1
|
Hao Z, Zhang M, Du Y, Liu J, Zeng G, Li H, Peng X. Invadopodia in cancer metastasis: dynamics, regulation, and targeted therapies. J Transl Med 2025; 23:548. [PMID: 40380267 PMCID: PMC12083038 DOI: 10.1186/s12967-025-06526-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/21/2025] [Indexed: 05/19/2025] Open
Abstract
Pseudopodia and invadopodia are dynamic, actin-rich membrane structures extending from the cell surface. While pseudopodia are found in various cell types, invadopodia are exclusive to tumor cells and play a key role in cancer progression. These specialized structures enable tumor cells to degrade the extracellular matrix, breach tissue barriers, and invade surrounding tissues and blood vessels, thus facilitating metastasis. Extensive research has elucidated the distinct structure of invadopodia, the signaling pathways driving their formation, and their interaction with the tumor microenvironment. Integrin- and Src kinase-mediated signaling pathways regulate invadopodia dynamics. This review explores the mechanisms underlying invadopodia stabilization and highlights recent insights into their regulation by the tumor microenvironment. Particular emphasis is placed on the role of cell surface signaling in modulating invadopodia activity and the intracellular targeting of matrix metalloproteinases (MMPs) in enhancing invasive potential. A deeper understanding of invadopodia-driven cancer cell migration and metastasis provides valuable implications for therapeutic development. These findings support the potential for receptor-mediated and molecularly targeted therapies to inhibit tumor metastasis, improve clinical outcomes, and enhance the efficacy of existing cancer treatments.
Collapse
Affiliation(s)
- Zhixiong Hao
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Manru Zhang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Yao Du
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jiaxing Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Guolong Zeng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hangyu Li
- The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121001, China.
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
2
|
Zhou P, Yao W, Liu L, Yan Q, Chen X, Wei X, Ding S, Lv Z, Zhu F. SPG21, a potential oncogene targeted by miR-128-3p, amplifies HBx-induced carcinogenesis and chemoresistance via activation of TRPM7-mediated JNK pathway in hepatocellular carcinoma. Cell Oncol (Dordr) 2024; 47:1757-1778. [PMID: 38753154 DOI: 10.1007/s13402-024-00955-5] [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] [Accepted: 05/01/2024] [Indexed: 07/31/2024] Open
Abstract
PURPOSE Chronic hepatitis B virus (HBV) infection is the primary risk factor for the malignant progression of hepatocellular carcinoma (HCC). It has been reported that HBV X protein (HBx) possesses oncogenic properties, promoting hepatocarcinogenesis and chemoresistance. However, the detailed molecular mechanisms are not fully understood. Here, we aim to investigate the effects of miR-128-3p/SPG21 axis on HBx-induced hepatocarcinogenesis and chemoresistance. METHODS The expression of SPG21 in HCC was determined using bioinformatics analysis, quantitative real-time PCR (qRT-PCR), western blotting, and immunohistochemistry (IHC). The roles of SPG21 in HCC were elucidated through a series of in vitro and in vivo experiments, including real-time cellular analysis (RTCA), matrigel invasion assay, and xenograft mouse model. Pharmacologic treatment and flow cytometry were performed to demonstrate the potential mechanism of SPG21 in HCC. RESULTS SPG21 expression was elevated in HCC tissues compared to adjacent non-tumor tissues (NTs). Moreover, higher SPG21 expression correlated with poor overall survival. Functional assays revealed that SPG21 fostered HCC tumorigenesis and invasion. MiR-128-3p, which targeted SPG21, was downregulated in HCC tissues. Subsequent analyses showed that HBx amplified TRPM7-mediated calcium influx via miR-128-3p/SPG21, thereby activating the c-Jun N-terminal kinase (JNK) pathway. Furthermore, HBx inhibited doxorubicin-induced apoptosis by engaging the JNK pathway through miR-128-3p/SPG21. CONCLUSION The study suggested that SPG21, targeted by miR-128-3p, might be involved in enhancing HBx-induced carcinogenesis and doxorubicin resistance in HCC via the TRPM7/Ca2+/JNK signaling pathway. This insight suggested that SPG21 could be recognized as a potential oncogene, offering a novel perspective on its role as a prognostic factor and a therapeutic target in the context of HCC.
Collapse
Affiliation(s)
- Ping Zhou
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, No. 185 Donghu Road, 430071, Wuhan, China
| | - Wei Yao
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, No. 185 Donghu Road, 430071, Wuhan, China
| | - Lijuan Liu
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, No. 185 Donghu Road, 430071, Wuhan, China
| | - Qiujin Yan
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, No. 185 Donghu Road, 430071, Wuhan, China
| | - Xiaobei Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060, Wuhan, China
| | - Xiaocui Wei
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, No. 185 Donghu Road, 430071, Wuhan, China
| | - Shuang Ding
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, No. 185 Donghu Road, 430071, Wuhan, China
| | - Zhao Lv
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, No. 185 Donghu Road, 430071, Wuhan, China
| | - Fan Zhu
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, No. 185 Donghu Road, 430071, Wuhan, China.
- Hubei Province Key Laboratory of Allergy & Immunology, Wuhan University, 430071, Wuhan, China.
| |
Collapse
|
3
|
Feroz W, Park BS, Siripurapu M, Ntim N, Kilroy MK, Sheikh AMA, Mishra R, Garrett JT. Non-Muscle Myosin II A: Friend or Foe in Cancer? Int J Mol Sci 2024; 25:9435. [PMID: 39273383 PMCID: PMC11395477 DOI: 10.3390/ijms25179435] [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: 07/23/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Non-muscle myosin IIA (NM IIA) is a motor protein that belongs to the myosin II family. The myosin heavy chain 9 (MYH9) gene encodes the heavy chain of NM IIA. NM IIA is a hexamer and contains three pairs of peptides, which include the dimer of heavy chains, essential light chains, and regulatory light chains. NM IIA is a part of the actomyosin complex that generates mechanical force and tension to carry out essential cellular functions, including adhesion, cytokinesis, migration, and the maintenance of cell shape and polarity. These functions are regulated via light and heavy chain phosphorylation at different amino acid residues. Apart from physiological functions, NM IIA is also linked to the development of cancer and genetic and neurological disorders. MYH9 gene mutations result in the development of several autosomal dominant disorders, such as May-Hegglin anomaly (MHA) and Epstein syndrome (EPS). Multiple studies have reported NM IIA as a tumor suppressor in melanoma and head and neck squamous cell carcinoma; however, studies also indicate that NM IIA is a critical player in promoting tumorigenesis, chemoradiotherapy resistance, and stemness. The ROCK-NM IIA pathway regulates cellular movement and shape via the control of cytoskeletal dynamics. In addition, the ROCK-NM IIA pathway is dysregulated in various solid tumors and leukemia. Currently, there are very few compounds targeting NM IIA, and most of these compounds are still being studied in preclinical models. This review provides comprehensive evidence highlighting the dual role of NM IIA in multiple cancer types and summarizes the signaling networks involved in tumorigenesis. Furthermore, we also discuss the role of NM IIA as a potential therapeutic target with a focus on the ROCK-NM IIA pathway.
Collapse
Affiliation(s)
- Wasim Feroz
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Briley SoYoung Park
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
- Cancer Research Scholars Program, College of Allied Health Sciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Meghna Siripurapu
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Nicole Ntim
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Mary Kate Kilroy
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | | | - Rosalin Mishra
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Joan T. Garrett
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| |
Collapse
|
4
|
Yao H, Tang L, Wang D, Pang H, Yang K. F-actin microfilaments affect the LIPUS-promoted osteogenic differentiation of BMSCs through TRPM7. Biotechnol J 2024; 19:e2400310. [PMID: 39212193 DOI: 10.1002/biot.202400310] [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: 05/07/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
The differentiation of bone marrow mesenchymal stem cells (BMSCs) toward osteogenesis can be induced by low-intensity pulsed ultrasound (LIPUS). However, the molecular mechanisms responsible for LIPUS stimulation are unclear. The possible molecular mechanisms by which LIPUS promotes osteogenic differentiation of BMSCs were investigated in this study. The quantification of alkaline phosphatase (ALP) activity, Alizarin Red S staining, ALP staining, and the establishment of a calvarial defect model were used to evaluate osteogenic effects. Immunofluorescence was performed to observe the expression of microfilaments and transient receptor potential melastatin 7 (TRPM7). The levels of F-actin/G-actin and osteogenesis-related proteins under LIPUS alone or LIPUS combined with cytoskeleton interfering drugs (Cytochalasin D [CytoD] or Jasplakinolide [JA]) were assayed by western blot. Quantitative real-time reverse transcription polymerase chain reaction was utilized to measure the expression of Trpm7 mRNA. Moreover, adenoviral Trpm7 knockdown was verified using western blot. The results demonstrated that LIPUS promoted bone formation in vivo. Under osteogenic induction in vitro, the osteogenesis of BMSCs induced by LIPUS was accompanied by the depolymerization and rearrangement of microfilaments and increased levels of TRPM7. By perturbing intracellular actin dynamics, CytoD enhanced the pro-osteogenicity of LIPUS and increased TRPM7 level, while JA inhibited the pro-osteogenicity of LIPUS and reduced TRPM7 level. Additionally, the knockdown of Trpm7 suppressed the osteogenic promotion of BMSCs induced by LIPUS. The transient depolymerization and rearrangement of the cytoskeleton microfilaments mediated by LIPUS can affect TRPM7 expression and subsequently promote the osteogenesis of BMSCs. This study provides further direction for exploring the molecular mechanism of LIPUS, as a mechanical stress, in facilitating the osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Huan Yao
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Tang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dong Wang
- Department of Ultrasound, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hua Pang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Yang
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
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
|
6
|
Chi A, Yang B, Dai H, Li X, Mo J, Gao Y, Chen Z, Feng X, Ma M, Li Y, Yang C, Liu J, Liu H, Wang Z, Gao F, Liao Y, Shi X, Deng C, Zhang M. Stem Leydig cells support macrophage immunological homeostasis through mitochondrial transfer in mice. Nat Commun 2024; 15:2120. [PMID: 38459012 PMCID: PMC10924100 DOI: 10.1038/s41467-024-46190-2] [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: 06/03/2023] [Accepted: 02/16/2024] [Indexed: 03/10/2024] Open
Abstract
As testicular mesenchymal stromal cells, stem Leydig cells (SLCs) show great promise in the treatment of male hypogonadism. The therapeutic functions of mesenchymal stromal cells are largely determined by their reciprocal regulation by immune responses. However, the immunoregulatory properties of SLCs remain unclear. Here, we observe that SLCs transplantation restore male fertility and testosterone production in an ischemia‒reperfusion injury mouse model. SLCs prevent inflammatory cascades through mitochondrial transfer to macrophages. Reactive oxygen species (ROS) released from activated macrophages inducing mitochondrial transfer from SLCs to macrophages in a transient receptor potential cation channel subfamily member 7 (TRPM7)-mediated manner. Notably, knockdown of TRPM7 in transplanted SLCs compromised therapeutic outcomes in both testicular ischemia‒reperfusion and testicular aging mouse models. These findings reveal a new mechanism of SLCs transplantation that may contribute to preserve testis function in male patients with hypogonadism related to immune disorders.
Collapse
Affiliation(s)
- Ani Chi
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Bicheng Yang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hao Dai
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Xinyu Li
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiahui Mo
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yong Gao
- Reproductive Medicine Center, The Key Laboratory for Reproductive Medicine of Guangdong Province, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhihong Chen
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xin Feng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Menghui Ma
- Center of Reproductive Medicine, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Yanqing Li
- Center of Reproductive Medicine, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Chao Yang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Jie Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Hanchao Liu
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhenqing Wang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Feng Gao
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Reproductive Medicine Center, The Key Laboratory for Reproductive Medicine of Guangdong Province, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yan Liao
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Xuetao Shi
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
- National Engineering Research Centre for Tissue Restoration and Reconstruction and Key Laboratory of Biomedical Engineering of Guangdong Province South China University of Technology, Guangzhou, 510640, China.
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, 518054, China.
| | - Chunhua Deng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Min Zhang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
7
|
Liu Q, Cheng C, Huang J, Yan W, Wen Y, Liu Z, Zhou B, Guo S, Fang W. MYH9: A key protein involved in tumor progression and virus-related diseases. Biomed Pharmacother 2024; 171:116118. [PMID: 38181716 DOI: 10.1016/j.biopha.2023.116118] [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: 09/03/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
The myosin heavy chain 9 (MYH9) gene encodes the heavy chain of non-muscle myosin IIA (NMIIA), which belongs to the myosin II subfamily of actin-based molecular motors. Previous studies have demonstrated that abnormal expression and mutations of MYH9 were correlated with MYH9-related diseases and tumors. Furthermore, earlier investigations identified MYH9 as a tumor suppressor. However, subsequent research revealed that MYH9 promoted tumorigenesis, progression and chemoradiotherapy resistance. Note-worthily, MYH9 has also been linked to viral infections, like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Epstein-Barr virus, and hepatitis B virus, as a receptor or co-receptor. In addition, MYH9 promotes the development of hepatocellular carcinoma by interacting with the hepatitis B virus-encoding X protein. Finally, various findings highlighted the role of MYH9 in the development of these illnesses, especially in tumors. This review summarizes the involvement of the MYH9-regulated signaling network in tumors and virus-related diseases and presents possible drug interventions on MYH9, providing insights for the use of MYH9 as a therapeutic target for tumors and virus-mediated diseases.
Collapse
Affiliation(s)
- Qing Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Chao Cheng
- Department of Otolaryngology, Shenzhen Longgang Otolaryngology hospital, Shenzhen 518000, China
| | - Jiyu Huang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Weiwei Yan
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Yinhao Wen
- Department of Oncology, Pingxiang People's Hospital, Pingxiang 337000, China
| | - Zhen Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China; Key Laboratory of Protein Modification and Degradation, Basic School of Guangzhou Medical University, Guangzhou 510315, China.
| | - Beixian Zhou
- The People's Hospital of Gaozhou, Gaozhou 525200, China.
| | - Suiqun Guo
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510315, China.
| | - Weiyi Fang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China; The People's Hospital of Gaozhou, Gaozhou 525200, China; Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510315, China.
| |
Collapse
|
8
|
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
|
9
|
Shahcheraghi SH, Asl ER, Lotfi M, Ayatollahi J, Khaleghinejad SH, Aljabali AAA, Bakshi HA, El-Tanani M, Charbe NB, Serrano-Aroca Á, Mishra V, Mishra Y, Goyal R, Hromić-Jahjefendić A, Uversky VN, Lotfi M, Tambuwala MM. Non-coding RNAs as Key Regulators of the Notch Signaling Pathway in Glioblastoma: Diagnostic, Prognostic, and Therapeutic Targets. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1203-1216. [PMID: 38279763 DOI: 10.2174/0118715273277458231213063147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/20/2023] [Accepted: 10/31/2023] [Indexed: 01/28/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly invasive brain malignancy originating from astrocytes, accounting for approximately 30% of central nervous system malignancies. Despite advancements in therapeutic strategies including surgery, chemotherapy, and radiopharmaceutical drugs, the prognosis for GBM patients remains dismal. The aggressive nature of GBM necessitates the identification of molecular targets and the exploration of effective treatments to inhibit its proliferation. The Notch signaling pathway, which plays a critical role in cellular homeostasis, becomes deregulated in GBM, leading to increased expression of pathway target genes such as MYC, Hes1, and Hey1, thereby promoting cellular proliferation and differentiation. Recent research has highlighted the regulatory role of non-coding RNAs (ncRNAs) in modulating Notch signaling by targeting critical mRNA expression at the post-transcriptional or transcriptional levels. Specifically, various types of ncRNAs, including long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), have been shown to control multiple target genes and significantly contribute to the carcinogenesis of GBM. Furthermore, these ncRNAs hold promise as prognostic and predictive markers for GBM. This review aims to summarize the latest studies investigating the regulatory effects of ncRNAs on the Notch signaling pathway in GBM.
Collapse
Affiliation(s)
- Seyed Hossein Shahcheraghi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elmira Roshani Asl
- Social Determinants of Health Research Center, Saveh University of Medical Sciences, Saveh, Iran
| | - Malihe Lotfi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jamshid Ayatollahi
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Hematology and Oncology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Hamid A Bakshi
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Mohamed El-Tanani
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Nitin B Charbe
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics (Lake Nona), University of Florida, Orlando, FL, USA
| | - Ángel Serrano-Aroca
- Biomaterials & Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia, San Vicente Mártir, Valencia, 46001, Spain
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Yachana Mishra
- Department of Zoology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan, India
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, 71000 Sarajevo, Bosnia and Herzegovina
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Marzieh Lotfi
- Abortion Research Center, Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK
| |
Collapse
|
10
|
Bai Y, Bentley L, Ma C, Naveenan N, Cleak J, Wu Y, Simon MM, Westerberg H, Cañas RC, Horner N, Pandey R, Paphiti K, Schulze U, Mianné J, Hough T, Teboul L, de Baaij JH, Cox RD. Cleft palate and minor metabolic disturbances in a mouse global Arl15 gene knockout. FASEB J 2023; 37:e23211. [PMID: 37773757 PMCID: PMC10631251 DOI: 10.1096/fj.202201918r] [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: 11/21/2022] [Revised: 07/27/2023] [Accepted: 09/08/2023] [Indexed: 10/01/2023]
Abstract
ARL15, a small GTPase protein, was linked to metabolic traits in association studies. We aimed to test the Arl15 gene as a functional candidate for metabolic traits in the mouse. CRISPR/Cas9 germline knockout (KO) of Arl15 showed that homozygotes were postnatal lethal and exhibited a complete cleft palate (CP). Also, decreased cell migration was observed from Arl15 KO mouse embryonic fibroblasts (MEFs). Metabolic phenotyping of heterozygotes showed that females had reduced fat mass on a chow diet from 14 weeks of age. Mild body composition phenotypes were also observed in heterozygous mice on a high-fat diet (HFD)/low-fat diet (LFD). Females on a HFD showed reduced body weight, gonadal fat depot weight and brown adipose tissue (BAT) weight. In contrast, in the LFD group, females showed increased bone mineral density (BMD), while males showed a trend toward reduced BMD. Clinical biochemistry analysis of plasma on HFD showed transient lower adiponectin at 20 weeks of age in females. Urinary and plasma Mg2+ concentrations were not significantly different. Our phenotyping data showed that Arl15 is essential for craniofacial development. Adult metabolic phenotyping revealed potential roles in brown adipose tissue and bone development.
Collapse
Affiliation(s)
- Ying Bai
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Liz Bentley
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Chao Ma
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | - James Cleak
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Yixing Wu
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Michelle M Simon
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Henrik Westerberg
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Ramón Casero Cañas
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Neil Horner
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Rajesh Pandey
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Keanu Paphiti
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | | | - Joffrey Mianné
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Tertius Hough
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Lydia Teboul
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Jeroen H.F. de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Roger D. Cox
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| |
Collapse
|
11
|
LaGuardia JS, Shariati K, Bedar M, Ren X, Moghadam S, Huang KX, Chen W, Kang Y, Yamaguchi DT, Lee JC. Convergence of Calcium Channel Regulation and Mechanotransduction in Skeletal Regenerative Biomaterial Design. Adv Healthc Mater 2023; 12:e2301081. [PMID: 37380172 PMCID: PMC10615747 DOI: 10.1002/adhm.202301081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/20/2023] [Indexed: 06/30/2023]
Abstract
Cells are known to perceive their microenvironment through extracellular and intracellular mechanical signals. Upon sensing mechanical stimuli, cells can initiate various downstream signaling pathways that are vital to regulating proliferation, growth, and homeostasis. One such physiologic activity modulated by mechanical stimuli is osteogenic differentiation. The process of osteogenic mechanotransduction is regulated by numerous calcium ion channels-including channels coupled to cilia, mechanosensitive and voltage-sensitive channels, and channels associated with the endoplasmic reticulum. Evidence suggests these channels are implicated in osteogenic pathways such as the YAP/TAZ and canonical Wnt pathways. This review aims to describe the involvement of calcium channels in regulating osteogenic differentiation in response to mechanical loading and characterize the fashion in which those channels directly or indirectly mediate this process. The mechanotransduction pathway is a promising target for the development of regenerative materials for clinical applications due to its independence from exogenous growth factor supplementation. As such, also described are examples of osteogenic biomaterial strategies that involve the discussed calcium ion channels, calcium-dependent cellular structures, or calcium ion-regulating cellular features. Understanding the distinct ways calcium channels and signaling regulate these processes may uncover potential targets for advancing biomaterials with regenerative osteogenic capabilities.
Collapse
Affiliation(s)
- Jonnby S. LaGuardia
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Kaavian Shariati
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Meiwand Bedar
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Xiaoyan Ren
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Shahrzad Moghadam
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Kelly X. Huang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Wei Chen
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Youngnam Kang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Dean T. Yamaguchi
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Justine C. Lee
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
- Department of Orthopaedic Surgery, Los Angeles, CA, 90095, USA
- UCLA Molecular Biology Institute, Los Angeles, CA, 90095, USA
| |
Collapse
|
12
|
Okada Y, Numata T, Sabirov RZ, Kashio M, Merzlyak PG, Sato-Numata K. Cell death induction and protection by activation of ubiquitously expressed anion/cation channels. Part 3: the roles and properties of TRPM2 and TRPM7. Front Cell Dev Biol 2023; 11:1246955. [PMID: 37842082 PMCID: PMC10576435 DOI: 10.3389/fcell.2023.1246955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Cell volume regulation (CVR) is a prerequisite for animal cells to survive and fulfill their functions. CVR dysfunction is essentially involved in the induction of cell death. In fact, sustained normotonic cell swelling and shrinkage are associated with necrosis and apoptosis, and thus called the necrotic volume increase (NVI) and the apoptotic volume decrease (AVD), respectively. Since a number of ubiquitously expressed ion channels are involved in the CVR processes, these volume-regulatory ion channels are also implicated in the NVI and AVD events. In Part 1 and Part 2 of this series of review articles, we described the roles of swelling-activated anion channels called VSOR or VRAC and acid-activated anion channels called ASOR or PAC in CVR and cell death processes. Here, Part 3 focuses on therein roles of Ca2+-permeable non-selective TRPM2 and TRPM7 cation channels activated by stress. First, we summarize their phenotypic properties and molecular structure. Second, we describe their roles in CVR. Since cell death induction is tightly coupled to dysfunction of CVR, third, we focus on their participation in the induction of or protection against cell death under oxidative, acidotoxic, excitotoxic, and ischemic conditions. In this regard, we pay attention to the sensitivity of TRPM2 and TRPM7 to a variety of stress as well as to their capability to physicall and functionally interact with other volume-related channels and membrane enzymes. Also, we summarize a large number of reports hitherto published in which TRPM2 and TRPM7 channels are shown to be involved in cell death associated with a variety of diseases or disorders, in some cases as double-edged swords. Lastly, we attempt to describe how TRPM2 and TRPM7 are organized in the ionic mechanisms leading to cell death induction and protection.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Cardiovascular Research Institute, Yokohama City University, Yokohama, Japan
| | - Tomohiro Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| | - Ravshan Z. Sabirov
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Makiko Kashio
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
| | - Peter G. Merzlyak
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Kaori Sato-Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| |
Collapse
|
13
|
Hoeger B, Nadolni W, Hampe S, Hoelting K, Fraticelli M, Zaborsky N, Madlmayr A, Sperrer V, Fraticelli L, Addington L, Steinritz D, Chubanov V, Geisberger R, Greil R, Breit A, Boekhoff I, Gudermann T, Zierler S. Inactivation of TRPM7 Kinase Targets AKT Signaling and Cyclooxygenase-2 Expression in Human CML Cells. FUNCTION 2023; 4:zqad053. [PMID: 37786778 PMCID: PMC10541797 DOI: 10.1093/function/zqad053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 10/04/2023] Open
Abstract
Cyclooxygenase-2 (COX-2) is a key regulator of inflammation. High constitutive COX-2 expression enhances survival and proliferation of cancer cells, and adversely impacts antitumor immunity. The expression of COX-2 is modulated by various signaling pathways. Recently, we identified the melastatin-like transient-receptor-potential-7 (TRPM7) channel-kinase as modulator of immune homeostasis. TRPM7 protein is essential for leukocyte proliferation and differentiation, and upregulated in several cancers. It comprises of a cation channel and an atypical α-kinase, linked to inflammatory cell signals and associated with hallmarks of tumor progression. A role in leukemia has not been established, and signaling pathways are yet to be deciphered. We show that inhibiting TRPM7 channel-kinase in chronic myeloid leukemia (CML) cells results in reduced constitutive COX-2 expression. By utilizing a CML-derived cell line, HAP1, harboring CRISPR/Cas9-mediated TRPM7 knockout, or a point mutation inactivating TRPM7 kinase, we could link this to reduced activation of AKT serine/threonine kinase and mothers against decapentaplegic homolog 2 (SMAD2). We identified AKT as a direct in vitro substrate of TRPM7 kinase. Pharmacologic blockade of TRPM7 in wildtype HAP1 cells confirmed the effect on COX-2 via altered AKT signaling. Addition of an AKT activator on TRPM7 kinase-dead cells reconstituted the wildtype phenotype. Inhibition of TRPM7 resulted in reduced phosphorylation of AKT and diminished COX-2 expression in peripheral blood mononuclear cells derived from CML patients, and reduced proliferation in patient-derived CD34+ cells. These results highlight a role of TRPM7 kinase in AKT-driven COX-2 expression and suggest a beneficial potential of TRPM7 blockade in COX-2-related inflammation and malignancy.
Collapse
Affiliation(s)
- Birgit Hoeger
- Institute of Pharmacology, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz and Krankenhausstr. 5, 4020 Linz, Austria
| | - Wiebke Nadolni
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Sarah Hampe
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Kilian Hoelting
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Marco Fraticelli
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Nadja Zaborsky
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute–Laboratory for Immunological and Molecular Cancer Research (SCRI–LIMCR), Müllner Hauptstr. 48, 5020 Salzburg, Austria
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Anna Madlmayr
- Institute of Pharmacology, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz and Krankenhausstr. 5, 4020 Linz, Austria
| | - Viktoria Sperrer
- Institute of Pharmacology, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz and Krankenhausstr. 5, 4020 Linz, Austria
| | - Laura Fraticelli
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Lynda Addington
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Dirk Steinritz
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Vladimir Chubanov
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Roland Geisberger
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute–Laboratory for Immunological and Molecular Cancer Research (SCRI–LIMCR), Müllner Hauptstr. 48, 5020 Salzburg, Austria
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute–Laboratory for Immunological and Molecular Cancer Research (SCRI–LIMCR), Müllner Hauptstr. 48, 5020 Salzburg, Austria
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Andreas Breit
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Ingrid Boekhoff
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| | - Susanna Zierler
- Institute of Pharmacology, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz and Krankenhausstr. 5, 4020 Linz, Austria
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany
| |
Collapse
|
14
|
Xing F, Dong H, Yang J, Fan C, Hou M, Zhang P, Hu F, Zhou J, Chen L, Pan L, Xu J. Mesenchymal Migration on Adhesive-Nonadhesive Alternate Surfaces in Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301337. [PMID: 37211690 PMCID: PMC10427406 DOI: 10.1002/advs.202301337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/14/2023] [Indexed: 05/23/2023]
Abstract
Mesenchymal migration usually happens on adhesive substrates, while cells adopt amoeboid migration on low/nonadhesive surfaces. Protein-repelling reagents, e.g., poly(ethylene) glycol (PEG), are routinely employed to resist cell adhering and migrating. Contrary to these perceptions, this work discovers a unique locomotion of macrophages on adhesive-nonadhesive alternate substrates in vitro that they can overcome nonadhesive PEG gaps to reach adhesive regions in the mesenchymal mode. Adhering to extracellular matrix regions is a prerequisite for macrophages to perform further locomotion on the PEG regions. Podosomes are found highly enriched on the PEG region in macrophages and support their migration across the nonadhesive regions. Increasing podosome density through myosin IIA inhibition facilitates cell motility on adhesive-nonadhesive alternate substrates. Moreover, a developed cellular Potts model reproduces this mesenchymal migration. These findings together uncover a new migratory behavior on adhesive-nonadhesive alternate substrates in macrophages.
Collapse
Affiliation(s)
- Fulin Xing
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin, 300071, China
| | - Hao Dong
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin, 300071, China
| | - Jianyu Yang
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin, 300071, China
| | - Chunhui Fan
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin, 300071, China
| | - Mengdi Hou
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin, 300071, China
| | - Ping Zhang
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin, 300071, China
| | - Fen Hu
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin, 300071, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Liangyi Chen
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, National Biomedical Imaging Center, Center for Life Sciences, School of Future Technology, Peking University, Beijing, 100871, China
| | - Leiting Pan
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
- Shenzhen Research Institute of Nankai University, Shenzhen, Guangdong, 518083, China
| | - Jingjun Xu
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin, 300071, China
- Shenzhen Research Institute of Nankai University, Shenzhen, Guangdong, 518083, China
| |
Collapse
|
15
|
Mahbub L, Kozlov G, Zong P, Lee EL, Tetteh S, Nethramangalath T, Knorn C, Jiang J, Shahsavan A, Yue L, Runnels L, Gehring K. Structural insights into regulation of CNNM-TRPM7 divalent cation uptake by the small GTPase ARL15. eLife 2023; 12:e86129. [PMID: 37449820 PMCID: PMC10348743 DOI: 10.7554/elife.86129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023] Open
Abstract
Cystathionine-β-synthase (CBS)-pair domain divalent metal cation transport mediators (CNNMs) are an evolutionarily conserved family of magnesium transporters. They promote efflux of Mg2+ ions on their own and influx of divalent cations when expressed with the transient receptor potential ion channel subfamily M member 7 (TRPM7). Recently, ADP-ribosylation factor-like GTPase 15 (ARL15) has been identified as CNNM-binding partner and an inhibitor of divalent cation influx by TRPM7. Here, we characterize ARL15 as a GTP and CNNM-binding protein and demonstrate that ARL15 also inhibits CNNM2 Mg2+ efflux. The crystal structure of a complex between ARL15 and CNNM2 CBS-pair domain reveals the molecular basis for binding and allowed the identification of mutations that specifically block binding. A binding deficient ARL15 mutant, R95A, failed to inhibit CNNM and TRPM7 transport of Mg2+ and Zn2+ ions. Structural analysis and binding experiments with phosphatase of regenerating liver 2 (PRL2 or PTP4A2) showed that ARL15 and PRLs compete for binding CNNM to coordinate regulation of ion transport by CNNM and TRPM7.
Collapse
Affiliation(s)
- Luba Mahbub
- Department of Biochemistry, McGill UniversityMontrealCanada
- Centre de recherche en biologie structurale, McGill UniversityMontréalCanada
| | - Guennadi Kozlov
- Department of Biochemistry, McGill UniversityMontrealCanada
- Centre de recherche en biologie structurale, McGill UniversityMontréalCanada
| | - Pengyu Zong
- Department of Cell Biology, UCONN Health CenterFarmingtonUnited States
| | - Emma L Lee
- Department of Biochemistry, McGill UniversityMontrealCanada
- Centre de recherche en biologie structurale, McGill UniversityMontréalCanada
| | - Sandra Tetteh
- Rutgers-Robert Wood Johnson Medical SchoolPiscatawayUnited States
| | | | - Caroline Knorn
- Department of Biochemistry, McGill UniversityMontrealCanada
- Centre de recherche en biologie structurale, McGill UniversityMontréalCanada
| | - Jianning Jiang
- Department of Biochemistry, McGill UniversityMontrealCanada
- Centre de recherche en biologie structurale, McGill UniversityMontréalCanada
| | - Ashkan Shahsavan
- Department of Biochemistry, McGill UniversityMontrealCanada
- Centre de recherche en biologie structurale, McGill UniversityMontréalCanada
| | - Lixia Yue
- Department of Cell Biology, UCONN Health CenterFarmingtonUnited States
| | - Loren Runnels
- Rutgers-Robert Wood Johnson Medical SchoolPiscatawayUnited States
| | - Kalle Gehring
- Department of Biochemistry, McGill UniversityMontrealCanada
- Centre de recherche en biologie structurale, McGill UniversityMontréalCanada
| |
Collapse
|
16
|
Zhang S, Cao F, Li W, Abumaria N. TRPM7 kinase activity induces amyloid-β degradation to reverse synaptic and cognitive deficits in mouse models of Alzheimer's disease. Sci Signal 2023; 16:eade6325. [PMID: 37433006 DOI: 10.1126/scisignal.ade6325] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 06/15/2023] [Indexed: 07/13/2023]
Abstract
Altered abundance or activity of the dual-function transient receptor potential melastatin-like 7 (TRPM7) protein is implicated in neurodegenerative disorders, including Alzheimer's disease (AD). Toxic aggregation of amyloid-β (Aβ) in neurons is implicated in AD pathology. Here, we found that the kinase activity of TRPM7 is important to stimulate the degradation of Aβ. TRPM7 expression was decreased in hippocampal tissue samples from patients with AD and two mouse models of AD (APP/PS1 and 5XFAD). In cultures of hippocampal neurons from mice, overexpression of full-length TRPM7 or of its functional kinase domain M7CK prevented synapse loss induced by exogenous Aβ. In contrast, this neuroprotection was not afforded by overexpression of either the functional ion channel portion alone or a TRPM7 mutant lacking kinase activity. M7CK overexpression in the hippocampus of young and old 5XFAD mice prevented and reversed, respectively, memory deficits, synapse loss, and Aβ plaque accumulation. In both neurons and mice, M7CK interacted with and activated the metalloprotease MMP14 to promote Aβ degradation. Thus, TRPM7 loss in patients with AD may contribute to the associated Aβ pathology.
Collapse
Affiliation(s)
- Shimeng Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Feifei Cao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Wei Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Nashat Abumaria
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
The TRPM7 channel reprograms cellular glycolysis to drive tumorigenesis and angiogenesis. Cell Death Dis 2023; 14:183. [PMID: 36878949 PMCID: PMC9988972 DOI: 10.1038/s41419-023-05701-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/08/2023]
Abstract
Cancer or endothelial cells preferably catabolize glucose through aerobic glycolysis rather than oxidative phosphorylation. Intracellular ionic signaling has been shown to regulate glucose metabolism, but the underlying ion channel has yet to be identified. RNA-seq, metabolomics and genetic assay revealed that the TRPM7 channel regulated cellular glycolysis. Deletion of TRPM7 suppressed cancer cell glycolysis and reduced the xenograft tumor burden. Deficiency of endothelial TRPM7 inhibited postnatal retinal angiogenesis in mice. Mechanistically, TRPM7 transcriptionally regulated the solute carrier family 2 member 3 (SLC2A3, also known as GLUT3) via Ca2+ influx-induced calcineurin activation. Furthermore, CREB-regulated transcription coactivator 2 (CRTC2) and CREB act downstream of calcineurin to relay Ca2+ signal to SLC2A3 transcription. Expression of the constitutively active CRTC2 or CREB in TRPM7 knockout cell normalized glycolytic metabolism and cell growth. The TRPM7 channel represents a novel regulator of glycolytic reprogramming. Inhibition of the TRPM7-dependent glycolysis could be harnessed for cancer therapy.
Collapse
|
18
|
Turlova E, Ji D, Deurloo M, Wong R, Fleig A, Horgen FD, Sun HS, Feng ZP. Hypoxia-Induced Neurite Outgrowth Involves Regulation Through TRPM7. Mol Neurobiol 2023; 60:836-850. [PMID: 36378470 DOI: 10.1007/s12035-022-03114-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 10/26/2022] [Indexed: 11/16/2022]
Abstract
Transient receptor potential melastatin 7 (TRPM7) is a ubiquitously expressed divalent cation channel that plays a key role in cell functions such as ion homeostasis, cell proliferation, survival, and cytoskeletal dynamics and mediates cells death in hypoxic and ischemic conditions. Previously, TRPM7 was found to play a role in the neurite outgrowth and maturation of primary hippocampal neurons. Either knockdown of TRPM7 with target-specific shRNA or blocking channel conductance by a specific blocker waixenicin A enhanced axonal outgrowth in the primary neuronal culture. In this study, we investigated whether and how TPRM7 is involved in hypoxia-altered neurite outgrowth patterns in E16 hippocampal neuron cultures. We demonstrate that short-term hypoxia activated the MEK/ERK and PI3K/Akt pathways, reduced TRPM7 activity, and enhanced axonal outgrowth of neuronal cultures. On the other hand, long-term hypoxia caused a progressive retraction of axons and dendrites that could be attenuated by the TRPM7-specific inhibitor waixenicin A. Further, we demonstrate that in the presence of astrocytes, axonal retraction in long-term hypoxic conditions was enhanced, and TRPM7 block by waixenicin A prevented this retraction. Our data demonstrate the effect of hypoxia on TRPM7 activity and axonal outgrowth/retraction in cultures with or without astrocytes present.
Collapse
Affiliation(s)
- Ekaterina Turlova
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Ontario, M5S 1A8, Toronto, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Delphine Ji
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Ontario, M5S 1A8, Toronto, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Marielle Deurloo
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Raymond Wong
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Ontario, M5S 1A8, Toronto, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine and Cancer Center at the, University of Hawaii, Honolulu, HI, 96720, USA
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI, 96744, USA
| | - Hong-Shuo Sun
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Ontario, M5S 1A8, Toronto, Canada.
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
- Department of Pharmacology, Temerty Faculty of Medicine, University of Toronto, King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
19
|
Adipose-specific deletion of the cation channel TRPM7 inhibits TAK1 kinase-dependent inflammation and obesity in male mice. Nat Commun 2023; 14:491. [PMID: 36717580 PMCID: PMC9887063 DOI: 10.1038/s41467-023-36154-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 01/18/2023] [Indexed: 01/31/2023] Open
Abstract
Chronic inflammation of white adipose tissue is a key link between obesity and the associated metabolic syndrome. Transient receptor potential melastatin-like 7 (TRPM7) is known to be related to inflammation; however, the role of TRPM7 in adipocyte phenotype and function in obesity remains unclear. Here, we observe that the activation of adipocyte TRPM7 plays an essential role in pro-inflammatory responses. Adult male mice are used in our experiments. Adipocyte-specific deficiency in TRPM7 attenuates the pro-inflammatory phenotype, improves glucose homeostasis, and suppresses weight gain in mice fed a high-fat diet. Mechanistically, the pro-inflammatory effect of TRPM7 is dependent on Ca2+ signaling. Ca2+ influx initiated by TRPM7 enhances transforming growth factor-β activated kinase 1 activation via the co-regulation of calcium/calmodulin-dependent protein kinase II and tumor necrosis factor receptor-associated factor 6, leading to exacerbated nuclear factor kappa B signaling. Additionally, obese mice treated with TRPM7 inhibitor are protected against obesity and insulin resistance. Our results demonstrate TRPM7 as a factor in the development of adipose inflammation that regulates insulin sensitivity in obesity.
Collapse
|
20
|
Forzisi E, Sesti F. Non-conducting functions of ion channels: The case of integrin-ion channel complexes. Channels (Austin) 2022; 16:185-197. [PMID: 35942524 PMCID: PMC9364710 DOI: 10.1080/19336950.2022.2108565] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Started as an academic curiosity more than two decades ago, the idea that ion channels can regulate cellular processes in ways that do not depend on their conducting properties (non-ionic functions) gained traction and is now a flourishing area of research. Channels can regulate physiological processes including actin cytoskeletal remodeling, cell motility, excitation-contraction coupling, non-associative learning and embryogenesis, just to mention some, through non-ionic functions. When defective, non-ionic functions can give rise to channelopathies involved in cancer, neurodegenerative disease and brain trauma. Ion channels exert their non-ionic functions through a variety of mechanisms that range from physical coupling with other proteins, to possessing enzymatic activity, to assembling with signaling molecules. In this article, we take stock of the field and review recent findings. The concept that emerges, is that one of the most common ways through which channels acquire non-ionic attributes, is by assembling with integrins. These integrin-channel complexes exhibit broad genotypic and phenotypic heterogeneity and reveal a pleiotropic nature, as they appear to be capable of influencing both physiological and pathological processes.
Collapse
Affiliation(s)
- Elena Forzisi
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| |
Collapse
|
21
|
Yao M, Tijore A, Cheng D, Li JV, Hariharan A, Martinac B, Tran Van Nhieu G, Cox CD, Sheetz M. Force- and cell state-dependent recruitment of Piezo1 drives focal adhesion dynamics and calcium entry. SCIENCE ADVANCES 2022; 8:eabo1461. [PMID: 36351022 PMCID: PMC9645726 DOI: 10.1126/sciadv.abo1461] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/21/2022] [Indexed: 11/11/2022]
Abstract
Mechanosensing is an integral part of many physiological processes including stem cell differentiation, fibrosis, and cancer progression. Two major mechanosensing systems-focal adhesions and mechanosensitive ion channels-can convert mechanical features of the microenvironment into biochemical signals. We report here unexpectedly that the mechanosensitive calcium-permeable channel Piezo1, previously perceived to be diffusive on plasma membranes, binds to matrix adhesions in a force-dependent manner, promoting cell spreading, adhesion dynamics, and calcium entry in normal but not in most cancer cells tested except some glioblastoma lines. A linker domain in Piezo1 is needed for binding to adhesions, and overexpression of the domain blocks Piezo1 binding to adhesions, decreasing adhesion size and cell spread area. Thus, we suggest that Piezo1 is a previously unidentified component of focal adhesions in nontransformed cells that catalyzes adhesion maturation and growth through force-dependent calcium signaling, but this function is absent in most cancer cells.
Collapse
Affiliation(s)
- Mingxi Yao
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Corresponding author. (M.Y); (C.C.); (M.S.)
| | - Ajay Tijore
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Center for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Delfine Cheng
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
| | - Jinyuan Vero Li
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
| | - Anushya Hariharan
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
| | - Guy Tran Van Nhieu
- Ecole Normale Supérieure Paris-Saclay Gif-sur-Yvette, France
- Team Ca Signaling and Microbial Infections, Institute for Integrative Biology of the Cell–CNRS UMR9198–Inserm U1280, 1, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Charles D. Cox
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
- Corresponding author. (M.Y); (C.C.); (M.S.)
| | - Michael Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Department of Biological Sciences, National University of Singapore, Singapore 117558
- Molecular MechanoMedicine Program, Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
- Corresponding author. (M.Y); (C.C.); (M.S.)
| |
Collapse
|
22
|
Svec KV, Howe AK. Protein Kinase A in cellular migration-Niche signaling of a ubiquitous kinase. Front Mol Biosci 2022; 9:953093. [PMID: 35959460 PMCID: PMC9361040 DOI: 10.3389/fmolb.2022.953093] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/30/2022] [Indexed: 12/28/2022] Open
Abstract
Cell migration requires establishment and maintenance of directional polarity, which in turn requires spatial heterogeneity in the regulation of protrusion, retraction, and adhesion. Thus, the signaling proteins that regulate these various structural processes must also be distinctly regulated in subcellular space. Protein Kinase A (PKA) is a ubiquitous serine/threonine kinase involved in innumerable cellular processes. In the context of cell migration, it has a paradoxical role in that global inhibition or activation of PKA inhibits migration. It follows, then, that the subcellular regulation of PKA is key to bringing its proper permissive and restrictive functions to the correct parts of the cell. Proper subcellular regulation of PKA controls not only when and where it is active but also specifies the targets for that activity, allowing the cell to use a single, promiscuous kinase to exert distinct functions within different subcellular niches to facilitate cell movement. In this way, understanding PKA signaling in migration is a study in context and in the elegant coordination of distinct functions of a single protein in a complex cellular process.
Collapse
Affiliation(s)
- Kathryn V. Svec
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
| | - Alan K. Howe
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, V T, United States
- University of Vermont Cancer Center, University of Vermont, Burlington, VT, United States
| |
Collapse
|
23
|
Efferocytosis requires periphagosomal Ca 2+-signaling and TRPM7-mediated electrical activity. Nat Commun 2022; 13:3230. [PMID: 35680919 PMCID: PMC9184625 DOI: 10.1038/s41467-022-30959-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 05/26/2022] [Indexed: 11/21/2022] Open
Abstract
Efficient clearance of apoptotic cells by phagocytosis, also known as efferocytosis, is fundamental to developmental biology, organ physiology, and immunology. Macrophages use multiple mechanisms to detect and engulf apoptotic cells, but the signaling pathways that regulate the digestion of the apoptotic cell cargo, such as the dynamic Ca2+ signals, are poorly understood. Using an siRNA screen, we identify TRPM7 as a Ca2+-conducting ion channel essential for phagosome maturation during efferocytosis. Trpm7-targeted macrophages fail to fully acidify or digest their phagosomal cargo in the absence of TRPM7. Through perforated patch electrophysiology, we demonstrate that TRPM7 mediates a pH-activated cationic current necessary to sustain phagosomal acidification. Using mice expressing a genetically-encoded Ca2+ sensor, we observe that phagosome maturation requires peri-phagosomal Ca2+-signals dependent on TRPM7. Overall, we reveal TRPM7 as a central regulator of phagosome maturation during macrophage efferocytosis. Efficient removal of apoptotic cells by phagocytosis underlies tissue development, wound repair, host defense and organ homeostasis. Here, authors identify TRPM7 as a regulator of cargo acidification and Ca2+ signaling during apoptotic cell clearance.
Collapse
|
24
|
LncRNA HOTAIR sponges miR-301a-3p to promote glioblastoma proliferation and invasion through upregulating FOSL1. Cell Signal 2022; 94:110306. [PMID: 35292358 PMCID: PMC9058208 DOI: 10.1016/j.cellsig.2022.110306] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 01/29/2023]
Abstract
Glioblastoma, one of the most fatal brain tumors, is associated with a dismal prognosis and an extremely short overall survival. We previously reported that the overexpressed transient receptor potential channel TRPM7 is an essential glioblastoma regulator. Accumulating evidence suggests that long noncoding RNAs (lncRNAs) play an important role in glioma's initiation and progression. However, the function of lncRNA, HOX transcript antisense intergenic RNA (HOTAIR) mediated by TRPM7 in glioma remains unclear. In this study, HOTAIR expression was found to be positively regulated by TRPM7, significantly upregulated in glioma tissues, and is a poor prognosis factor for glioma patients. Moreover, reduced HOTAIR expression impeded the proliferation and invasion of glioma cells. Mechanistically, HOTAIR directly interacted with miR-301a-3p, and downregulation of miR-301a-3p efficiently reversed FOSL1 suppression induced by siRNA HOTAIR, which implied that HOTAIR positively regulated FOSL1 level through sponging miR-301a-3p and played an oncogenic role in glioma progression. In contrast to HOTAIR's role, miR-301a-3p alone served as a tumor suppressor to decrease glioma cell viability and migration/invasion. In agreement with HOTAIR's role, FOSL1 functioned as a tumorigenic gene in glioma pathogenesis, which was highly expressed in glioma tissues, and was shown to be an unfavorable prognostic factor for glioma patients. Mechanically, FOSL1 inhibition by siRNA FOSL1 efficiently rescued the oncogenic-like phenotypes caused by the miR-301a-3p inhibitor in glioma pathogenesis. SIGNIFICANCE: Our study elucidated the role of TRPM7-mediated HOTAIR as a miRNA sponge to target downstream FOSL1 oncogene and therefore consequently contribute to gliomagenesis, which shed new light on TRPM7/lncRNA-directed diagnostic and therapeutic approach in glioma.
Collapse
|
25
|
Souza Bomfim GH, Niemeyer BA, Lacruz RS, Lis A. On the Connections between TRPM Channels and SOCE. Cells 2022; 11:1190. [PMID: 35406753 PMCID: PMC8997886 DOI: 10.3390/cells11071190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 12/02/2022] Open
Abstract
Plasma membrane protein channels provide a passageway for ions to access the intracellular milieu. Rapid entry of calcium ions into cells is controlled mostly by ion channels, while Ca2+-ATPases and Ca2+ exchangers ensure that cytosolic Ca2+ levels ([Ca2+]cyt) are maintained at low (~100 nM) concentrations. Some channels, such as the Ca2+-release-activated Ca2+ (CRAC) channels and voltage-dependent Ca2+ channels (CACNAs), are highly Ca2+-selective, while others, including the Transient Receptor Potential Melastatin (TRPM) family, have broader selectivity and are mostly permeable to monovalent and divalent cations. Activation of CRAC channels involves the coupling between ORAI1-3 channels with the endoplasmic reticulum (ER) located Ca2+ store sensor, Stromal Interaction Molecules 1-2 (STIM1/2), a pathway also termed store-operated Ca2+ entry (SOCE). The TRPM family is formed by 8 members (TRPM1-8) permeable to Mg2+, Ca2+, Zn2+ and Na+ cations, and is activated by multiple stimuli. Recent studies indicated that SOCE and TRPM structure-function are interlinked in some instances, although the molecular details of this interaction are only emerging. Here we review the role of TRPM and SOCE in Ca2+ handling and highlight the available evidence for this interaction.
Collapse
Affiliation(s)
- Guilherme H. Souza Bomfim
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Barbara A. Niemeyer
- Department of Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany;
| | - Rodrigo S. Lacruz
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Annette Lis
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
26
|
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
|
27
|
TRPM7 Ion Channel: Oncogenic Roles and Therapeutic Potential in Breast Cancer. Cancers (Basel) 2021; 13:cancers13246322. [PMID: 34944940 PMCID: PMC8699295 DOI: 10.3390/cancers13246322] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Breast cancer is the most frequently diagnosed malignant tumor and the second leading cause of cancer death in women worldwide. The risk of developing breast cancer is 12.8%, i.e., 1 in 8 people, and a woman’s risk of dying is approximately 1 in 39. Calcium signals play an important role in various cancers and transport calcium ions may have altered expression in breast cancer, such as the TRPM7 calcium permeant ion channel, where overexpression may be associated with a poor prognosis. This review focuses on the TRPM7 channel, and the oncogenic roles studied so far in breast cancer. The TRPM7 ion channel is suggested as a potential and prospective target in the diagnosis and treatment of breast cancer. Abstract The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a divalent cations permeant channel but also has intrinsic serine/threonine kinase activity. It is ubiquitously expressed in normal tissues and studies have indicated that it participates in important physiological and pharmacological processes through its channel-kinase activity, such as calcium/magnesium homeostasis, phosphorylation of proteins involved in embryogenesis or the cellular process. Accumulating evidence has shown that TRPM7 is overexpressed in human pathologies including breast cancer. Breast cancer is the second leading cause of cancer death in women with an incidence rate increase of around 0.5% per year since 2004. The overexpression of TRPM7 may be associated with a poor prognosis in breast cancer patients, so more efforts are needed to research a new therapeutic target. TRPM7 regulates the levels of Ca2+, which can alter the signaling pathways involved in survival, cell cycle progression, proliferation, growth, migration, invasion, epithelial-mesenchymal transition and thus determines cell behavior, promoting tumor development. This work provides a complete overview of the TRPM7 ion channel and its main involvements in breast cancer. Special consideration is given to the modulation of the channel as a potential target in breast cancer treatment by inhibition of proliferation, migration and invasion. Taken together, these data suggest the potential exploitation of TRPM7 channel-kinase as a therapeutic target and a diagnostic biomarker.
Collapse
|
28
|
Characterization of IL-2 Stimulation and TRPM7 Pharmacomodulation in NK Cell Cytotoxicity and Channel Co-Localization with PIP 2 in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Patients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182211879. [PMID: 34831634 PMCID: PMC8618557 DOI: 10.3390/ijerph182211879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/30/2021] [Accepted: 11/08/2021] [Indexed: 12/01/2022]
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex multisystemic disorder responsible for significant disability. Although a unifying etiology for ME/CFS is uncertain, impaired natural killer (NK) cell cytotoxicity represents a consistent and measurable feature of this disorder. Research utilizing patient-derived NK cells has implicated dysregulated calcium (Ca2+) signaling, dysfunction of the phosphatidylinositol-4,5-bisphosphate (PIP2)-dependent cation channel, transient receptor potential melastatin (TRPM) 3, as well as altered surface expression patterns of TRPM3 and TRPM2 in the pathophysiology of ME/CFS. TRPM7 is a related channel that is modulated by PIP2 and participates in Ca2+ signaling. Though TRPM7 is expressed on NK cells, the role of TRPM7 with IL-2 and intracellular signaling mechanisms in the NK cells of ME/CFS patients is unknown. This study examined the effect of IL-2 stimulation and TRPM7 pharmacomodulation on NK cell cytotoxicity using flow cytometric assays as well as co-localization of TRPM7 with PIP2 and cortical actin using confocal microscopy in 17 ME/CFS patients and 17 age- and sex-matched healthy controls. The outcomes of this investigation are preliminary and indicate that crosstalk between IL-2 and TRMP7 exists. A larger sample size to confirm these findings and characterization of TRPM7 in ME/CFS using other experimental modalities are warranted.
Collapse
|
29
|
Trophectoderm cell failure leads to peri-implantation lethality in Trpm7-deficient mouse embryos. Cell Rep 2021; 37:109851. [PMID: 34686339 DOI: 10.1016/j.celrep.2021.109851] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/28/2021] [Accepted: 09/28/2021] [Indexed: 11/24/2022] Open
Abstract
Early embryogenesis depends on proper control of intracellular homeostasis of ions including Ca2+ and Mg2+. Deletion of the Ca2+ and Mg2+ conducting the TRPM7 channel is embryonically lethal in mice but leaves compaction, blastomere polarization, blastocoel formation, and correct specification of the lineages of the trophectoderm and inner cell mass unaltered despite that free cytoplasmic Ca2+ and Mg2+ is reduced at the two-cell stage. Although Trpm7-/- embryos are able to hatch from the zona pellucida, no expansion of Trpm7-/- trophoblast cells can be observed, and Trpm7-/- embryos are not identifiable in utero at E6.5 or later. Given the proliferation and adhesion defect of Trpm7-/- trophoblast stem cells and the ability of Trpm7-/- ESCs to develop to embryos in tetraploid embryo complementation assays, we postulate a critical role of TRPM7 in trophectoderm cells and their failure during implantation as the most likely explanation of the developmental arrest of Trpm7-deficient mouse embryos.
Collapse
|
30
|
Du Preez S, Cabanas H, Staines D, Marshall-Gradisnik S. Potential Implications of Mammalian Transient Receptor Potential Melastatin 7 in the Pathophysiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:10708. [PMID: 34682454 PMCID: PMC8535478 DOI: 10.3390/ijerph182010708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 11/25/2022]
Abstract
The transient receptor potential (TRP) superfamily of ion channels is involved in the molecular mechanisms that mediate neuroimmune interactions and activities. Recent advancements in neuroimmunology have identified a role for TRP cation channels in several neuroimmune disorders including amyotropic lateral sclerosis, multiple sclerosis, and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). ME/CFS is a debilitating disorder with an obscure aetiology, hence considerable examination of its pathobiology is warranted. Dysregulation of TRP melastatin (TRPM) subfamily members and calcium signalling processes are implicated in the neurological, immunological, cardiovascular, and metabolic impairments inherent in ME/CFS. In this review, we present TRPM7 as a potential candidate in the pathomechanism of ME/CFS, as TRPM7 is increasingly recognized as a key mediator of physiological and pathophysiological mechanisms affecting neurological, immunological, cardiovascular, and metabolic processes. A focused examination of the biochemistry of TRPM7, the role of this protein in the aforementioned systems, and the potential of TRPM7 as a molecular mechanism in the pathophysiology of ME/CFS will be discussed in this review. TRPM7 is a compelling candidate to examine in the pathobiology of ME/CFS as TRPM7 fulfils several key roles in multiple organ systems, and there is a paucity of literature reporting on its role in ME/CFS.
Collapse
Affiliation(s)
- Stanley Du Preez
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast 4215, Australia; (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Menzies Health Institute Queensland, Griffith University, Gold Coast 4215, Australia;
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast 4215, Australia
- School of Medicine and Dentistry, Griffith University, Gold Coast 4215, Australia
| | - Helene Cabanas
- Consortium Health International for Myalgic Encephalomyelitis, Menzies Health Institute Queensland, Griffith University, Gold Coast 4215, Australia;
- Institut de Recherche Saint Louis, Université de Paris, INSERM U944 and CNRS UMR 7212, Hôpital Saint Louis, APHP, 75010 Paris, France
| | - Donald Staines
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast 4215, Australia; (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Menzies Health Institute Queensland, Griffith University, Gold Coast 4215, Australia;
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast 4215, Australia; (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Menzies Health Institute Queensland, Griffith University, Gold Coast 4215, Australia;
| |
Collapse
|
31
|
Liang HY, Chen Y, Wei X, Ma GG, Ding J, Lu C, Zhou RP, Hu W. Immunomodulatory functions of TRPM7 and its implications in autoimmune diseases. Immunology 2021; 165:3-21. [PMID: 34558663 DOI: 10.1111/imm.13420] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 08/17/2021] [Accepted: 09/14/2021] [Indexed: 12/18/2022] Open
Abstract
An autoimmune disease is an inappropriate response to one's tissues due to a break in immune tolerance and exposure to self-antigens. It often leads to structural and functional damage to organs and systemic disorders. To date, there are no effective interventions to prevent the progression of autoimmune diseases. Hence, there is an urgent need for new treatment targets. TRPM7 is an enzyme-coupled, transient receptor ion channel of the subfamily M that plays a vital role in pathologic and physiologic conditions. While TRPM7 is constitutively activated under certain conditions, it can regulate cell migration, polarization, proliferation and cytokine secretion. However, a growing body of evidence highlights the critical role of TRPM7 in autoimmune diseases, including rheumatoid arthritis, multiple sclerosis and diabetes. Herein, we present (a) a review of the channel kinase properties of TRPM7 and its pharmacological properties, (b) discuss the role of TRPM7 in immune cells (neutrophils, macrophages, lymphocytes and mast cells) and its upstream immunoreactive substances, and (c) highlight TRPM7 as a potential therapeutic target for autoimmune diseases.
Collapse
Affiliation(s)
- Hong-Yu Liang
- The Second School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Xin Wei
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Gang-Gang Ma
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Jie Ding
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Chao Lu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Ren-Peng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
32
|
Rodat-Despoix L, Chamlali M, Ouadid-Ahidouch H. Ion channels as key partners of cytoskeleton in cancer disease. Biochim Biophys Acta Rev Cancer 2021; 1876:188627. [PMID: 34520803 DOI: 10.1016/j.bbcan.2021.188627] [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: 04/30/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022]
Abstract
Several processes occur during tumor development including changes in cell morphology, a reorganization of the expression and distribution of the cytoskeleton proteins as well as ion channels. If cytoskeleton proteins and ion channels have been widely investigated in understanding cancer mechanisms, the interaction between these two elements and the identification of the associated signaling pathways are only beginning to emerge. In this review, we summarize the work published over the past 15 years relating to the roles played by ion channels in these mechanisms of reorganization of the cellular morphology, essential to metastatic dissemination, both through the physical interactions with elements of the cytoskeleton and by intracellular signaling pathways involved.
Collapse
Affiliation(s)
- Lise Rodat-Despoix
- Laboratoire de Physiologie Cellulaire et Moléculaire (UR 4667), Université de Picardie Jules Verne, UFR des Sciences, 33 Rue St Leu, 80039 Amiens, France.
| | - Mohamed Chamlali
- Laboratoire de Physiologie Cellulaire et Moléculaire (UR 4667), Université de Picardie Jules Verne, UFR des Sciences, 33 Rue St Leu, 80039 Amiens, France
| | - Halima Ouadid-Ahidouch
- Laboratoire de Physiologie Cellulaire et Moléculaire (UR 4667), Université de Picardie Jules Verne, UFR des Sciences, 33 Rue St Leu, 80039 Amiens, France
| |
Collapse
|
33
|
Starostina I, Jang YK, Kim HS, Suh JS, Ahn SH, Choi GH, Suk M, Kim TJ. Distinct calcium regulation of TRPM7 mechanosensitive channels at plasma membrane microdomains visualized by FRET-based single cell imaging. Sci Rep 2021; 11:17893. [PMID: 34504177 PMCID: PMC8429465 DOI: 10.1038/s41598-021-97326-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/17/2021] [Indexed: 11/09/2022] Open
Abstract
Transient receptor potential subfamily M member 7 (TRPM7), a mechanosensitive Ca2+ channel, plays a crucial role in intracellular Ca2+ homeostasis. However, it is currently unclear how cell mechanical cues control TRPM7 activity and its associated Ca2+ influx at plasma membrane microdomains. Using two different types of Ca2+ biosensors (Lyn-D3cpv and Kras-D3cpv) based on fluorescence resonance energy transfer, we investigate how Ca2+ influx generated by the TRPM7-specific agonist naltriben is mediated at the detergent-resistant membrane (DRM) and non-DRM regions. This study reveals that TRPM7-induced Ca2+ influx mainly occurs at the DRM, and chemically induced mechanical perturbations in the cell mechanosensitive apparatus substantially reduce Ca2+ influx through TRPM7, preferably located at the DRM. Such perturbations include the disintegration of lipid rafts, microtubules, or actomyosin filaments; the alteration of actomyosin contractility; and the inhibition of focal adhesion and Src kinases. These results suggest that the mechanical membrane environment contributes to the TRPM7 function and activity. Thus, this study provides a fundamental understanding of how the mechanical aspects of the cell membrane regulate the function of mechanosensitive channels.
Collapse
Affiliation(s)
- Irina Starostina
- Department of Integrated Biological Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Yoon-Kwan Jang
- Department of Integrated Biological Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Heon-Su Kim
- Department of Integrated Biological Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Jung-Soo Suh
- Department of Integrated Biological Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Sang-Hyun Ahn
- Department of Integrated Biological Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Gyu-Ho Choi
- Department of Integrated Biological Science, Pusan National University, Pusan, 46241, Republic of Korea.,Department of Biological Sciences, Pusan National University, Pusan, 46241, Republic of Korea
| | - Myungeun Suk
- Department of Mechanical Engineering, Dong-Eui University, Pusan, 47340, Republic of Korea.
| | - Tae-Jin Kim
- Department of Integrated Biological Science, Pusan National University, Pusan, 46241, Republic of Korea. .,Department of Biological Sciences, Pusan National University, Pusan, 46241, Republic of Korea.
| |
Collapse
|
34
|
Rivier P, Mubalama M, Destaing O. Small GTPases all over invadosomes. Small GTPases 2021; 12:429-439. [PMID: 33487105 PMCID: PMC8583085 DOI: 10.1080/21541248.2021.1877081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/30/2020] [Accepted: 01/10/2021] [Indexed: 12/19/2022] Open
Abstract
Cell invasion is associated with numerous patho-physiologic states including cell development and metastatic dissemination. This process couples the activation of cell motility with the capacity to degrade the extracellular matrix, thereby permitting cells to pass through basal membranes. Invasion is sustained by the actions of invadosomes, an ensemble of subcellular structures with high functional homology. Invadosomes are 3D acto-adhesive structures that can also mediate local extracellular matrix degradation through the controlled delivery of proteases. Intracellular RHO GTPases play a central role in the regulation of invadosomes where their complex interplay regulates multiple invadosome functions. This review aims to provide an overview of the synergistic activities of the small GTPases in invadosome biology. This broad-based review also reinforces the importance of the spatiotemporal regulation of small GTPases and the impact of this process on invadosome dynamics.
Collapse
Affiliation(s)
- Paul Rivier
- Team DYSAD, Dept2, Institute for Advanced Biosciences, Centre de Recherche Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Grenoble, France
| | - Michel Mubalama
- Team DYSAD, Dept2, Institute for Advanced Biosciences, Centre de Recherche Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Grenoble, France
| | - Olivier Destaing
- Team DYSAD, Dept2, Institute for Advanced Biosciences, Centre de Recherche Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Grenoble, France
| |
Collapse
|
35
|
Farina AR, Cappabianca LA, Zelli V, Sebastiano M, Mackay AR. Mechanisms involved in selecting and maintaining neuroblastoma cancer stem cell populations, and perspectives for therapeutic targeting. World J Stem Cells 2021; 13:685-736. [PMID: 34367474 PMCID: PMC8316860 DOI: 10.4252/wjsc.v13.i7.685] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/09/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric neuroblastomas (NBs) are heterogeneous, aggressive, therapy-resistant embryonal tumours that originate from cells of neural crest (NC) origin and in particular neuroblasts committed to the sympathoadrenal progenitor cell lineage. Therapeutic resistance, post-therapeutic relapse and subsequent metastatic NB progression are driven primarily by cancer stem cell (CSC)-like subpopulations, which through their self-renewing capacity, intermittent and slow cell cycles, drug-resistant and reversibly adaptive plastic phenotypes, represent the most important obstacle to improving therapeutic outcomes in unfavourable NBs. In this review, dedicated to NB CSCs and the prospects for their therapeutic eradication, we initiate with brief descriptions of the unique transient vertebrate embryonic NC structure and salient molecular protagonists involved NC induction, specification, epithelial to mesenchymal transition and migratory behaviour, in order to familiarise the reader with the embryonic cellular and molecular origins and background to NB. We follow this by introducing NB and the potential NC-derived stem/progenitor cell origins of NBs, before providing a comprehensive review of the salient molecules, signalling pathways, mechanisms, tumour microenvironmental and therapeutic conditions involved in promoting, selecting and maintaining NB CSC subpopulations, and that underpin their therapy-resistant, self-renewing metastatic behaviour. Finally, we review potential therapeutic strategies and future prospects for targeting and eradication of these bastions of NB therapeutic resistance, post-therapeutic relapse and metastatic progression.
Collapse
Affiliation(s)
- Antonietta Rosella Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Lucia Annamaria Cappabianca
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Veronica Zelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Michela Sebastiano
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Andrew Reay Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy.
| |
Collapse
|
36
|
Hiraishi K, Kurahara LH, Feng J, Yamamura A, Cui Y, Yahiro E, Yokomise H, Go T, Ishikawa K, Yokota N, Fujiwara A, Onitsuka M, Abe K, Ohga S, Satoh T, Okada Y, Yue L, Inoue R, Hirano K. Substantial involvement of TRPM7 inhibition in the therapeutic effect of Ophiocordyceps sinensis on pulmonary hypertension. Transl Res 2021; 233:127-143. [PMID: 33691194 PMCID: PMC9225677 DOI: 10.1016/j.trsl.2021.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 01/08/2023]
Abstract
Ophiocordyceps sinensis (OCS), an entomopathogenic fungus, is known to exert antiproliferative and antitissue remodeling effects. Vascular remodeling and vasoconstriction play critical roles in the development of pulmonary hypertension (PH). The therapeutic potential of OCS for PH was investigated using rodent PH models, and cultured pulmonary artery endothelial and smooth muscle cells (PAECs and PASMCs), with a focus on the involvement of TRPM7. OCS ameliorated the development of PH, right ventricular hypertrophy and dysfunction in the monocrotaline-induced PH rats. The genetic knockout of TRPM7 attenuated the development of PH in mice with monocrotaline pyrrole-induced PH. TRPM7 was associated with medial hypertrophy and the plexiform lesions in rats and humans with PH. OCS suppressed proliferation of PASMCs derived from the PH patients. Ethanol extracts of OCS inhibited TRPM7-like current, TGF-β2-induced endothelial-mesenchymal transition, IL-6-induced STAT3 phosphorylation, and PDGF-induced Akt phosphorylation in PAECs or PASMCs. These inhibitory effects were recapitulated by either siRNA-mediated TRPM7 knockdown or treatment with TRPM7 antagonist FTY-720. OCS and FTY-720 induced vasorelaxation in the isolated normal human pulmonary artery. As a result, the present study proposes the therapeutic potential of OCS for the treatment of PH. The inhibition of TRPM7 is suggested to underlie the therapeutic effect of OCS.
Collapse
Affiliation(s)
- Keizo Hiraishi
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa, Japan; Department of Physiology, Fukuoka University School of Medicine, Johnan-ku, Fukuoka, Japan
| | - Lin Hai Kurahara
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa, Japan; Department of Physiology, Fukuoka University School of Medicine, Johnan-ku, Fukuoka, Japan.
| | - Jianlin Feng
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Aya Yamamura
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yuanyuan Cui
- Department of Physiology, Fukuoka University School of Medicine, Johnan-ku, Fukuoka, Japan; School of Basic Medical Sciences & Shanxi key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, China
| | - Eiji Yahiro
- Fukuoka University Medical Education Center, Fukuoka University School of Medicine, Fukuoka University, Johnan-ku, Fukuoka, Japan
| | - Hiroyasu Yokomise
- Department of General Thoracic Surgery, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan
| | - Tetsuhiko Go
- Department of General Thoracic Surgery, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan
| | - Kaori Ishikawa
- Department of General Medicine, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan
| | - Naoya Yokota
- Department of General Thoracic Surgery, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan
| | - Atsushi Fujiwara
- Department of General Thoracic Surgery, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan
| | - Miki Onitsuka
- Department of Pathology, School of Medicine, Fukuoka University, Fukuoka, Japan
| | - Kohtaro Abe
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Higashi-ku, Fukuoka, Japan
| | - Shoji Ohga
- Faculty of Agriculture, Kyusyu University Professor Emeritus, Kasuya-gun, Fukuoka, Japan
| | - Toru Satoh
- Division of Cardiology, Department of Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Yasumasa Okada
- Division of Internal Medicine and Laboratory of Electrophysiology, Murayama Medical Center, Tokyo, Japan
| | - Lixia Yue
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Ryuji Inoue
- Department of Physiology, Fukuoka University School of Medicine, Johnan-ku, Fukuoka, Japan.
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa, Japan
| |
Collapse
|
37
|
Saldías MP, Maureira D, Orellana-Serradell O, Silva I, Lavanderos B, Cruz P, Torres C, Cáceres M, Cerda O. TRP Channels Interactome as a Novel Therapeutic Target in Breast Cancer. Front Oncol 2021; 11:621614. [PMID: 34178620 PMCID: PMC8222984 DOI: 10.3389/fonc.2021.621614] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/31/2021] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is one of the most frequent cancer types worldwide and the first cause of cancer-related deaths in women. Although significant therapeutic advances have been achieved with drugs such as tamoxifen and trastuzumab, breast cancer still caused 627,000 deaths in 2018. Since cancer is a multifactorial disease, it has become necessary to develop new molecular therapies that can target several relevant cellular processes at once. Ion channels are versatile regulators of several physiological- and pathophysiological-related mechanisms, including cancer-relevant processes such as tumor progression, apoptosis inhibition, proliferation, migration, invasion, and chemoresistance. Ion channels are the main regulators of cellular functions, conducting ions selectively through a pore-forming structure located in the plasma membrane, protein–protein interactions one of their main regulatory mechanisms. Among the different ion channel families, the Transient Receptor Potential (TRP) family stands out in the context of breast cancer since several members have been proposed as prognostic markers in this pathology. However, only a few approaches exist to block their specific activity during tumoral progress. In this article, we describe several TRP channels that have been involved in breast cancer progress with a particular focus on their binding partners that have also been described as drivers of breast cancer progression. Here, we propose disrupting these interactions as attractive and potential new therapeutic targets for treating this neoplastic disease.
Collapse
Affiliation(s)
- María Paz Saldías
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile
| | - Diego Maureira
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile
| | - Octavio Orellana-Serradell
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile
| | - Ian Silva
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile
| | - Boris Lavanderos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile
| | - Pablo Cruz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile
| | - Camila Torres
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile
| | - Mónica Cáceres
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment, and Health (WoRTH) Initiative, Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment, and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
38
|
Meng S, Alanazi R, Ji D, Bandura J, Luo ZW, Fleig A, Feng ZP, Sun HS. Role of TRPM7 kinase in cancer. Cell Calcium 2021; 96:102400. [PMID: 33784560 DOI: 10.1016/j.ceca.2021.102400] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 01/09/2023]
Abstract
Cancer is the second leading cause of death worldwide and accounted for an estimated 9.6 million deaths, or 1 in 6 deaths, in 2018. Despite recent advances in cancer prevention, diagnosis, and treatment strategies, the burden of this disease continues to grow with each year, with dire physical, emotional, and economic consequences for all levels of society. Classic characteristics of cancer include rapid, uncontrolled cell proliferation and spread of cancerous cells to other parts of the body, a process known as metastasis. Transient receptor potential melastatin 7 (TRPM7), a Ca2+- and Mg2+-permeable nonselective divalent cation channel defined by the atypical presence of an α-kinase within its C-terminal domain, has been implicated, due to its modulation of Ca2+ and Mg2+ influx, in a wide variety of physiological and pathological processes, including cancer. TRPM7 is overexpressed in several cancer types and has been shown to variably increase cellular proliferation, migration, and invasion of tumour cells. However, the relative contribution of TRPM7 kinase domain activity to cancer as opposed to ion flux through its channel pore remains an area of active discovery. In this review, we describe the specific role of the TRPM7 kinase domain in cancer processes as well as mechanisms of regulation and inhibition of the kinase domain.
Collapse
Affiliation(s)
- Selena Meng
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Rahmah Alanazi
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Delphine Ji
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Julia Bandura
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Zheng-Wei Luo
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine and Cancer Center at the University of Hawaii, Honolulu, HI, 96720, USA
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada.
| | - Hong-Shuo Sun
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Department of Pharmacology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada.
| |
Collapse
|
39
|
Abstract
Mechanosensing is a key feature through which organisms can receive inputs from the environment and convert them into specific functional and behavioral outputs. Mechanosensation occurs in many cells and tissues, regulating a plethora of molecular processes based on the distribution of forces and stresses both at the cell membrane and at the intracellular organelles levels, through complex interactions between cells’ microstructures, cytoskeleton, and extracellular matrix. Although several primary and secondary mechanisms have been shown to contribute to mechanosensation, a fundamental pathway in simple organisms and mammals involves the presence of specialized sensory neurons and the presence of different types of mechanosensitive ion channels on the neuronal cell membrane. In this contribution, we present a review of the main ion channels which have been proven to be significantly involved in mechanotransduction in neurons. Further, we discuss recent studies focused on the biological mechanisms and modeling of mechanosensitive ion channels’ gating, and on mechanotransduction modeling at different scales and levels of details.
Collapse
|
40
|
Cojocaru F, Şelescu T, Domocoş D, Măruţescu L, Chiritoiu G, Chelaru NR, Dima S, Mihăilescu D, Babes A, Cucu D. Functional expression of the transient receptor potential ankyrin type 1 channel in pancreatic adenocarcinoma cells. Sci Rep 2021; 11:2018. [PMID: 33479347 PMCID: PMC7819973 DOI: 10.1038/s41598-021-81250-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 01/03/2021] [Indexed: 11/09/2022] Open
Abstract
The transient receptor potential ankyrin type 1 (TRPA1) channel belongs to the TRP superfamily of ion channels. TRPA1 is a membrane protein with multiple functions able to respond to noxious stimuli, reactive oxygen species, inflammatory cytokines or pungent substances, and it participates in pain signalling, taste, inflammation and various steps of the tumorigenic process. To date, no reports have addressed the expression and function of TRPA1 in pancreatic ductal adenocarcinoma (PDAC) cells. This work reports the endogenous expression of TRPA1 channels in human pancreatic adenocarcinoma cell lines and provides insights into the function of the TRPA1 protein in the Panc-1 cell line. This study reports that cell lines isolated from PDAC patients had different levels of TRPA1 expression. The channel activity in Panc-1 cells, as assessed with electrophysiological (whole-cell patch clamp) and microfluorimetry methods, showed that non-selective cationic currents were activated by allyl isothiocyanate (AITC) in Panc-1 cells and inhibited by the selective TRPA1 antagonist A-967079. The current elicited by the specific agonist was associated with a robust increase in intracellular Ca2+. Furthermore, siRNA-induced downregulation of TRPA1 enhanced cell migration in the wound healing assay, indicating a possible role of ion channels independent from pore function. Finally, TRPA1 activation changed the cell cycle progression. Taken together, these results support the idea of channel-dependent and independent role for TRPA1 in tumoral processes.
Collapse
Affiliation(s)
- Florentina Cojocaru
- Department DAFAB, Faculty of Biology, University of Bucharest, Splaiul Independenței 91-95, Bucharest, Romania
| | - Tudor Şelescu
- Department DAFAB, Faculty of Biology, University of Bucharest, Splaiul Independenței 91-95, Bucharest, Romania
| | - Dan Domocoş
- Department DAFAB, Faculty of Biology, University of Bucharest, Splaiul Independenței 91-95, Bucharest, Romania
| | - Luminiţa Măruţescu
- Faculty of Biology, Research Institute of the University of Bucharest (ICUB), University of Bucharest, Bucharest, Romania
| | - Gabriela Chiritoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, Splaiul Independenței 296, 060031, Bucharest, Romania
| | - Nicoleta-Raluca Chelaru
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328, Bucharest, Romania
| | - Simona Dima
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328, Bucharest, Romania
| | - Dan Mihăilescu
- Department DAFAB, Faculty of Biology, University of Bucharest, Splaiul Independenței 91-95, Bucharest, Romania
| | - Alexandru Babes
- Department DAFAB, Faculty of Biology, University of Bucharest, Splaiul Independenței 91-95, Bucharest, Romania.
| | - Dana Cucu
- Department DAFAB, Faculty of Biology, University of Bucharest, Splaiul Independenței 91-95, Bucharest, Romania.
| |
Collapse
|
41
|
Acheva A, Kärki T, Schaible N, Krishnan R, Tojkander S. Adipokine Leptin Co-operates With Mechanosensitive Ca 2 +-Channels and Triggers Actomyosin-Mediated Motility of Breast Epithelial Cells. Front Cell Dev Biol 2021; 8:607038. [PMID: 33490070 PMCID: PMC7815691 DOI: 10.3389/fcell.2020.607038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
In postmenopausal women, a major risk factor for the development of breast cancer is obesity. In particular, the adipose tissue-derived adipokine leptin has been strongly linked to tumor cell proliferation, migration, and metastasis, but the underlying mechanisms remain unclear. Here we show that treatment of normal mammary epithelial cells with leptin induces EMT-like features characterized by higher cellular migration speeds, loss of structural ordering of 3D-mammo spheres, and enhancement of epithelial traction forces. Mechanistically, leptin triggers the phosphorylation of myosin light chain kinase-2 (MLC-2) through the interdependent activity of leptin receptor and Ca2+ channels. These data provide evidence that leptin-activated leptin receptors, in co-operation with mechanosensitive Ca2+ channels, play a role in the development of breast carcinomas through the regulation of actomyosin dynamics.
Collapse
Affiliation(s)
- Anna Acheva
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Tytti Kärki
- Department of Applied Physics, School of Science, Aalto University, Espoo, Finland
| | - Niccole Schaible
- Beth Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ramaswamy Krishnan
- Beth Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Sari Tojkander
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
42
|
Vellino S, Oddou C, Rivier P, Boyault C, Hiriart-Bryant E, Kraut A, Martin R, Coute Y, Knölker HJ, Valverde MA, Albigès-Rizo C, Destaing O. Cross-talk between the calcium channel TRPV4 and reactive oxygen species interlocks adhesive and degradative functions of invadosomes. J Cell Biol 2021; 220:211651. [PMID: 33399853 PMCID: PMC7788461 DOI: 10.1083/jcb.201910079] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 07/23/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
Invadosomes support cell invasion by coupling both acto-adhesive and extracellular matrix degradative functions, which are apparently antagonistic. β1-integrin dynamics regulate this coupling, but the actual sensing mechanism and effectors involved have not yet been elucidated. Using genetic and reverse genetic approaches combined with biochemical and imaging techniques, we now show that the calcium channel TRPV4 colocalizes with β1-integrins at the invadosome periphery and regulates its activation and the coupling of acto-adhesive and degradative functions. TRPV4-mediated regulation of podosome function depends on its ability to sense reactive oxygen species (ROS) in invadosomes' microenvironment and involves activation of the ROS/calcium-sensitive kinase Ask1 and binding of the motor MYO1C. Furthermore, disease-associated TRPV4 gain-of-function mutations that modulate ECM degradation are also implicated in the ROS response, which provides new perspectives in our understanding of the pathophysiology of TRPV4 channelopathies.
Collapse
Affiliation(s)
- Sanela Vellino
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Christiane Oddou
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Paul Rivier
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Cyril Boyault
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Edwige Hiriart-Bryant
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Alexandra Kraut
- Laboratoire EDyP, Institute of Biosciences and Biotechnologies of Grenoble-Biologie à Grande Echelle, Commissariat à l'Énergie Atomique Grenoble, Grenoble, France
| | - René Martin
- Faculty of Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Yohann Coute
- Laboratoire EDyP, Institute of Biosciences and Biotechnologies of Grenoble-Biologie à Grande Echelle, Commissariat à l'Énergie Atomique Grenoble, Grenoble, France
| | | | - Miguel A. Valverde
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Corinne Albigès-Rizo
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Olivier Destaing
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France,Correspondence to Olivier Destaing:
| |
Collapse
|
43
|
Hu F, Li M, Han F, Zhang Q, Zeng Y, Zhang W, Cheng X. Role of TRPM7 in cardiac fibrosis: A potential therapeutic target (Review). Exp Ther Med 2020; 21:173. [PMID: 33456540 PMCID: PMC7792474 DOI: 10.3892/etm.2020.9604] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiac fibrosis is a hallmark of cardiac remodeling associated with nearly all forms of heart disease. Clinically, no effective therapeutic drugs aim to inhibit cardiac fibrosis, owing to the complex etiological heterogeneity and pathogenesis of this disease. A two-in-one protein structure, a ubiquitous expression profile and unique biophysical characteristics enable the involvement of transient receptor potential melastatin-subfamily member 7 (TRPM7) in the pathogenesis and development of fibrosis-related cardiac diseases, such as heart failure (HF), cardiomyopathies, arrhythmia and hyperaldosteronism. In response to a variety of stimuli, multiple bioactive molecules can activate TRPM7 and related signaling pathways, leading to fibroblast proliferation, differentiation and extracellular matrix production in cardiac fibroblasts. TRPM7-mediated Ca2+ signaling and TGF-β1 signaling pathways are critical for the formation of fibrosis. Accumulating evidence has demonstrated that TRPM7 is a potential pharmacological target for halting the development of fibrotic cardiac diseases. Reliable drug-like molecules for further development of high-affinity in vivo drugs targeting TRPM7 are urgently needed. The present review discusses the widespread and significant role of TRPM7 in cardiac fibrosis and focuses on its potential as a therapeutic target for alleviating heart fibrogenesis.
Collapse
Affiliation(s)
- Feng Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Meiyong Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fengyu Han
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qing Zhang
- Department of Cardiology, The Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yuhao Zeng
- Department of Medical Education, The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Weifang Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaoshu Cheng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China.,Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
Kärki T, Rajakylä EK, Acheva A, Tojkander S. TRPV6 calcium channel directs homeostasis of the mammary epithelial sheets and controls epithelial mesenchymal transition. Sci Rep 2020; 10:14683. [PMID: 32895467 PMCID: PMC7477193 DOI: 10.1038/s41598-020-71645-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022] Open
Abstract
Epithelial integrity is lost upon cancer progression as cancer cells detach from the primary tumor site and start to invade to the surrounding tissues. Invasive cancers of epithelial origin often express altered levels of TRP-family cation channels. Upregulation of TRPV6 Ca2+-channel has been associated with a number of human malignancies and its high expression in breast cancer has been linked to both proliferation and invasive disease. The mechanisms behind the potential of TRPV6 to induce invasive progression have, however, not been well elucidated. Here we show that TRPV6 is connected to both E-cadherin-based adherens junctions and intracellular cytoskeletal structures. Loss of TRPV6 from normal mammary epithelial cells led to disruption of epithelial integrity and abnormal 3D-mammo sphere morphology. Furthermore, expression level of TRPV6 was tightly linked to the levels of common EMT markers, suggesting that TRPV6 may have a role in the mesenchymal invasion of breast cancer cells. Thus, either too low or too high TRPV6 levels compromise homeostasis of the mammary epithelial sheets and may promote the progression of pathophysiological conditions.
Collapse
Affiliation(s)
- Tytti Kärki
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, 00014, Helsinki, Finland
- Department of Applied Physics, Aalto University School of Science, Puumiehenkuja 2, 02150, Espoo, Finland
| | - Eeva Kaisa Rajakylä
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, 00014, Helsinki, Finland
| | - Anna Acheva
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, 00014, Helsinki, Finland
| | - Sari Tojkander
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, 00014, Helsinki, Finland.
| |
Collapse
|
47
|
Inoue H, Inazu M, Konishi M, Yokoyama U. Functional expression of TRPM7 as a Ca 2+ influx pathway in adipocytes. Physiol Rep 2020; 7:e14272. [PMID: 31650715 PMCID: PMC6813326 DOI: 10.14814/phy2.14272] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 09/20/2019] [Accepted: 10/04/2019] [Indexed: 02/01/2023] Open
Abstract
In adipocytes, intracellular Ca2+ and Mg2+ modulates physiological functions, such as insulin action and the secretion of adipokines. TRPM7 is a Ca2+/Mg2+‐permeable non‐selective cation channel. TRPM7 mRNA is highly expressed in adipose tissue, however, its functional expression in adipocytes remains to be elucidated. In this study, we demonstrated for the first time that TRPM7 was functionally expressed in both freshly isolated white adipocytes and in 3T3‐L1 adipocytes differentiated from a 3T3‐L1 pre‐adipocyte cell line by whole‐cell patch‐clamp recordings. Consistent with known properties of TRPM7 current, the current in adipocytes was activated by the elimination of extracellular divalent cations and the reduction of intracellular free Mg2+ concentrations, and was inhibited by the TRPM7 inhibitors, 2‐aminoethyl diphenylborinate (2‐APB), hydrogen peroxide (H2O2), N‐methyl maleimide (NMM), NS8593, and 2‐amino‐2‐[2‐(4‐octylphenyl)ethyl]‐1,3‐propanediol (FTY720). Treatment with small‐interfering (si) RNA targeting TRPM7 resulted in a reduction in the current to 23 ± 7% of nontargeting siRNA‐treated adipocytes. Moreover a TRPM7 activator, naltriben, increased the TRPM7‐like current and [Ca2+]i in 3T3‐L1 adipocytes but not in TRPM7‐knockdown adipocytes. These findings indicate that TRPM7 is functionally expressed, and plays a role as a Ca2+ influx pathway in adipocytes.
Collapse
Affiliation(s)
- Hana Inoue
- Department of Physiology, Tokyo Medical University, Tokyo, Japan
| | - Masato Inazu
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Masato Konishi
- Department of Physiology, Tokyo Medical University, Tokyo, Japan
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
48
|
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: 35] [Impact Index Per Article: 7.0] [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
|
49
|
Hu Q, Wolfner MF. Regulation of Trpm activation and calcium wave initiation during Drosophila egg activation. Mol Reprod Dev 2020; 87:880-886. [PMID: 32735035 DOI: 10.1002/mrd.23403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/24/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022]
Abstract
The transition from a developmentally arrested mature oocyte to a developing embryo requires a series of highly conserved events, collectively known as egg activation. All of these events are preceded by a ubiquitous rise of intracellular calcium, which results from influx of external calcium and/or calcium release from internal storage. In Drosophila, this calcium rise initiates from the pole(s) of the oocyte by influx of external calcium in response to mechanical triggers. It is thought to trigger calcium responsive kinases and/or phosphatases, which in turn alter the oocyte phospho-proteome to initiate downstream events. Recent studies revealed that external calcium enters the activating Drosophila oocyte through Trpm channels, a feature conserved in mouse. The local entry of calcium raises the question of whether Trpm channels are found locally at the poles of the oocyte or are localized around the oocyte periphery, but activated only at the poles. Here, we show that Trpm is distributed all around the oocyte. This requires that it thus be specially regulated at the poles to allow calcium wave initiation. We show that neither egg shape nor local pressure is sufficient to explain this local activation of Trpm channels.
Collapse
Affiliation(s)
- Qinan Hu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York
| | - Mariana F Wolfner
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York
| |
Collapse
|
50
|
Svandova E, Peterkova R, Matalova E, Lesot H. Formation and Developmental Specification of the Odontogenic and Osteogenic Mesenchymes. Front Cell Dev Biol 2020; 8:640. [PMID: 32850793 PMCID: PMC7396701 DOI: 10.3389/fcell.2020.00640] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022] Open
Abstract
Within the mandible, the odontogenic and osteogenic mesenchymes develop in a close proximity and form at about the same time. They both originate from the cranial neural crest. These two condensing ecto-mesenchymes are soon separated from each other by a very loose interstitial mesenchyme, whose cells do not express markers suggesting a neural crest origin. The two condensations give rise to mineralized tissues while the loose interstitial mesenchyme, remains as a soft tissue. This is crucial for proper anchorage of mammalian teeth. The situation in all three regions of the mesenchyme was compared with regard to cell heterogeneity. As the development progresses, the early phenotypic differences and the complexity in cell heterogeneity increases. The differences reported here and their evolution during development progressively specifies each of the three compartments. The aim of this review was to discuss the mechanisms underlying condensation in both the odontogenic and osteogenic compartments as well as the progressive differentiation of all three mesenchymes during development. Very early, they show physical and structural differences including cell density, shape and organization as well as the secretion of three distinct matrices, two of which will mineralize. Based on these data, this review highlights the consecutive differences in cell-cell and cell-matrix interactions, which support the cohesion as well as mechanosensing and mechanotransduction. These are involved in the conversion of mechanical energy into biochemical signals, cytoskeletal rearrangements cell differentiation, or collective cell behavior.
Collapse
Affiliation(s)
- Eva Svandova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, Brno, Czechia
| | - Renata Peterkova
- Department of Histology and Embryology, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Eva Matalova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, Brno, Czechia.,Department of Physiology, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
| | - Herve Lesot
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, Brno, Czechia
| |
Collapse
|