinical and clinical evidence indicates that alterations in calcium signaling are crucial for the progression of HNSCC. This review examines the role of calcium channels in HNSCC development and evaluates current clinical trials targeting these channels to assess the feasibility of calcium signaling-based therapies for HNSCC.
Collapse
Affiliation(s)
| | | | | | | | | | - Dongcai Li
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, China
| | - Xianhai Zeng
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, China
| |
Collapse
|
2
|
Zhao F, Yu W, Hu J, Xia Y, Li Y, Liu S, Liu A, Wang C, Zhang H, Zhang L, Shi J. Hypoxia-induced TRPM7 promotes glycolytic metabolism and progression in hepatocellular carcinoma. Eur J Pharmacol 2024; 974:176601. [PMID: 38677534 DOI: 10.1016/j.ejphar.2024.176601] [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: 10/22/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Hypoxia disrupts glucose metabolism in hepatocellular carcinoma (HCC). Transient receptor potential cation channel, subfamily M, member 7 (TRPM7) plays an ontogenetic role. Thus, we aimed to explore the regulation of TRPM7 by hypoxia-induced factor (HIF) and its underlying mechanisms in HCC. METHODS hypoxia was induced in multiple HCC cells using 1% O2 or CoCl2 treatment, and subsequently blocked using siRNAs targeting HIF-1α or HIF-2α as well as a HIF-1α protein synthesis inhibitor. The levels of HIF-1α and TRPM7 were assessed using quantitative PCR (qPCR) and Western blot analysis. Chromatin immunoprecipitation (ChIP) and luciferase assays were performed to observe the regulation of TRPM7 promoter regions by HIF-1α. A PCR array was utilized to screen glucose metabolism-related enzymes in HEK293 cells overexpressing TRPM7 induced by tetracycline, and then verified in TRPM7-overexpressed huh7 cells. Finally, CCK-8, transwell, scratch and tumor formation experiments in nude mice were conducted to examine the effect of TRPM7 on proliferation and metastasis in HCC. RESULTS Exposure to hypoxia led to increase the levels of TRPM7 and HIF-1α in HCC cells, which were inhibited by HIF-1α siRNA or enhanced by HIF-1α overexpression. HIF-1α directly bound to two hypoxia response elements (HREs) in the TRPM7 promoter. Several glycolytic metabolism-related enzymes, were simultaneously upregulated in HEK293 and huh7 cells overexpressing TRPM7 during hypoxia. In vitro and in vivo experiments demonstrated that TRPM7 promoted the proliferation and metastasis of HCC cells. CONCLUSIONS TRPM7 was directly transcriptionally regulated by HIF-1α, leading to glycolytic metabolic reprogramming and the promotion of HCC proliferation and metastasis in vitro and in vivo. Our findings suggest that TRPM7 might be a potential diagnostic indicator and therapeutic target for HCC.
Collapse
Affiliation(s)
- Fengbo Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Medical School of Nantong University, Nantong, 226001, China
| | - Weili Yu
- Institute of Interdisciplinary Integrative Medicine Research, Medical School of Nantong University, Nantong, 226001, China
| | - Jingyan Hu
- Institute of Interdisciplinary Integrative Medicine Research, Medical School of Nantong University, Nantong, 226001, China
| | - Yi Xia
- Institute of Interdisciplinary Integrative Medicine Research, Medical School of Nantong University, Nantong, 226001, China
| | - YuXuan Li
- Institute of Interdisciplinary Integrative Medicine Research, Medical School of Nantong University, Nantong, 226001, China
| | - Siqi Liu
- Institute of Interdisciplinary Integrative Medicine Research, Medical School of Nantong University, Nantong, 226001, China
| | - Aifen Liu
- Institute of Interdisciplinary Integrative Medicine Research, Medical School of Nantong University, Nantong, 226001, China
| | - Chengniu Wang
- Institute of Interdisciplinary Integrative Medicine Research, Medical School of Nantong University, Nantong, 226001, China
| | - Hong Zhang
- Innovative Drug R&D Center, College of Life Sciences, Huaibei Normal University, Huaibei, Anhui, 235000, China
| | - Lei Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Medical School of Nantong University, Nantong, 226001, China; Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, 12 Shanghai, 200433, China.
| | - Jianwu Shi
- Institute of Interdisciplinary Integrative Medicine Research, Medical School of Nantong University, Nantong, 226001, China.
| |
Collapse
|
3
|
Köles L, Ribiczey P, Szebeni A, Kádár K, Zelles T, Zsembery Á. The Role of TRPM7 in Oncogenesis. Int J Mol Sci 2024; 25:719. [PMID: 38255793 PMCID: PMC10815510 DOI: 10.3390/ijms25020719] [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: 11/28/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
This review summarizes the current understanding of the role of transient receptor potential melastatin-subfamily member 7 (TRPM7) channels in the pathophysiology of neoplastic diseases. The TRPM family represents the largest and most diverse group in the TRP superfamily. Its subtypes are expressed in virtually all human organs playing a central role in (patho)physiological events. The TRPM7 protein (along with TRPM2 and TRPM6) is unique in that it has kinase activity in addition to the channel function. Numerous studies demonstrate the role of TRPM7 chanzyme in tumorigenesis and in other tumor hallmarks such as proliferation, migration, invasion and metastasis. Here we provide an up-to-date overview about the possible role of TRMP7 in a broad range of malignancies such as tumors of the nervous system, head and neck cancers, malignant neoplasms of the upper gastrointestinal tract, colorectal carcinoma, lung cancer, neoplasms of the urinary system, breast cancer, malignant tumors of the female reproductive organs, prostate cancer and other neoplastic pathologies. Experimental data show that the increased expression and/or function of TRPM7 are observed in most malignant tumor types. Thus, TRPM7 chanzyme may be a promising target in tumor therapy.
Collapse
Affiliation(s)
- László Köles
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Polett Ribiczey
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Andrea Szebeni
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
| | - Kristóf Kádár
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
| | - Tibor Zelles
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, H-1083, Budapest, Hungary
| | - Ákos Zsembery
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
| |
Collapse
|
4
|
Ciaglia T, Vestuto V, Bertamino A, González-Muñiz R, Gómez-Monterrey I. On the modulation of TRPM channels: Current perspectives and anticancer therapeutic implications. Front Oncol 2023; 12:1065935. [PMID: 36844925 PMCID: PMC9948629 DOI: 10.3389/fonc.2022.1065935] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/15/2022] [Indexed: 02/11/2023] Open
Abstract
The transient melastatin receptor potential (TRPM) ion channel subfamily functions as cellular sensors and transducers of critical biological signal pathways by regulating ion homeostasis. Some members of TRPM have been cloned from cancerous tissues, and their abnormal expressions in various solid malignancies have been correlated with cancer cell growth, survival, or death. Recent evidence also highlights the mechanisms underlying the role of TRPMs in tumor epithelial-mesenchymal transition (EMT), autophagy, and cancer metabolic reprogramming. These implications support TRPM channels as potential molecular targets and their modulation as an innovative therapeutic approach against cancer. Here, we discuss the general characteristics of the different TRPMs, focusing on current knowledge about the connection between TRPM channels and critical features of cancer. We also cover TRPM modulators used as pharmaceutical tools in biological trials and an indication of the only clinical trial with a TRPM modulator about cancer. To conclude, the authors describe the prospects for TRPM channels in oncology.
Collapse
Affiliation(s)
- Tania Ciaglia
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | - Vincenzo Vestuto
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | - Alessia Bertamino
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | | | | |
Collapse
|
5
|
Liu H, Dilger JP, Lin J. A pan-cancer-bioinformatic-based literature review of TRPM7 in cancers. Pharmacol Ther 2022; 240:108302. [PMID: 36332746 DOI: 10.1016/j.pharmthera.2022.108302] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
TRPM7, a divalent cation-selective channel with kinase domains, has been widely reported to potentially affect cancers. In this study, we conducted multiple bioinformatic analyses based on open databases and reviewed articles that provided evidence for the effects of TRPM7 on cancers. The purposes of this paper are 1) to provide a pan-cancer overview of TRPM7 in cancers; 2) to summarize evidence of TRPM7 effects on cancers; 3) to identify potential future studies of TRPM7 in cancer. Bioinformatics analysis revealed that no cancer-related TRPM7 mutation was found. TRPM7 is aberrantly expressed in most cancer types but the cancer-noncancer expression pattern varies across cancer types. TRPM7 was not associated with survival, TMB, or cancer stemness in most cancer types. TRPM7 affected drug sensitivity and tumor immunity in some cancer types. The in vitro evidence, preclinical in vivo evidence, and clinical evidence for TRPM7 effects on cancers as well as TRPM7 kinase substrate and TRPM7-targeting drugs associated with cancers were summarized to facilitate comparison. We matched the bioinformatics evidence to literature evidence, thereby unveiling potential avenues for future investigation of TRPM7 in cancers. We believe that this paper will help orient research toward important and relevant aspects of the role of TRPM7 in cancers.
Collapse
Affiliation(s)
- Hengrui Liu
- Department of Anesthesiology, Health Science Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - James P Dilger
- Department of Anesthesiology, Health Science Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jun Lin
- Department of Anesthesiology, Health Science Center, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
6
|
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
|
7
|
Zhong T, Zhang W, Guo H, Pan X, Chen X, He Q, Yang B, Ding L. The regulatory and modulatory roles of TRP family channels in malignant tumors and relevant therapeutic strategies. Acta Pharm Sin B 2022; 12:1761-1780. [PMID: 35847486 PMCID: PMC9279634 DOI: 10.1016/j.apsb.2021.11.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/11/2021] [Accepted: 10/19/2021] [Indexed: 02/08/2023] Open
Abstract
Transient receptor potential (TRP) channels are one primary type of calcium (Ca2+) permeable channels, and those relevant transmembrane and intracellular TRP channels were previously thought to be mainly associated with the regulation of cardiovascular and neuronal systems. Nowadays, however, accumulating evidence shows that those TRP channels are also responsible for tumorigenesis and progression, inducing tumor invasion and metastasis. However, the overall underlying mechanisms and possible signaling transduction pathways that TRP channels in malignant tumors might still remain elusive. Therefore, in this review, we focus on the linkage between TRP channels and the significant characteristics of tumors such as multi-drug resistance (MDR), metastasis, apoptosis, proliferation, immune surveillance evasion, and the alterations of relevant tumor micro-environment. Moreover, we also have discussed the expression of relevant TRP channels in various forms of cancer and the relevant inhibitors' efficacy. The chemo-sensitivity of the anti-cancer drugs of various acting mechanisms and the potential clinical applications are also presented. Furthermore, it would be enlightening to provide possible novel therapeutic approaches to counteract malignant tumors regarding the intervention of calcium channels of this type.
Collapse
Key Words
- 4α-PDD, 4α-phorbol-12,13-didecanoate
- ABCB, ATP-binding cassette B1
- AKT, protein kinase B
- ALA, alpha lipoic acid
- AMPK, AMP-activated protein kinase
- APB, aminoethoxydiphenyl borate
- ATP, adenosine triphosphate
- CBD, cannabidiol
- CRAC, Ca2+ release-activated Ca2+ channel
- CaR, calcium-sensing receptor
- CaSR, calcium sensing receptor
- Cancer progression
- DAG, diacylglycerol
- DBTRG, Denver Brain Tumor Research Group
- ECFC, endothelial colony-forming cells
- ECM, enhanced extracellular matrix
- EGF, epidermal growth factor
- EMT, epithelial–mesenchymal transition
- ER, endoplasmic reticulum
- ERK, extracellular signal-regulated kinase
- ETS, erythroblastosis virus E26 oncogene homolog
- FAK, focal adhesion kinase
- GADD, growth arrest and DNA damage-inducible gene
- GC, gastric cancer
- GPCR, G-protein coupled receptor
- GSC, glioma stem-like cells
- GSK, glycogen synthase kinase
- HCC, hepatocellular carcinoma
- HIF, hypoxia-induced factor
- HSC, hematopoietic stem cells
- IP3R, inositol triphosphate receptor
- Intracellular mechanism
- KO, knockout
- LOX, lipoxygenase
- LPS, lipopolysaccharide
- LRP, lipoprotein receptor-related protein
- MAPK, mitogen-activated protein kinase
- MLKL, mixed lineage kinase domain-like protein
- MMP, matrix metalloproteinases
- NEDD4, neural precursor cell expressed, developmentally down-regulated 4
- NFAT, nuclear factor of activated T-cells
- NLRP3, NLR family pyrin domain containing 3
- NO, nitro oxide
- NSCLC, non-small cell lung cancer
- Nrf2, nuclear factor erythroid 2-related factor 2
- P-gp, P-glycoprotein
- PCa, prostate cancer
- PDAC, pancreatic ductal adenocarcinoma
- PHD, prolyl hydroxylases
- PI3K, phosphoinositide 3-kinase
- PKC, protein kinase C
- PKD, polycystic kidney disease
- PLC, phospholipase C
- Programmed cancer cell death
- RNS/ROS, reactive nitrogen species/reactive oxygen species
- RTX, resiniferatoxin
- SMAD, Caenorhabditis elegans protein (Sma) and mothers against decapentaplegic (Mad)
- SOCE, store operated calcium entry
- SOR, soricimed
- STIM1, stromal interaction molecules 1
- TEC, tumor endothelial cells
- TGF, transforming growth factor-β
- TNF-α, tumor necrosis factor-α
- TRP channels
- TRPA/C/M/ML/N/P/V, transient receptor potential ankyrin/canonical/melastatin/mucolipon/NOMPC/polycystin/vanilloid
- Targeted tumor therapy
- Tumor microenvironment
- Tumor-associated immunocytes
- UPR, unfolded protein response
- VEGF, vascular endothelial growth factor
- VIP, vasoactive intestinal peptide
- VPAC, vasoactive intestinal peptide receptor subtype
- mTOR, mammalian target of rapamycin
- pFRG/RTN, parafacial respiratory group/retrotrapezoid nucleus
Collapse
|
8
|
Alanazi R, Nakatogawa H, Wang H, Ji D, Luo Z, Golbourn B, Feng Z, Rutka JT, Sun H. Inhibition of TRPM7 with carvacrol suppresses glioblastoma functions
in vivo. Eur J Neurosci 2022; 55:1483-1491. [DOI: 10.1111/ejn.15647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 11/28/2022]
Affiliation(s)
| | | | - Haitao Wang
- Departments of Surgery
- Departments of Surgery Physiology
| | | | - Zhengwei Luo
- Departments of Surgery
- Departments of Surgery Physiology
| | - Brian Golbourn
- Departments of Cell Biology SickKids Research Institute, The Hospital for Sick Children Toronto Canada
| | | | | | - Hong‐Shuo Sun
- Departments of Surgery
- Departments of Surgery Physiology
- Pharmacology, Temerty Faculty of Medicine
- Leslie Dan Faculty of Pharmacy University of Toronto Toronto Canada
| |
Collapse
|
9
|
Zheng L, Lindsay A, McSweeney K, Aplin J, Forbes K, Smith S, Tunwell R, Mackrill JJ. Ryanodine receptor calcium release channels in trophoblasts and their role in cell migration. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119139. [PMID: 34624436 DOI: 10.1016/j.bbamcr.2021.119139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 09/03/2021] [Accepted: 09/19/2021] [Indexed: 10/20/2022]
Abstract
Trophoblasts are specialized epithelial cells of the placenta that are involved in invasion, communication and the exchange of materials between the mother and fetus. Cytoplasmic Ca2+ ([Ca2+]c) plays critical roles in regulating such processes in other cell types, but relatively little is known about the mechanisms that control this second messenger in trophoblasts. In the current study, the presence of RyRs and their accessory proteins in placental tissues and in the BeWo choriocarcinoma, a model trophoblast cell-line, were examined using immunohistochemistry and Western immunoblotting. Contributions of RyRs to Ca2+ signalling and to random migration in BeWo cells were investigated using fura-2 fluorescent and brightfield videomicroscopy. The effect of RyR inhibition on reorganization of the F-actin cytoskeleton elicited by the hormone angiotensin II, was determined using phalloidin-labelling and confocal microscopy. RyR1 and RyR3 proteins were detected in trophoblasts of human first trimester and term placental villi, along with the accessory proteins triadin and calsequestrin. Similarly, RyR1, RyR3, triadin and calsequestrin were detected in BeWo cells. In this cell-line, activation of RyRs with micromolar ryanodine increased [Ca2+]c, whereas pharmacological inhibition of these channels reduced Ca2+ transients elicited by the peptide hormones angiotensin II, arginine vasopressin and endothelin 1. Angiotensin II increased the velocity, total distance and Euclidean distance of random migration by BeWo cells and these effects were significantly reduced by tetracaine and by inhibitory concentrations of ryanodine. RyRs contribute to reorganization of the F-actin cytoskeleton elicited by angiotensin II, since inhibition of these channels restores the parallelness of these structures to control levels. These findings demonstrate that trophoblasts contain a suite of proteins similar to those in other cell types possessing highly developed Ca2+ signal transduction systems, such as skeletal muscle. They also indicate that these channels regulate the migration of trophoblast cells, a process that plays a key role in development of the placenta.
Collapse
Affiliation(s)
- Limian Zheng
- Department of Physiology, University College Cork, Ireland
| | - Andrew Lindsay
- School of Biochemistry and Cell Biology, University College Cork, Ireland
| | - Kate McSweeney
- Department of Physiology, University College Cork, Ireland
| | - John Aplin
- Maternal and Fetal Health Research Centre, University of Manchester, UK
| | - Karen Forbes
- Maternal and Fetal Health Research Centre, University of Manchester, UK; Leeds Centre for Reproduction and Early Development, University of Leeds, UK
| | - Samantha Smith
- Maternal and Fetal Health Research Centre, University of Manchester, UK
| | - Richard Tunwell
- Division of Biosciences, University College London, Gower Street, London, UK
| | | |
Collapse
|
10
|
Maklad A, Sedeeq M, Milevskiy MJG, Azimi I. Calcium Signalling in Medulloblastoma: An In Silico Analysis of the Expression of Calcium Regulating Genes in Patient Samples. Genes (Basel) 2021; 12:1329. [PMID: 34573310 PMCID: PMC8468187 DOI: 10.3390/genes12091329] [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: 07/05/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 11/16/2022] Open
Abstract
Dysregulation in calcium signalling is implicated in several cancer-associated processes, including cell proliferation, migration, invasion and therapy resistance. Modulators of specific calcium-regulating proteins have been proposed as promising future therapeutic agents for some cancers. Alterations in calcium signalling have been extensively studied in some cancers; however, this area of research is highly underexplored in medulloblastoma (MB), the most common paediatric malignant brain tumour. Current MB treatment modalities are not completely effective and can result in several long-lasting mental complications. Hence, new treatment strategies are needed. In this study, we sought to probe the landscape of calcium signalling regulators to uncover those most likely to be involved in MB tumours. We investigated the expression of calcium signalling regulator genes in MB patients using publicly available datasets. We stratified the expression level of these genes with MB molecular subgroups, tumour metastasis and patient survival to uncover correlations with clinical features. Of particular interest was CACNA1 genes, in which we were able to show a developmentally-driven change in expression within the cerebellum, MB's tissue of origin, highlighting a potential influence on tumour incidence. This study lays a platform for future investigations into molecular regulators of calcium signalling in MB formation and progression.
Collapse
Affiliation(s)
- Ahmed Maklad
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, TAS 7005, Australia; (A.M.); (M.S.)
| | - Mohammed Sedeeq
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, TAS 7005, Australia; (A.M.); (M.S.)
| | - Michael J. G. Milevskiy
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia;
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Iman Azimi
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, TAS 7005, Australia; (A.M.); (M.S.)
| |
Collapse
|
11
|
Gong H, Bandura J, Wang GL, Feng ZP, Sun HS. Xyloketal B: A marine compound with medicinal potential. Pharmacol Ther 2021; 230:107963. [PMID: 34375691 DOI: 10.1016/j.pharmthera.2021.107963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 06/01/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022]
Abstract
In recent decades, technological advantages have allowed scientists to isolate medicinal compounds from marine organisms that exhibit unique structure and bioactivity. The mangrove fungus Xylaria sp. from the South China Sea is rich in metabolites and produces a potent therapeutic compound, xyloketal B. Since its isolation in 2001, xyloketal B has been extensively studied in a wide variety of cell types and in vitro and in vivo disease models. Xyloketal B and its derivatives exhibit cytoprotective effects in cardiovascular and neurodegenerative diseases by reducing oxidative stress, regulating the apoptosis pathway, maintaining ionic balance, mitigating inflammatory responses, and preventing protein aggregation. Xyloketal B has also shown to alleviate lipid accumulation in a non-alcoholic fatty liver disease model. Moreover, xyloketal B treatment induces glioblastoma cell death. This review summarizes our current understanding of xyloketal B in various disease models.
Collapse
Affiliation(s)
- Haifan Gong
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Julia Bandura
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Guan-Lei Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory of Functional Molecules from Oceanic Microorganisms (Sun Yat-Sen University), Department of Education of Guangdong Province, 510080, China.
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Hong-Shuo Sun
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada.
| |
Collapse
|
12
|
Arbabian A, Iftinca M, Altier C, Singh PP, Isambert H, Coscoy S. Mutations in calmodulin-binding domains of TRPV4/6 channels confer invasive properties to colon adenocarcinoma cells. Channels (Austin) 2021; 14:101-109. [PMID: 32186440 PMCID: PMC7153789 DOI: 10.1080/19336950.2020.1740506] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Transient receptor potential (TRP) channels form a family of polymodal cation channels gated by thermal, mechanical, or chemical stimuli, with many of them involved in the control of proliferation, apoptosis, or cell cycle. From an evolutionary point of view, TRP family is characterized by high conservation of duplicated genes originating from whole-genome duplication at the onset of vertebrates. The conservation of such “ohnolog” genes is theoretically linked to an increased probability of generating phenotypes deleterious for the organism upon gene mutation. We aimed to test experimentally the hypothesis that TRP mutations, in particular gain-of-function, could be involved in the generation of deleterious phenotypes involved in cancer, such as gain of invasiveness. Indeed, a number of TRP channels have been linked to cancer progression, and exhibit changes in expression levels in various types of cancers. However, TRP mutations in cancer have been poorly documented. We focused on 2 TRPV family members, TRPV4 and TRPV6, and studied the effect of putative gain-of-function mutations on invasiveness properties. TRPV channels have a C-terminal calmodulin-binding domain (CaMBD) that has important functions for regulating protein function, through different mechanisms depending on the channel (channel inactivation/potentiation, cytoskeleton regulation). We studied the effect of mutations mimicking constitutive phosphorylation in TRPV4 and TRPV6 CaMBDs: TRPV4 S823D, S824D and T813D, TRPV6 S691D, S692D and T702. We found that most of these mutants induced a strong gain of invasiveness of colon adenocarcinoma SW480 cells, both for TRPV4 and TRPV6. While increased invasion with TRPV6 S692D and T702D mutants was correlated to increased mutant channel activity, it was not the case for TRPV4 mutants, suggesting different mechanisms with the same global effect of gain in deleterious phenotype. This highlights the potential importance to search for TRP mutations involved in cancer.
Collapse
Affiliation(s)
- Atousa Arbabian
- Laboratoire Physico Chimie Curie, Institut Curie, CNRS UMR168, PSL Research University, Paris, France.,Sorbonne Université, Paris, France
| | - Mircea Iftinca
- Department of Physiology and Pharmacology. Inflammation Research Network, Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute (ACHRI), University of Calgary, Calgary, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology. Inflammation Research Network, Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute (ACHRI), University of Calgary, Calgary, Canada
| | - Param Priya Singh
- Laboratoire Physico Chimie Curie, Institut Curie, CNRS UMR168, PSL Research University, Paris, France.,Sorbonne Université, Paris, France
| | - Hervé Isambert
- Laboratoire Physico Chimie Curie, Institut Curie, CNRS UMR168, PSL Research University, Paris, France.,Sorbonne Université, Paris, France
| | - Sylvie Coscoy
- Laboratoire Physico Chimie Curie, Institut Curie, CNRS UMR168, PSL Research University, Paris, France.,Sorbonne Université, Paris, France.,Equipe Labellisée « Ligue contre le Cancer »
| |
Collapse
|
13
|
Liang X, Zhang N, Pan H, Xie J, Han W. Development of Store-Operated Calcium Entry-Targeted Compounds in Cancer. Front Pharmacol 2021; 12:688244. [PMID: 34122115 PMCID: PMC8194303 DOI: 10.3389/fphar.2021.688244] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/17/2021] [Indexed: 11/17/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) is the major pathway of Ca2+ entry in mammalian cells, and regulates a variety of cellular functions including proliferation, motility, apoptosis, and death. Accumulating evidence has indicated that augmented SOCE is related to the generation and development of cancer, including tumor formation, proliferation, angiogenesis, metastasis, and antitumor immunity. Therefore, the development of compounds targeting SOCE has been proposed as a potential and effective strategy for use in cancer therapy. In this review, we summarize the current research on SOCE inhibitors and blockers, discuss their effects and possible mechanisms of action in cancer therapy, and induce a new perspective on the treatment of cancer.
Collapse
Affiliation(s)
- Xiaojing Liang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ningxia Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiansheng Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
Store Operated Calcium Entry in Cell Migration and Cancer Metastasis. Cells 2021; 10:cells10051246. [PMID: 34069353 PMCID: PMC8158756 DOI: 10.3390/cells10051246] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/12/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023] Open
Abstract
Ca2+ signaling is ubiquitous in eukaryotic cells and modulates many cellular events including cell migration. Directional cell migration requires the polarization of both signaling and structural elements. This polarization is reflected in various Ca2+ signaling pathways that impinge on cell movement. In particular, store-operated Ca2+ entry (SOCE) plays important roles in regulating cell movement at both the front and rear of migrating cells. SOCE represents a predominant Ca2+ influx pathway in non-excitable cells, which are the primary migrating cells in multicellular organisms. In this review, we summarize the role of Ca2+ signaling in cell migration with a focus on SOCE and its diverse functions in migrating cells and cancer metastasis. SOCE has been implicated in regulating focal adhesion turnover in a polarized fashion and the mechanisms involved are beginning to be elucidated. However, SOCE is also involved is other aspects of cell migration with a less well-defined mechanistic understanding. Therefore, much remains to be learned regarding the role and regulation of SOCE in migrating cells.
Collapse
|
15
|
Nadolni W, Immler R, Hoelting K, Fraticelli M, Ripphahn M, Rothmiller S, Matsushita M, Boekhoff I, Gudermann T, Sperandio M, Zierler S. TRPM7 Kinase Is Essential for Neutrophil Recruitment and Function via Regulation of Akt/mTOR Signaling. Front Immunol 2021; 11:606893. [PMID: 33658993 PMCID: PMC7917126 DOI: 10.3389/fimmu.2020.606893] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/22/2020] [Indexed: 12/22/2022] Open
Abstract
During inflammation, neutrophils are one of the first responding cells of innate immunity, contributing to a fast clearance of infection and return to homeostasis. However, excessive neutrophil infiltration accelerates unsolicited disproportionate inflammation for instance in autoimmune diseases such as rheumatoid arthritis. The transient-receptor-potential channel-kinase TRPM7 is an essential regulator of immune system homeostasis. Naïve murine T cells with genetic inactivation of the TRPM7 enzyme, due to a point mutation at the active site, are unable to differentiate into pro-inflammatory T cells, whereas regulatory T cells develop normally. Moreover, TRPM7 is vital for lipopolysaccharides (LPS)-induced activation of murine macrophages. Within this study, we show that the channel-kinase TRPM7 is functionally expressed in neutrophils and has an important impact on neutrophil recruitment during inflammation. We find that human neutrophils cannot transmigrate along a CXCL8 chemokine gradient or produce reactive oxygen species in response to gram-negative bacterial lipopolysaccharide LPS, if TRPM7 channel or kinase activity are blocked. Using a recently identified TRPM7 kinase inhibitor, TG100-115, as well as murine neutrophils with genetic ablation of the kinase activity, we confirm the importance of both TRPM7 channel and kinase function in murine neutrophil transmigration and unravel that TRPM7 kinase affects Akt1/mTOR signaling thereby regulating neutrophil transmigration and effector function. Hence, TRPM7 represents an interesting potential target to treat unwanted excessive neutrophil invasion.
Collapse
Affiliation(s)
- Wiebke Nadolni
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Roland Immler
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Kilian Hoelting
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marco Fraticelli
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Myriam Ripphahn
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Simone Rothmiller
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany
| | - Masayuki Matsushita
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Ingrid Boekhoff
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Markus Sperandio
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Susanna Zierler
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany.,Institute of Pharmacology, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|
16
|
He F, Yu J, Yang J, Wang S, Zhuang A, Shi H, Gu X, Xu X, Chai P, Jia R. m 6A RNA hypermethylation-induced BACE2 boosts intracellular calcium release and accelerates tumorigenesis of ocular melanoma. Mol Ther 2021; 29:2121-2133. [PMID: 33601055 DOI: 10.1016/j.ymthe.2021.02.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/22/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Ocular melanoma, including uveal melanoma (UM) and conjunctival melanoma (CM), is the most common and deadly eye cancer in adults. Both UM and CM originate from melanocytes and exhibit an aggressive growth pattern with high rates of metastasis and mortality. The integral membrane glycoprotein beta-secretase 2 (BACE2), an enzyme that cleaves amyloid precursor protein into amyloid beta peptide, has been reported to play a vital role in vertebrate pigmentation and metastatic melanoma. However, the role of BACE2 in ocular melanoma remains unclear. In this study, we showed that BACE2 was significantly upregulated in ocular melanoma, and inhibition of BACE2 significantly impaired tumor progression both in vitro and in vivo. Notably, we identified that transmembrane protein 38B (TMEM38B), whose expression was highly dependent on BACE2, modulated calcium release from endoplasmic reticulum (ER). Inhibition of the BACE2/TMEM38B axis could trigger exhaustion of intracellular calcium release and inhibit tumor progression. We further demonstrated that BACE2 presented an increased level of N6-methyladenosine (m6A) RNA methylation, which led to the upregulation of BACE2 mRNA. To our knowledge, this study provides a novel pattern of BACE2-mediated intracellular calcium release in ocular melanoma progression, and our findings suggest that m6A/BACE2/TMEM38b could be a potential therapeutic axis for ocular melanoma.
Collapse
Affiliation(s)
- Fanglin He
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Jie Yu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Jie Yang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Shaoyun Wang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Ai Zhuang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Hanhan Shi
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Xiang Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Xiaofang Xu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Peiwei Chai
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China.
| |
Collapse
|
17
|
Huang SY, Chen Y, Tan XR, Gong S, Yang XJ, He QM, He SW, Liu N, Li YQ. Serum Calcium Levels Before Antitumour Therapy Predict Clinical Outcomes in Patients with Nasopharyngeal Carcinoma. Onco Targets Ther 2021; 13:13111-13119. [PMID: 33380801 PMCID: PMC7767708 DOI: 10.2147/ott.s275613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/09/2020] [Indexed: 01/19/2023] Open
Abstract
PURPOSE The prognostic value of serum calcium levels in nasopharyngeal carcinoma (NPC) remains unknown. This study aimed to evaluate the prognostic value of serum calcium levels in patients with NPC. PATIENTS AND METHODS A total of 2094 patients diagnosed with NPC between April 2009 and September 2012 were enrolled in this retrospective analysis. The median follow-up time was 96.3 months (range: 4.1-120.0 months). Univariate and multivariable Cox proportional hazards models were used to identify significant and independent prognostic predictors of overall survival (OS), disease-free survival (DFS), distant metastasis-free survival (DMFS), and relapse-free survival (RFS). RESULTS Overall, low serum calcium levels were detected in 1109/2094 (53.00%) patients and tended to be more frequently detected in older (P<0.001) and female (P=0.001) patients. Patients with low serum calcium levels had poorer OS (P=0.011), DFS (P=0.012) and DMFS (P=0.004) than those with high serum calcium levels, but serum calcium levels had no significant effect on RFS (P=0.376). In univariate and multivariable analyses, low serum calcium levels were a statistically significant and independent prognostic factor for OS, DFS, and DMFS but had no prognostic value for RFS. CONCLUSION Serum calcium levels can serve as a prognostic predictor and guide more individualized treatment for NPC patients.
Collapse
Affiliation(s)
- Sheng-Yan Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, People's Republic of China
| | - Yang Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, People's Republic of China
| | - Xi-Rong Tan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, People's Republic of China
| | - Sha Gong
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, People's Republic of China
| | - Xiao-Jing Yang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, People's Republic of China
| | - Qing-Mei He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, People's Republic of China
| | - Shi-Wei He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, People's Republic of China
| | - Na Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, People's Republic of China
| | - Ying-Qing Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, People's Republic of China
| |
Collapse
|
18
|
Adiga D, Radhakrishnan R, Chakrabarty S, Kumar P, Kabekkodu SP. The Role of Calcium Signaling in Regulation of Epithelial-Mesenchymal Transition. Cells Tissues Organs 2020; 211:134-156. [PMID: 33316804 DOI: 10.1159/000512277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 10/13/2020] [Indexed: 11/19/2022] Open
Abstract
Despite substantial advances in the field of cancer therapeutics, metastasis is a significant challenge for a favorable clinical outcome. Epithelial to mesenchymal transition (EMT) is a process of acquiring increased motility, invasiveness, and therapeutic resistance by cancer cells for their sustained growth and survival. A plethora of intrinsic mechanisms and extrinsic microenvironmental factors drive the process of cancer metastasis. Calcium (Ca2+) signaling plays a critical role in dictating the adaptive metastatic cell behavior comprising of cell migration, invasion, angiogenesis, and intravasation. By modulating EMT, Ca2+ signaling can regulate the complexity and dynamics of events leading to metastasis. This review summarizes the role of Ca2+ signal remodeling in the regulation of EMT and metastasis in cancer.
Collapse
Affiliation(s)
- Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, India
| | - Prashant Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India,
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, India,
| |
Collapse
|
19
|
Abstract
Transient receptor potential (TRP) channels comprise a diverse family of ion channels, the majority of which are calcium permeable and show sophisticated regulatory patterns in response to various environmental cues. Early studies led to the recognition of TRP channels as environmental and chemical sensors. Later studies revealed that TRP channels mediated the regulation of intracellular calcium. Mutations in TRP channel genes result in abnormal regulation of TRP channel function or expression, and interfere with normal spatial and temporal patterns of intracellular local Ca2+ distribution. The resulting dysregulation of multiple downstream effectors, depending on Ca2+ homeostasis, is associated with hallmarks of cancer pathophysiology, including enhanced proliferation, survival and invasion of cancer cells. These findings indicate that TRP channels affect multiple events that control cellular fate and play a key role in cancer progression. This review discusses the accumulating evidence supporting the role of TRP channels in tumorigenesis, with emphasis on prostate cancer. [BMB Reports 2020; 53(3): 125-132].
Collapse
Affiliation(s)
- Dongki Yang
- Departments of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea
| | - Jaehong Kim
- Departments of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea
| |
Collapse
|
20
|
Wong R, Gong H, Alanazi R, Bondoc A, Luck A, Sabha N, Horgen FD, Fleig A, Rutka JT, Feng ZP, Sun HS. Inhibition of TRPM7 with waixenicin A reduces glioblastoma cellular functions. Cell Calcium 2020; 92:102307. [PMID: 33080445 DOI: 10.1016/j.ceca.2020.102307] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/27/2020] [Accepted: 10/04/2020] [Indexed: 12/28/2022]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumour originating in the CNS. Median patient survival is <15 months with standard treatment which consists of surgery alongside radiation therapy and temozolomide chemotherapy. However, because of the aggressive nature of GBM, and the significant toxicity of these adjuvant therapies, long-term therapeutic effects are unsatisfactory. Thus, there is urgency to identify new drug targets for GBM. Recent evidence shows that the transient receptor potential melastatin 7 (TRPM7) cation channel is aberrantly upregulated in GBM and its inhibition leads to reduction of GBM cellular functions. This suggests that TRPM7 may be a potential drug target for GBM treatment. In this study, we assessed the effects of the specific TRPM7 antagonist waixenicin A on human GBM cell lines U87 or U251 both in vitro and in vivo. First, we demonstrated in vitro that application of waixenicin A reduced TRPM7 protein expression and inhibited the TRPM7-like currents in GBM cells. We also observed reduction of GBM cell viability, migration, and invasion. Using an intracranial xenograft GBM mouse model, we showed that with treatment of waixenicin A, there was increased cleaved caspase 3 activity, alongside reduction in Ki-67, cofilin, and Akt activity in vivo. Together, these data demonstrate higher GBM cell apoptosis, and lower proliferation, migration, invasion and survivability following treatment with waixenicin A.
Collapse
Affiliation(s)
- Raymond Wong
- Departments of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Haifan Gong
- Departments of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Rahmah Alanazi
- Departments of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Andrew Bondoc
- Departments of Cell Biology SickKids Research Institute, The Hospital for Sick Children, Toronto, Canada
| | - Amanda Luck
- Departments of Cell Biology SickKids Research Institute, The Hospital for Sick Children, Toronto, Canada
| | - Nesrin Sabha
- Departments of Genetics and Genome Biology, SickKids Research Institute, The Hospital for Sick Children, Toronto, Canada
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, Hawaii, 96744, USA
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, Hawaii, 96720, USA
| | - James T Rutka
- Departments of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Hong-Shuo Sun
- Departments of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada.
| |
Collapse
|
21
|
Mapping TRPM7 Function by NS8593. Int J Mol Sci 2020; 21:ijms21197017. [PMID: 32977698 PMCID: PMC7582524 DOI: 10.3390/ijms21197017] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
The transient receptor potential cation channel, subfamily M, member 7 (TRPM7) is a ubiquitously expressed membrane protein, which forms a channel linked to a cytosolic protein kinase. Genetic inactivation of TRPM7 in animal models uncovered the critical role of TRPM7 in early embryonic development, immune responses, and the organismal balance of Zn2+, Mg2+, and Ca2+. TRPM7 emerged as a new therapeutic target because malfunctions of TRPM7 have been associated with anoxic neuronal death, tissue fibrosis, tumour progression, and giant platelet disorder. Recently, several laboratories have identified pharmacological compounds allowing to modulate either channel or kinase activity of TRPM7. Among other small molecules, NS8593 has been defined as a potent negative gating regulator of the TRPM7 channel. Consequently, several groups applied NS8593 to investigate cellular pathways regulated by TRPM7. Here, we summarize the progress in this research area. In particular, two notable milestones have been reached in the assessment of TRPM7 druggability. Firstly, several laboratories demonstrated that NS8593 treatment reliably mirrors prominent phenotypes of cells manipulated by genetic inactivation of TRPM7. Secondly, it has been shown that NS8593 allows us to probe the therapeutic potential of TRPM7 in animal models of human diseases. Collectively, these studies employing NS8593 may serve as a blueprint for the preclinical assessment of TRPM7-targeting drugs.
Collapse
|
22
|
Pethő Z, Najder K, Carvalho T, McMorrow R, Todesca LM, Rugi M, Bulk E, Chan A, Löwik CWGM, Reshkin SJ, Schwab A. pH-Channeling in Cancer: How pH-Dependence of Cation Channels Shapes Cancer Pathophysiology. Cancers (Basel) 2020; 12:E2484. [PMID: 32887220 PMCID: PMC7565548 DOI: 10.3390/cancers12092484] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/20/2022] Open
Abstract
Tissue acidosis plays a pivotal role in tumor progression: in particular, interstitial acidosis promotes tumor cell invasion, and is a major contributor to the dysregulation of tumor immunity and tumor stromal cells. The cell membrane and integral membrane proteins commonly act as important sensors and transducers of altered pH. Cell adhesion molecules and cation channels are prominent membrane proteins, the majority of which is regulated by protons. The pathophysiological consequences of proton-sensitive ion channel function in cancer, however, are scarcely considered in the literature. Thus, the main focus of this review is to highlight possible events in tumor progression and tumor immunity where the pH sensitivity of cation channels could be of great importance.
Collapse
Affiliation(s)
- Zoltán Pethő
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Karolina Najder
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Tiago Carvalho
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
| | - Luca Matteo Todesca
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Micol Rugi
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Etmar Bulk
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Alan Chan
- Percuros B.V., 2333 CL Leiden, The Netherlands;
| | - Clemens W. G. M. Löwik
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
- Department of Oncology CHUV, UNIL and Ludwig Cancer Center, 1011 Lausanne, Switzerland
| | - Stephan J. Reshkin
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| |
Collapse
|
23
|
Kiss F, Pohóczky K, Szállási A, Helyes Z. Transient Receptor Potential (TRP) Channels in Head-and-Neck Squamous Cell Carcinomas: Diagnostic, Prognostic, and Therapeutic Potentials. Int J Mol Sci 2020; 21:E6374. [PMID: 32887395 PMCID: PMC7569891 DOI: 10.3390/ijms21176374] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/24/2022] Open
Abstract
Head-and-neck squamous cell carcinomas (HNSCC) remain a leading cause of cancer morbidity and mortality worldwide. This is a largely preventable disease with smoking, alcohol abuse, and human papilloma virus (HPV) being the main risk factors. Yet, many patients are diagnosed with advanced disease, and no survival improvement has been seen for oral SCC in the past decade. Clearly, new diagnostic and prognostic markers are needed for early diagnosis and to guide therapy. Gene expression studies implied the involvement of transient receptor potential (TRP) channels in the pathogenesis of HNSCC. TRPs are expressed in normal epithelium where they play a key role in proliferation and differentiation. There is increasing evidence that the expression of TRP channels may change in HNSCC with important implications for diagnosis, prognosis, and therapy. In this review, we propose that TRP channel expression may afford a novel opportunity for early diagnosis of HNSCC and targeted molecular treatment.
Collapse
Affiliation(s)
- Fruzsina Kiss
- Somogy County Kaposi Mór Teaching Hospital, H-7400 Kaposvár, Hungary;
| | - Krisztina Pohóczky
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary;
- János Szentágothai Research Centre, Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Arpad Szállási
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary;
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary;
- János Szentágothai Research Centre, Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- PharmInVivo Ltd., H-7629 Pécs, Hungary
| |
Collapse
|
24
|
Bruce JIE, James AD. Targeting the Calcium Signalling Machinery in Cancer. Cancers (Basel) 2020; 12:cancers12092351. [PMID: 32825277 PMCID: PMC7565467 DOI: 10.3390/cancers12092351] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/30/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer is caused by excessive cell proliferation and a propensity to avoid cell death, while the spread of cancer is facilitated by enhanced cellular migration, invasion, and vascularization. Cytosolic Ca2+ is central to each of these important processes, yet to date, there are no cancer drugs currently being used clinically, and very few undergoing clinical trials, that target the Ca2+ signalling machinery. The aim of this review is to highlight some of the emerging evidence that targeting key components of the Ca2+ signalling machinery represents a novel and relatively untapped therapeutic strategy for the treatment of cancer.
Collapse
Affiliation(s)
- Jason I. E. Bruce
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
- Correspondence: ; Tel.: +44-(0)-161-275-5484
| | - Andrew D. James
- Department of Biology, University of York, Heslington, York YO10 5DD, UK;
| |
Collapse
|
25
|
Hu W, Yao W, Li H, Chen L. MiR-30e-5p inhibits the migration and invasion of nasopharyngeal carcinoma via regulating the expression of MTA1. Biosci Rep 2020; 40:BSR20194309. [PMID: 32458989 PMCID: PMC7253402 DOI: 10.1042/bsr20194309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/14/2020] [Accepted: 04/21/2020] [Indexed: 11/17/2022] Open
Abstract
The study explored the effect of miR-30e-5p on nasopharyngeal carcinoma (NPC). MiR-30e-5p levels in NPC cancer and adjacent normal samples, in metastatic and non-metastatic cancer samples of NPC, and in NP69 cell and five NPC cell lines were determined by quantitative real-time polymerase chain reaction (qRT-PCR). The relationship between miR-30e-5p and MTA1 was confirmed by dual-luciferase reporter assay, Western blot and qRT-PCR. The viability, migration and invasion of 5-8F and 6-10B cells were determined by CCK-8, scratch test and transwell assays, respectively. The levels of migration-related proteins (vimentin and Snail) and invasion-related proteins (MMP2 and MMP3) in NPC cells were detected by Western blot. The results showed that low expression of miR-30e-5p was associated with HNSC cancer, NPC, metastasis of NPC and NPC cell lines. Overexpressed miR-30e-5p in HNSC cancer and NPC was predictive of a better prognosis of patients. In addition, the viability, migration and invasion were reduced by up-regulating miR-30e-5p in 5-8F cells, but promoted by down-regulated miR-30e-5p in 6-10B cells. MiR-30e-5p reversed the migration and invasion of NPC cells regulated by MTA1, and inhibited migration and invasion of NPC cells via regulating MTA1 expression.
Collapse
Affiliation(s)
- Weiqun Hu
- Department of Otorhinolaryngology, The Affiliated Hospital of Putian University, China
| | - Wenfeng Yao
- Department of Otorhinolaryngology, Xinxiang First People’s Hospital, China
| | - Haolin Li
- Department of Otorhinolaryngology, Xinxiang First People’s Hospital, China
| | - Li Chen
- Department of Otorhinolaryngology, Zaozhuang Municipal Hospital, China
| |
Collapse
|
26
|
Morishita K, Watanabe K, Ichijo H. Cell volume regulation in cancer cell migration driven by osmotic water flow. Cancer Sci 2019; 110:2337-2347. [PMID: 31120184 PMCID: PMC6676112 DOI: 10.1111/cas.14079] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer metastasis is the most frequent cause of death for patients with cancer. The main current treatment for cancer metastasis is chemotherapy targeting cancer cells’ ability to proliferate. However, some types of cancer cells show resistance to chemotherapy. Recently, cancer cell migration has become the subject of interest as a novel target of cancer therapy. Cell migration requires many factors, such as the cytoskeleton, cell‐matrix adhesion and cell volume regulation. Here, we focus on cell volume regulation and the role of ion/water transport systems in cell migration. Transport proteins, such as ion channels, ion carriers, and aquaporins, are indispensable for cell volume regulation under steady‐state conditions and during exposure to osmotic stress. Studies from the last ~25 years have revealed that cell volume regulation also plays an important role in the process of cell migration. Water flow in accordance with localized osmotic gradients generated by ion transport contributes to the driving force for cell migration. Moreover, it has been reported that metastatic cancer cells have higher expression of these transport proteins than nonmetastatic cancer cells. Thus, ion/water transport proteins involved in cell volume regulation and cell migration could be novel therapeutic targets for cancer metastasis. In this review, after presenting the importance of ion/water transport systems in cell volume regulation, we discuss the roles of transport proteins in a pathophysiological context, especially in the context of cancer cell migration.
Collapse
Affiliation(s)
- Kazuhiro Morishita
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kengo Watanabe
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
27
|
Canales J, Morales D, Blanco C, Rivas J, Díaz N, Angelopoulos I, Cerda O. A TR(i)P to Cell Migration: New Roles of TRP Channels in Mechanotransduction and Cancer. Front Physiol 2019; 10:757. [PMID: 31275168 PMCID: PMC6591513 DOI: 10.3389/fphys.2019.00757] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/31/2019] [Indexed: 12/20/2022] Open
Abstract
Cell migration is a key process in cancer metastasis, allowing malignant cells to spread from the primary tumor to distant organs. At the molecular level, migration is the result of several coordinated events involving mechanical forces and cellular signaling, where the second messenger Ca2+ plays a pivotal role. Therefore, elucidating the regulation of intracellular Ca2+ levels is key for a complete understanding of the mechanisms controlling cellular migration. In this regard, understanding the function of Transient Receptor Potential (TRP) channels, which are fundamental determinants of Ca2+ signaling, is critical to uncovering mechanisms of mechanotransduction during cell migration and, consequently, in pathologies closely linked to it, such as cancer. Here, we review recent studies on the association between TRP channels and migration-related mechanotransduction events, as well as in the involvement of TRP channels in the migration-dependent pathophysiological process of metastasis.
Collapse
Affiliation(s)
- Jimena Canales
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - Diego Morales
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - Constanza Blanco
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - José Rivas
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - Nicolás Díaz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - Ioannis Angelopoulos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
28
|
Pethő Z, Najder K, Bulk E, Schwab A. Mechanosensitive ion channels push cancer progression. Cell Calcium 2019; 80:79-90. [PMID: 30991298 DOI: 10.1016/j.ceca.2019.03.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023]
Abstract
In many cases, the mechanical properties of a tumor are different from those of the host tissue. Mechanical cues regulate cancer development by affecting both tumor cells and their microenvironment, by altering cell migration, proliferation, extracellular matrix remodeling and metastatic spread. Cancer cells sense mechanical stimuli such as tissue stiffness, shear stress, tissue pressure of the extracellular space (outside-in mechanosensation). These mechanical cues are transduced into a cellular response (e. g. cell migration and proliferation; inside-in mechanotransduction) or to a response affecting the microenvironment (e. g. inducing a fibrosis or building up growth-induced pressure; inside-out mechanotransduction). These processes heavily rely on mechanosensitive membrane proteins, prominently ion channels. Mechanosensitive ion channels are involved in the Ca2+-signaling of the tumor and stroma cells, both directly, by mediating Ca2+ influx (e. g. Piezo and TRP channels), or indirectly, by maintaining the electrochemical gradient necessary for Ca2+ influx (e. g. K2P, KCa channels). This review aims to discuss the diverse roles of mechanosenstive ion channels in cancer progression, especially those involved in Ca2+-signaling, by pinpointing their functional relevance in tumor pathophysiology.
Collapse
Affiliation(s)
- Zoltán Pethő
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany.
| | - Karolina Najder
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Etmar Bulk
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Albrecht Schwab
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| |
Collapse
|
29
|
Liao J, Ng SH, Luk AC, Suen HC, Qian Y, Lee AWT, Tu J, Fung JCL, Tang NLS, Feng B, Chan WY, Fouchet P, Hobbs RM, Lee TL. Revealing cellular and molecular transitions in neonatal germ cell differentiation using single cell RNA sequencing. Development 2019; 146:dev174953. [PMID: 30824552 DOI: 10.1242/dev.174953] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/17/2019] [Indexed: 12/22/2022]
Abstract
Neonatal germ cell development provides the foundation of spermatogenesis. However, a systematic understanding of this process is still limited. To resolve cellular and molecular heterogeneity in this process, we profiled single cell transcriptomes of undifferentiated germ cells from neonatal mouse testes and employed unbiased clustering and pseudotime ordering analysis to assign cells to distinct cell states in the developmental continuum. We defined the unique transcriptional programs underlying migratory capacity, resting cellular states and apoptosis regulation in transitional gonocytes. We also identified a subpopulation of primitive spermatogonia marked by CD87 (plasminogen activator, urokinase receptor), which exhibited a higher level of self-renewal gene expression and migration potential. We further revealed a differentiation-primed state within the undifferentiated compartment, in which elevated Oct4 expression correlates with lower expression of self-renewal pathway factors, higher Rarg expression, and enhanced retinoic acid responsiveness. Lastly, a knockdown experiment revealed the role of Oct4 in the regulation of gene expression related to the MAPK pathway and cell adhesion, which may contribute to stem cell differentiation. Our study thus provides novel insights into cellular and molecular regulation during early germ cell development.
Collapse
Affiliation(s)
- Jinyue Liao
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Shuk Han Ng
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Alfred Chun Luk
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Hoi Ching Suen
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Yan Qian
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Annie Wing Tung Lee
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Jiajie Tu
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Jacqueline Chak Lam Fung
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Nelson Leung Sang Tang
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Bo Feng
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Wai Yee Chan
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Joint CUHK-UoS (University of Southampton) Joint Laboratories for Stem Cells and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- CUHK-BGI Innovation Institute of Trans-omics Hong Kong, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Pierre Fouchet
- CEA DRF IBFJ IRCM, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France
- Université Paris Diderot, Sorbonne Paris Cité, INSERM, UMR 967, 92265 Fontenay-aux-Roses, France
- Université Paris Sud, INSERM, UMR 967, 92265 Fontenay-aux-Roses, France
| | - Robin M Hobbs
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Tin Lap Lee
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Joint CUHK-UoS (University of Southampton) Joint Laboratories for Stem Cells and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- CUHK-BGI Innovation Institute of Trans-omics Hong Kong, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
30
|
Taylor J, Bebawy M. Proteins Regulating Microvesicle Biogenesis and Multidrug Resistance in Cancer. Proteomics 2019; 19:e1800165. [PMID: 30520565 DOI: 10.1002/pmic.201800165] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/02/2018] [Indexed: 01/01/2023]
Abstract
Microvesicles (MV) are emerging as important mediators of intercellular communication. While MVs are important signaling vectors for many physiological processes, they are also implicated in cancer pathology and progression. Cellular activation is perhaps the most widely reported initiator of MV biogenesis, however, the precise mechanism remains undefined. Uncovering the proteins involved in regulating MV biogenesis is of interest given their role in the dissemination of deleterious cancer traits. MVs shed from drug-resistant cancer cells transfer multidrug resistance (MDR) proteins to drug-sensitive cells and confer the MDR phenotype in a matter of hours. MDR is attributed to the overexpression of ABC transporters, primarily P-glycoprotein and MRP1. Their expression and functionality is dependent on a number of proteins. In particular, FERM domain proteins have been implicated in supporting the functionality of efflux transporters in drug-resistant cells and in recipient cells during intercellular transfer by vesicles. Herein, the most recent research on the proteins involved in MV biogenesis and in the dissemination of MV-mediated MDR are discussed. Attention is drawn to unanswered questions in the literature that may prove to be of benefit in ongoing efforts to improve clinical response to chemotherapy and circumventing MDR.
Collapse
Affiliation(s)
- Jack Taylor
- Discipline of Pharmacy, Graduate School of Health, The University of Technology Sydney, Sydney, Australia
| | - Mary Bebawy
- Discipline of Pharmacy, Graduate School of Health, The University of Technology Sydney, Sydney, Australia
| |
Collapse
|
31
|
Broertjes J, Klarenbeek J, Habani Y, Langeslag M, Jalink K. TRPM7 residue S1269 mediates cAMP dependence of Ca2+ influx. PLoS One 2019; 14:e0209563. [PMID: 30615643 PMCID: PMC6322742 DOI: 10.1371/journal.pone.0209563] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023] Open
Abstract
The nonspecific divalent cation channel TRPM7 (transient receptor potential-melastatin-like 7) is involved in many Ca2+ and Mg2+-dependent cellular processes, including survival, proliferation and migration. TRPM7 expression predicts metastasis and recurrence in breast cancer and several other cancers. In cultured cells, it can induce an invasive phenotype by promoting Ca2+-mediated epithelial-mesenchymal transition. We previously showed that in neuroblastoma cells that overexpress TRPM7 moderately, stimulation with Ca2+-mobilizing agonists leads to a characteristic sustained influx of Ca2+. Here we report that sustained influx through TRPM7 is abruptly abrogated by elevating intracellular levels of cyclic adenosine monophosphate (cAMP). Using pharmacological inhibitors and overexpression studies we show that this blockage is mediated by the cAMP effector Protein Kinase A (PKA). Mutational analysis demonstrates that the Serine residue S1269, which is present proximal to the coiled-coil domain within the protein c-terminus, is responsible for sensitivity to cAMP.
Collapse
Affiliation(s)
- Jorrit Broertjes
- Division of Cell Biology I, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeffrey Klarenbeek
- Division of Cell Biology I, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Yasmin Habani
- Division of Cell Biology I, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michiel Langeslag
- Division of Cell Biology I, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kees Jalink
- Division of Cell Biology I, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
32
|
Prevarskaya N, Skryma R, Shuba Y. Ion Channels in Cancer: Are Cancer Hallmarks Oncochannelopathies? Physiol Rev 2018; 98:559-621. [PMID: 29412049 DOI: 10.1152/physrev.00044.2016] [Citation(s) in RCA: 304] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Genomic instability is a primary cause and fundamental feature of human cancer. However, all cancer cell genotypes generally translate into several common pathophysiological features, often referred to as cancer hallmarks. Although nowadays the catalog of cancer hallmarks is quite broad, the most common and obvious of them are 1) uncontrolled proliferation, 2) resistance to programmed cell death (apoptosis), 3) tissue invasion and metastasis, and 4) sustained angiogenesis. Among the genes affected by cancer, those encoding ion channels are present. Membrane proteins responsible for signaling within cell and among cells, for coupling of extracellular events with intracellular responses, and for maintaining intracellular ionic homeostasis ion channels contribute to various extents to pathophysiological features of each cancer hallmark. Moreover, tight association of these hallmarks with ion channel dysfunction gives a good reason to classify them as special type of channelopathies, namely oncochannelopathies. Although the relation of cancer hallmarks to ion channel dysfunction differs from classical definition of channelopathies, as disease states causally linked with inherited mutations of ion channel genes that alter channel's biophysical properties, in a broader context of the disease state, to which pathogenesis ion channels essentially contribute, such classification seems absolutely appropriate. In this review the authors provide arguments to substantiate such point of view.
Collapse
Affiliation(s)
- Natalia Prevarskaya
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| | - Roman Skryma
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| | - Yaroslav Shuba
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| |
Collapse
|
33
|
Nadolni W, Zierler S. The Channel-Kinase TRPM7 as Novel Regulator of Immune System Homeostasis. Cells 2018; 7:cells7080109. [PMID: 30126133 PMCID: PMC6115979 DOI: 10.3390/cells7080109] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/13/2018] [Accepted: 08/13/2018] [Indexed: 12/16/2022] Open
Abstract
The enzyme-coupled transient receptor potential channel subfamily M member 7, TRPM7, has been associated with immunity and immune cell signalling. Here, we review the role of this remarkable signalling protein in lymphocyte proliferation, differentiation, activation and survival. We also discuss its role in mast cell, neutrophil and macrophage function and highlight the potential of TRPM7 to regulate immune system homeostasis. Further, we shed light on how the cellular signalling cascades involving TRPM7 channel and/or kinase activity culminate in pathologies as diverse as allergic hypersensitivity, arterial thrombosis and graft versus host disease (GVHD), stressing the need for TRPM7 specific pharmacological modulators.
Collapse
Affiliation(s)
- Wiebke Nadolni
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Goethestr. 33, 80336 Munich, Germany.
| | - Susanna Zierler
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Goethestr. 33, 80336 Munich, Germany.
| |
Collapse
|
34
|
Fels B, Bulk E, Pethő Z, Schwab A. The Role of TRP Channels in the Metastatic Cascade. Pharmaceuticals (Basel) 2018; 11:E48. [PMID: 29772843 PMCID: PMC6027473 DOI: 10.3390/ph11020048] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/16/2022] Open
Abstract
A dysregulated cellular Ca2+ homeostasis is involved in multiple pathologies including cancer. Changes in Ca2+ signaling caused by altered fluxes through ion channels and transporters (the transportome) are involved in all steps of the metastatic cascade. Cancer cells thereby "re-program" and "misuse" the cellular transportome to regulate proliferation, apoptosis, metabolism, growth factor signaling, migration and invasion. Cancer cells use their transportome to cope with diverse environmental challenges during the metastatic cascade, like hypoxic, acidic and mechanical cues. Hence, ion channels and transporters are key modulators of cancer progression. This review focuses on the role of transient receptor potential (TRP) channels in the metastatic cascade. After briefly introducing the role of the transportome in cancer, we discuss TRP channel functions in cancer cell migration. We highlight the role of TRP channels in sensing and transmitting cues from the tumor microenvironment and discuss their role in cancer cell invasion. We identify open questions concerning the role of TRP channels in circulating tumor cells and in the processes of intra- and extravasation of tumor cells. We emphasize the importance of TRP channels in different steps of cancer metastasis and propose cancer-specific TRP channel blockade as a therapeutic option in cancer treatment.
Collapse
Affiliation(s)
- Benedikt Fels
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany.
| | - Etmar Bulk
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany.
| | - Zoltán Pethő
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany.
| | - Albrecht Schwab
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany.
| |
Collapse
|
35
|
|
36
|
Decreased TRPM7 inhibits activities and induces apoptosis of bladder cancer cells via ERK1/2 pathway. Oncotarget 2018; 7:72941-72960. [PMID: 27662662 PMCID: PMC5341955 DOI: 10.18632/oncotarget.12146] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 09/13/2016] [Indexed: 12/15/2022] Open
Abstract
Transient receptor potential melastatin 7 (TRPM7) functions as a Mg2+/Ca2+-permeable channel fused with a kinase domain and regulates various physical processes and diseases. However, its effects on pathogenesis of human bladder cancer (BCa) has not been clarified yet. Our microarray analysis has suggested that calcium signaling pathway is connected with bladder cancer via MAPK pathway. Therefore, we aim to investigate the mechanism of TRPM7 in BCa tumorigenesis by using BCa tissues compared with normal bladder epithelium tissues, as well as using distinct BCa cell lines (EJ, 5637 and T24). We observed increased TRPM7 expression and dysregulation of proteins involved in Epithelial-Mesenchymal Transition (EMT) in BCa tissues. Moreover, knockdown of TRPM7 in BCa cells reversed the EMT status, accompanied by increase of reactive oxygen species (ROS). Furthermore, TRPM7 deficiency could inhibit BCa cell proliferation, migration and invasion, as well as induce p-ERK1/2 and suppress PI3K/AKT at the protein level. Downregulation of TRPM7 promoted cell cycle arrest at G0/G1 phase and apoptosis in vitro, which could be recovered by pre-treatment with U0126 to deactivate ERK1/2, suggesting a close correlation between TRPM7 and the MAPK signaling pathway. Furthermore, a NOD/SCID mouse model transplanted using the BCa cells was established, revealing delayed tumor growth by reduced protein activity and mRNA transcription of TRPM7 in vivo. Our results suggested TRPM7 might be essential for BCa tumorigenesis by interfering BCa cell proliferation, motility and apoptosis.
Collapse
|
37
|
Huang X, Jin M, Chen YX, Wang J, Zhai K, Chang Y, Yuan Q, Yao KT, Ji G. ERP44 inhibits human lung cancer cell migration mainly via IP3R2. Aging (Albany NY) 2017; 8:1276-86. [PMID: 27347718 PMCID: PMC4931832 DOI: 10.18632/aging.100984] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 06/10/2016] [Indexed: 12/21/2022]
Abstract
Cancer cell migration is involved in tumour metastasis. However, the relationship between calcium signalling and cancer migration is not well elucidated. In this study, we used the human lung adenocarcinoma A549 cell line to examine the role of endoplasmic reticulum protein 44 (ERP44), which has been reported to regulate calcium release inside of the endoplasmic reticulum (ER), in cell migration. We found that the inositol 1,4,5-trisphosphate receptors (IP3Rs/ITPRs) inhibitor 2-APB significantly inhibited A549 cell migration by inhibiting cell polarization and pseudopodium protrusion, which suggests that Ca2+ is necessary for A549 cell migration. Similarly, the overexpression of ERP44 reduced intracellular Ca2+ release via IP3Rs, altered cell morphology and significantly inhibited the migration of A549 cells. These phenomena were primarily dependent on IP3R2 because wound healing in A549 cells with IP3R2 rather than IP3R1 or IP3R3 siRNA was markedly inhibited. Moreover, the overexpression of ERP44 did not affect the migration of the human neuroblastoma cell line SH-SY5Y, which mainly expresses IP3R1. Based on the above observations, we conclude that ERP44 regulates A549 cell migration mainly via an IP3R2-dependent pathway.
Collapse
Affiliation(s)
- Xue Huang
- Cancer Research Institute of Southern Medical University, Guangzhou, China
| | - Meng Jin
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ying-Xiao Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Current address: Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jun Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Kui Zhai
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yan Chang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Qi Yuan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Kai-Tai Yao
- Cancer Research Institute of Southern Medical University, Guangzhou, China
| | - Guangju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
38
|
Assessment of TRPM7 functions by drug-like small molecules. Cell Calcium 2017; 67:166-173. [DOI: 10.1016/j.ceca.2017.03.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 12/28/2022]
|
39
|
Gao SL, Kong CZ, Zhang Z, Li ZL, Bi JB, Liu XK. TRPM7 is overexpressed in bladder cancer and promotes proliferation, migration, invasion and tumor growth. Oncol Rep 2017; 38:1967-1976. [PMID: 28791418 PMCID: PMC5652943 DOI: 10.3892/or.2017.5883] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 06/23/2017] [Indexed: 12/22/2022] Open
Abstract
Recent findings suggest that the melastatin transient receptor potential channel 7 (TRPM7) is overexpressed in many types of cancers and is involved in tumorigenesis. However, its expression pattern and the potential role in bladder cancer remain unclear. The aim of the present study was to investigate the expression status of TRPM7 and its relationship with the development of bladder cancer. In the present study, we observed that the expression of TRPM7 was significantly elevated in bladder cancer tissues compared with that noted in the adjacent non-tumor tissues. Furthermore, increased TRPM7 expression was significantly associated with recurrence, metastasis and prognosis. In addition, after knockdown of the expression of TRPM7 by siRNA, the proliferation and the motility of T24 and 5637 cells were obviously inhibited, and downregulation of TRPM7 was found to play an important role in bladder cancer cell apoptosis. In conclusion, our findings suggest that TRPM7 plays an important role in bladder cancer, and TRPM7 may serve as a potentially unfavorable factor and novel target for human bladder cancer.
Collapse
Affiliation(s)
- Sheng-Lin Gao
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Chui-Ze Kong
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Zhe Zhang
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Ze-Liang Li
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Jian-Bin Bi
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Xian-Kui Liu
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
40
|
Wong R, Turlova E, Feng ZP, Rutka JT, Sun HS. Activation of TRPM7 by naltriben enhances migration and invasion of glioblastoma cells. Oncotarget 2017; 8:11239-11248. [PMID: 28061441 PMCID: PMC5355261 DOI: 10.18632/oncotarget.14496] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/26/2016] [Indexed: 01/09/2023] Open
Abstract
Glioblastoma (GBM), the most common and aggressive brain tumor in the central nervous system, remains a lethal diagnosis with a median survival of < 15 months. Aberrant expression of the TRPM7 channel has been linked to GBM functions. In this study, using the human GBM cell line U87, we evaluated the TRPM7 activator naltriben on GBM viability, migration, and invasiveness. First, using the whole-cell patch-clamp technique, we showed that naltriben enhanced the endogenous TRPM7-like current in U87 cells. In addition, with Fura-2 Ca2+ imaging, we observed robust Ca2+ influx following naltriben application. Naltriben significantly enhanced U87 cell migration and invasion (assessed with scratch wound assays, Matrigel invasion experiments, and MMP-2 protein expression), but not viability and proliferation (evaluated with MTT assays). Using Western immunoblots, we also detected the protein levels of p-Akt/t-Akt, and p-ERK1|2/t-ERK1|2. We found that naltriben enhanced the MAPK/ERK signaling pathway, but not the PI3k/Akt pathway. Therefore, potentiated TRPM7 activity contributes to the devastating migratory and invasive characteristics of GBM.
Collapse
Affiliation(s)
- Raymond Wong
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Ekaterina Turlova
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - James T Rutka
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Hong-Shuo Sun
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.,Department of Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
41
|
Mignen O, Constantin B, Potier-Cartereau M, Penna A, Gautier M, Guéguinou M, Renaudineau Y, Shoji KF, Félix R, Bayet E, Buscaglia P, Debant M, Chantôme A, Vandier C. Constitutive calcium entry and cancer: updated views and insights. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:395-413. [PMID: 28516266 DOI: 10.1007/s00249-017-1216-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 03/10/2017] [Accepted: 04/26/2017] [Indexed: 12/20/2022]
Abstract
Tight control of basal cytosolic Ca2+ concentration is essential for cell survival and to fine-tune Ca2+-dependent cell functions. A way to control this basal cytosolic Ca2+ concentration is to regulate membrane Ca2+ channels including store-operated Ca2+ channels and secondary messenger-operated channels linked to G-protein-coupled or tyrosine kinase receptor activation. Orai, with or without its reticular STIM partner and Transient Receptor Potential (TRP) proteins, were considered to be the main Ca2+ channels involved. It is well accepted that, in response to cell stimulation, opening of these Ca2+ channels contributes to Ca2+ entry and the transient increase in cytosolic Ca2+ concentration involved in intracellular signaling. However, in various experimental conditions, Ca2+ entry and/or Ca2+ currents can be recorded at rest, without application of any experimental stimulation. This led to the proposition that some plasma membrane Ca2+ channels are already open/activated in basal condition, contributing therefore to constitutive Ca2+ entry. This article focuses on direct and indirect observations supporting constitutive activity of channels belonging to the Orai and TRP families and on the mechanisms underlying their basal/constitutive activities.
Collapse
Affiliation(s)
- Olivier Mignen
- Inserm UMR 1078 IFR148 Université de Bretagne Occidentale, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Bruno Constantin
- STIM, ERL 7368 CNRS Université de Poitiers, Poitiers, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Marie Potier-Cartereau
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Aubin Penna
- IRSET, Inserm U1085, University of Rennes 1, 36043, Rennes, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Mathieu Gautier
- EA4667, Université de Picardie Jules Verne, 80039, Amiens, France
| | - Maxime Guéguinou
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Yves Renaudineau
- EA 2216, Inserm ESPRI, ERI 29, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Kenji F Shoji
- IRSET, Inserm U1085, University of Rennes 1, 36043, Rennes, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Romain Félix
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Elsa Bayet
- STIM, ERL 7368 CNRS Université de Poitiers, Poitiers, France
- IRSET, Inserm U1085, University of Rennes 1, 36043, Rennes, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Paul Buscaglia
- Inserm UMR 1078 IFR148 Université de Bretagne Occidentale, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Marjolaine Debant
- Inserm UMR 1078 IFR148 Université de Bretagne Occidentale, Brest, France
- EA 2216, Inserm ESPRI, ERI 29, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Aurélie Chantôme
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Christophe Vandier
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France.
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France.
| |
Collapse
|
42
|
Zierler S, Hampe S, Nadolni W. TRPM channels as potential therapeutic targets against pro-inflammatory diseases. Cell Calcium 2017; 67:105-115. [PMID: 28549569 DOI: 10.1016/j.ceca.2017.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/02/2017] [Indexed: 02/08/2023]
Abstract
The immune system protects our body against foreign pathogens. However, if it overshoots or turns against itself, pro-inflammatory diseases, such as rheumatoid arthritis, inflammatory bowel disease, or diabetes develop. Ions, the most basic signaling molecules, shape intracellular signaling cascades resulting in immune cell activation and subsequent immune responses. Mutations in ion channels required for calcium signaling result in human immunodeficiencies and highlight those ion channels as valued targets for therapies against pro-inflammatory diseases. Signaling pathways regulated by melastatin-like transient receptor potential (TRPM) cation channels also play crucial roles in calcium signaling and leukocyte physiology, affecting phagocytosis, degranulation, chemokine and cytokine expression, chemotaxis and invasion, as well as lymphocyte development and proliferation. Therefore, this review discusses their regulation, possible interactions and whether they can be exploited as targets for therapeutic approaches to pro-inflammatory diseases.
Collapse
Affiliation(s)
- Susanna Zierler
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Germany.
| | - Sarah Hampe
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Germany
| | - Wiebke Nadolni
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Germany
| |
Collapse
|
43
|
Ciardo MG, Ferrer-Montiel A. Lipids as central modulators of sensory TRP channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1615-1628. [PMID: 28432033 DOI: 10.1016/j.bbamem.2017.04.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/13/2017] [Accepted: 04/15/2017] [Indexed: 12/13/2022]
Abstract
The transient receptor potential (TRP) ion channel family is involved in a diversity of physiological processes including sensory and homeostatic functions, as well as muscle contraction and vasomotor control. Their dysfunction contributes to the etiology of several diseases, being validated as therapeutic targets. These ion channels may be activated by physical or chemical stimuli and their function is highly influenced by signaling molecules activated by extracellular signals. Notably, as integral membrane proteins, lipid molecules also modulate their membrane location and function either by direct interaction with the channel structure or by modulating the physico-chemical properties of the cellular membrane. This lipid-based modulatory effect is being considered an alternative and promising approach to regulate TRP channel dysfunction in diseases. Here, we review the current progress in this exciting field highlighting a complex channel regulation by a large diversity of lipid molecules and suggesting some diseases that may benefit from a membrane lipid therapy. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Av. De la Universidad s/n, Elche, Spain.
| |
Collapse
|
44
|
Yee NS. Role of TRPM7 in Cancer: Potential as Molecular Biomarker and Therapeutic Target. Pharmaceuticals (Basel) 2017; 10:39. [PMID: 28379203 PMCID: PMC5490396 DOI: 10.3390/ph10020039] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 02/22/2017] [Accepted: 03/29/2017] [Indexed: 12/31/2022] Open
Abstract
The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed ion channel with intrinsic kinase activity. Molecular and electrophysiological analyses of the structure and activity of TRPM7 have revealed functional coupling of its channel and kinase activity. Studies have indicated the important roles of TRPM7 channel-kinase in fundamental cellular processes, physiological responses, and embryonic development. Accumulating evidence has shown that TRPM7 is aberrantly expressed and/or activated in human diseases including cancer. TRPM7 plays a variety of functional roles in cancer cells including survival, cell cycle progression, proliferation, growth, migration, invasion, and epithelial-mesenchymal transition (EMT). Data from a study using mouse xenograft of human cancer show that TRPM7 is required for tumor growth and metastasis. The aberrant expression of TRPM7 and its genetic mutations/polymorphisms have been identified in various types of carcinoma. Chemical modulators of TRPM7 channel produced inhibition of proliferation, growth, migration, invasion, invadosome formation, and markers of EMT in cancer cells. Taken together, these studies suggest the potential value of exploiting TRPM7 channel-kinase as a molecular biomarker and therapeutic target in human malignancies.
Collapse
Affiliation(s)
- Nelson S Yee
- Division of Hematology-Oncology, Department of Medicine, PennState Health Milton S. Hershey Medical Center, Program of Experimental Therapeutics, PennState Cancer Institute, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA.
| |
Collapse
|
45
|
Rybarczyk P, Vanlaeys A, Brassart B, Dhennin-Duthille I, Chatelain D, Sevestre H, Ouadid-Ahidouch H, Gautier M. The Transient Receptor Potential Melastatin 7 Channel Regulates Pancreatic Cancer Cell Invasion through the Hsp90α/uPA/MMP2 pathway. Neoplasia 2017; 19:288-300. [PMID: 28284058 PMCID: PMC5345960 DOI: 10.1016/j.neo.2017.01.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/05/2017] [Accepted: 01/13/2017] [Indexed: 12/28/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a very poor prognosis. There is an urgent need to better understand the molecular mechanisms that regulate PDAC cell aggressiveness. The transient receptor potential melastatin 7 (TRPM7) is a nonselective cationic channel that mainly conducts Ca2+ and Mg2+. TRPM7 is overexpressed in numerous malignancies including PDAC. In the present study, we used the PANC-1 and MIA PaCa-2 cell lines to specifically assess the role of TRPM7 in cell invasion and matrix metalloproteinase secretion. We show that TRPM7 regulates Mg2+ homeostasis and constitutive cation entry in both PDAC cell lines. Moreover, cell invasion is strongly reduced by TRPM7 silencing without affecting the cell viability. Conditioned media were further studied, by gel zymography, to detect matrix metalloproteinase (MMP) secretion in PDAC cells. Our results show that MMP-2, urokinase plasminogen activator (uPA), and heat-shock protein 90α (Hsp90α) secretions are significantly decreased in TRPM7-deficient PDAC cells. Moreover, TRPM7 expression in human PDAC lymph node metastasis is correlated to the channel expression in primary tumor. Taken together, our results show that TRPM7 is involved in PDAC cell invasion through regulation of Hsp90α/uPA/MMP-2 proteolytic axis, confirming that this channel could be a promising biomarker and possibly a target for PDAC metastasis therapy.
Collapse
Affiliation(s)
- Pierre Rybarczyk
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231)
| | - Alison Vanlaeys
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231)
| | - Bertrand Brassart
- SFR CAP-Santé (FED 4231); UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), F-51095 Reims, France
| | - Isabelle Dhennin-Duthille
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231)
| | - Denis Chatelain
- Service d'anatomie pathologique, CHU d'Amiens, Université de Picardie Jules Verne, F-80000 Amiens, France, France
| | - Henri Sevestre
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231); Service d'anatomie pathologique, CHU d'Amiens, Université de Picardie Jules Verne, F-80000 Amiens, France, France
| | - Halima Ouadid-Ahidouch
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231)
| | - Mathieu Gautier
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231).
| |
Collapse
|
46
|
Mudit M, El Sayed KA. Cancer control potential of marine natural product scaffolds through inhibition of tumor cell migration and invasion. Drug Discov Today 2016; 21:1745-1760. [DOI: 10.1016/j.drudis.2016.06.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 01/14/2023]
|
47
|
Chen Y, Yu Y, Sun S, Wang Z, Liu P, Liu S, Jiang J. Bradykinin promotes migration and invasion of hepatocellular carcinoma cells through TRPM7 and MMP2. Exp Cell Res 2016; 349:68-76. [DOI: 10.1016/j.yexcr.2016.09.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/26/2016] [Accepted: 09/28/2016] [Indexed: 12/26/2022]
|
48
|
Kim BJ, Hong C. Role of transient receptor potential melastatin type 7 channel in gastric cancer. Integr Med Res 2016; 5:124-130. [PMID: 28462107 PMCID: PMC5381434 DOI: 10.1016/j.imr.2016.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/01/2016] [Accepted: 04/05/2016] [Indexed: 01/11/2023] Open
Abstract
Transient receptor potential (TRP) proteins are a family of ion channels, which are responsible for a wide array of cellular functions. In particular, TRP melastatin type (TRPM) 7 is expressed everywhere and permeable to divalent cations such as Mg2+ and Ca2+. It contains a channel and a kinase domain. Recent studies indicate that activation of TRPM7 plays an important role in the growth and survival of gastric cancer cells. In this review, we describe and discuss the findings of recent studies that have provided novel insights of the relation between TRPM7 and gastric cancer.
Collapse
Affiliation(s)
- Byung Joo Kim
- Division of Longevity and Biofunctional Medicine, School of Korean Medicine, Pusan National University, Yangsan, Korea
- Healthy Aging Korean Medical Research Center, School of Korean Medicine, Pusan National University, Yangsan, Korea
| | - Chansik Hong
- Department of Physiology, Seoul National University, College of Medicine, Seoul, Korea
| |
Collapse
|
49
|
Chen WL, Barszczyk A, Turlova E, Deurloo M, Liu B, Yang BB, Rutka JT, Feng ZP, Sun HS. Inhibition of TRPM7 by carvacrol suppresses glioblastoma cell proliferation, migration and invasion. Oncotarget 2016; 6:16321-40. [PMID: 25965832 PMCID: PMC4599272 DOI: 10.18632/oncotarget.3872] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 04/02/2015] [Indexed: 01/27/2023] Open
Abstract
Glioblastomas are progressive brain tumors with devastating proliferative and invasive characteristics. Ion channels are the second largest target class for drug development. In this study, we investigated the effects of the TRPM7 inhibitor carvacrol on the viability, resistance to apoptosis, migration, and invasiveness of the human U87 glioblastoma cell line. The expression levels of TRPM7 mRNA and protein in U87 cells were detected by RT-PCR, western blotting and immunofluorescence. TRPM7 currents were recorded using whole-cell patch-clamp techniques. An MTT assay was used to assess cell viability and proliferation. Wound healing and transwell experiments were used to evaluate cell migration and invasion. Protein levels of p-Akt/t-Akt, p-ERK1/2/t-ERK1/2, cleaved caspase-3, MMP-2 and phosphorylated cofilin were also detected. TRPM7 mRNA and protein expression in U87 cells is higher than in normal human astrocytes. Whole-cell patch-clamp recording showed that carvacrol blocks recombinant TRPM7 current in HEK293 cells and endogenous TRPM7-like current in U87 cells. Carvacrol treatment reduced the viability, migration and invasion of U87 cells. Carvacrol also decreased MMP-2 protein expression and promoted the phosphorylation of cofilin. Furthermore, carvacrol inhibited the Ras/MEK/MAPK and PI3K/Akt signaling pathways. Therefore, carvacrol may have therapeutic potential for the treatment of glioblastomas through its inhibition of TRPM7 channels.
Collapse
Affiliation(s)
- Wen-Liang Chen
- Department of Surgery, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - Andrew Barszczyk
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Ekaterina Turlova
- Department of Surgery, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - Marielle Deurloo
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Baosong Liu
- Department of Surgery, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - Burton B Yang
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - James T Rutka
- Department of Surgery, University of Toronto, Toronto, Canada
| | - Zhong-Ping Feng
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Hong-Shuo Sun
- Department of Surgery, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada.,Department of Pharmacology, University of Toronto, Toronto, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
50
|
Qin Y, Liao ZW, Luo JY, Wu WZ, Lu AS, Su PX, Lai BQ, Wang XX. Functional characterization of TRPM7 in nasopharyngeal carcinoma and its knockdown effects on tumorigenesis. Tumour Biol 2016; 37:9273-83. [PMID: 26779625 DOI: 10.1007/s13277-015-4636-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/10/2015] [Indexed: 01/18/2023] Open
Abstract
The aim of this study was to evaluate the association of functional expression of TRPM7 with nasopharyngeal carcinoma (NPC) growth. We examined the correlation of TRPM7 expression with cell growth and proliferation, cell cycle, and apoptosis in vitro in NPC cell lines and NPC tumorigenesis in mice by conducting experiments in mice and by further analyzing the tumor volume and growth. We further explored to see whether there is any positive correlation with the TRPM7 knockdown in NPC cells with their sensitivity to radiation. We found that the functional expression of TRPM7 in nasopharyngeal carcinoma is a critical requirement for physiological processes such as cell cycle, resistance to apoptosis, and cell proliferation. TRPM7 knockdown also enhanced sensitivity to radiotherapy of nasopharyngeal carcinoma. Moreover, we identified TRPM7 as a novel potential regulator of cell proliferation in NPC, through signal transducer and activator of transcription 3 (STAT3)-mediated signaling pathway and other anti-apoptotic factors. TRPM7 and STAT3 activation might be critical for the growth of NPC cells and could be an effective target for treatment of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Yi Qin
- Department of Orthopedics, Zhuhai People's Hospital, Zhuhai, 519000, China
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhi-Wei Liao
- Department of Radiation Oncology, Cancer Center of Guangzhou Medical University, Guangzhou, China
| | - Jing-Yan Luo
- Forevergen Biosciences Center, R&D Unit 602, Guangzhou, 510000, China
| | - Wen-Zhe Wu
- Forevergen Biosciences Center, R&D Unit 602, Guangzhou, 510000, China
| | - An-Shang Lu
- Forevergen Biosciences Center, R&D Unit 602, Guangzhou, 510000, China
| | - Pu-Xia Su
- Forevergen Biosciences Center, R&D Unit 602, Guangzhou, 510000, China
| | - Bing-Quan Lai
- Forevergen Biosciences Center, R&D Unit 602, Guangzhou, 510000, China.
| | - Xiao-Xiao Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|