1
|
Tian Y, Lin Y, Ma Y, Li J, Sahu SK, Fan J, Lin C, Li Z, Shi M, He F, Bai L, Fu Y, Deng Z, Guo H, Li H, Li Q, Xu Y, Lan T, Hou Z, Xia Y, Yang S. Population Genomics Reveals Elevated Inbreeding and Accumulation of Deleterious Mutations in White Raccoon Dogs. BIOLOGY 2025; 14:30. [PMID: 39857261 PMCID: PMC11760849 DOI: 10.3390/biology14010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025]
Abstract
The formation of animal breeds usually begins with a small subsample from their ancestral population. Deleterious mutations accumulate in the population under genetic drift, inbreeding, and artificial selection during the development and maintenance of traits desired by humans. White raccoon dogs are among the most popular breeds of farmed raccoon dogs, but white raccoon dogs are more susceptible to disease and have a lower reproductive ability. However, the accumulation of deleterious mutations in this white breed is largely unknown. By analyzing and comparing whole-genome sequencing data from 20 white raccoon dogs and 38 normal raccoon dogs, we detected an increased occurrence of loss-of-function (LoF) mutations in white raccoon dogs compared with normal raccoon dogs. With the finding of a significantly higher dosage of homozygous missense mutations in the white raccoon dog genome, we detected a greater fitness cost in white raccoon dogs. Although a much higher FROH level for ROH fragments longer than 1 Mb has been reported in white raccoon dogs, we did not detect a genetic signal of genetic purging in white raccoon dogs. This study provides valuable genomic resources and new insights into the accumulation of mutation loads in farmed raccoon dogs.
Collapse
Affiliation(s)
- Yinping Tian
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| | - Yu Lin
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| | - Yue Ma
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| | - Jiayi Li
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| | - Sunil Kumar Sahu
- BGI Research, Wuhan 430074, China;
- State Key Laboratory of Agricultural Genomics, BGI-Shenzhen, Shenzhen 518083, China;
| | - Jiale Fan
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| | - Chen Lin
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| | - Zhiang Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (Z.L.); (Q.L.)
| | - Minhui Shi
- State Key Laboratory of Agricultural Genomics, BGI-Shenzhen, Shenzhen 518083, China;
| | - Fengping He
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China;
| | - Lianduo Bai
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| | - Yuan Fu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| | - Zhangwen Deng
- Guangxi Zhuang Autonomous Region Forest Inventory and Planning Institute, Nanning 530011, China;
| | - Huabing Guo
- Forest Inventory and Planning Institute of Jilin Province, Changchun 130022, China;
| | - Haimeng Li
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
- Heilongjiang Key Laboratory of Complex Traits and Protein Machines in Organisms, Harbin 150040, China
| | - Qiye Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (Z.L.); (Q.L.)
| | - Yanchun Xu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
- BGI Life Science Joint Research Center, Northeast Forestry University, Harbin 150040, China
| | - Tianming Lan
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
- Heilongjiang Key Laboratory of Complex Traits and Protein Machines in Organisms, Harbin 150040, China
- BGI Life Science Joint Research Center, Northeast Forestry University, Harbin 150040, China
| | - Zhijun Hou
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| | - Yanling Xia
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| | - Shuhui Yang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China; (Y.T.); (Y.L.); (Y.M.); (J.L.); (J.F.); (C.L.); (L.B.); (Y.F.); (H.L.); (Y.X.); (T.L.); (Z.H.)
| |
Collapse
|
2
|
Chen Z, Xie H, Liu J, Zhao J, Huang R, Xiang Y, Wu H, Tian D, Bian E, Xiong Z. Roles of TRPM channels in glioma. Cancer Biol Ther 2024; 25:2338955. [PMID: 38680092 PMCID: PMC11062369 DOI: 10.1080/15384047.2024.2338955] [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: 12/03/2023] [Accepted: 04/01/2024] [Indexed: 05/01/2024] Open
Abstract
Gliomas are the most common type of primary brain tumor. Despite advances in treatment, it remains one of the most aggressive and deadly tumor of the central nervous system (CNS). Gliomas are characterized by high malignancy, heterogeneity, invasiveness, and high resistance to radiotherapy and chemotherapy. It is urgent to find potential new molecular targets for glioma. The TRPM channels consist of TRPM1-TPRM8 and play a role in many cellular functions, including proliferation, migration, invasion, angiogenesis, etc. More and more studies have shown that TRPM channels can be used as new therapeutic targets for glioma. In this review, we first introduce the structure, activation patterns, and physiological functions of TRPM channels. Additionally, the pathological mechanism of glioma mediated by TRPM2, 3, 7, and 8 and the related signaling pathways are described. Finally, we discuss the therapeutic potential of targeting TRPM for glioma.
Collapse
Affiliation(s)
- Zhigang Chen
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders, The First Affiliated Hospital (Yijishan Hospital), Wannan Medical College, Wuhu, P. R. China
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Han Xie
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - JiaJia Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Ruixiang Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Yufei Xiang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Haoyuan Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Dasheng Tian
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Erbao Bian
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhang Xiong
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders, The First Affiliated Hospital (Yijishan Hospital), Wannan Medical College, Wuhu, P. R. China
| |
Collapse
|
3
|
Schlieben LD, Achleitner MT, Bourke B, Diesner M, Feichtinger RG, Fichtner A, Flechtenmacher C, Hadzic N, Hegarty R, Heilos A, Janecke A, Konstantopoulou V, Lenz D, Mayr JA, Müller T, Prokisch H, Vogel GF. Missense variants in the TRPM7 α-kinase domain are associated with recurrent pediatric acute liver failure. Hepatol Commun 2024; 8:e0598. [PMID: 39621058 PMCID: PMC11608757 DOI: 10.1097/hc9.0000000000000598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/20/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Pediatric acute liver failure (PALF) is a rare and life-threatening condition. In up to 50% of PALF cases, the underlying etiology remains unknown during routine clinical testing. This lack of knowledge complicates clinical management and liver transplantation decisions. Recently, whole-exome sequencing has identified genetic disorders in a large number of cases without specific laboratory biomarkers or metabolic fingerprints. METHODS We describe how further analysis of whole-exome sequencing data combined with proteomic analyses in 5 previously unsolved PALF patients, where no pathogenic variants in genes previously associated with acute liver failure were identified, revealed rare biallelic variants in transient receptor potential cation channel subfamily M member 7 (TRPM7). RESULTS We establishe TRPM7 as a novel disease gene for PALF. Yet, the cation channel kinase TRPM7 has not been associated with any Mendelian disorder. No homozygous loss-of-function variants were found in in-house exomes or publicly available databases. Rare biallelic TRPM7-variants were significantly enriched in the PALF cohort compared with a pediatric control cohort. Viral infections preceded the majority of PALF episodes. Recurrent PALF episodes characterized the disease course with rapid progression, leading to early death in 3 cases. Proteomic analyses of patient fibroblasts unveiled significantly reduced TRPM7 protein levels, indicative of functional impairment. Severely reduced Mg2+ levels in one individual with a mutation in the channel domain suggests a potential interaction between disturbed Mg2+ homeostasis and PALF. The consistent presence of mutations in the TRPM7 protein-kinase-domain across all patients suggests its specific relevance in PALF. CONCLUSIONS Our data extend the genetic spectrum of recurrent PALF and prompt consideration of TRPM7 in children with unexplained liver failure.
Collapse
Affiliation(s)
- Lea D. Schlieben
- School of Medicine, Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Melanie T. Achleitner
- University Children’s Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU) Salzburg, Salzburg, Austria
| | - Billy Bourke
- UCD School of Medicine & Medical Science, Crumlin, Dublin, Ireland
| | | | - René G. Feichtinger
- University Children’s Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU) Salzburg, Salzburg, Austria
| | - Alexander Fichtner
- Department I, Division of Pediatric Neurology and Metabolic Medicine Heidelberg University, Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Heidelberg, Germany
| | | | - Nedim Hadzic
- King’s College Hospital, Paediatric Liver, GI & Nutrition Centre, London, United Kingdom
| | - Robert Hegarty
- King’s College Hospital, Paediatric Liver, GI & Nutrition Centre, London, United Kingdom
| | - Andreas Heilos
- Department of Paediatric Gastroenterology, Medical University of Vienna, Vienna, Austria
- Department of Paediatrics, Medical University of Vienna, Vienna, Austria
| | - Andreas Janecke
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Dominic Lenz
- Department I, Division of Pediatric Neurology and Metabolic Medicine Heidelberg University, Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Heidelberg, Germany
| | - Johannes A. Mayr
- University Children’s Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU) Salzburg, Salzburg, Austria
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Holger Prokisch
- School of Medicine, Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Georg F. Vogel
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
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
|
5
|
Xie Z, Abumaria N. Effect of truncation on TRPM7 channel activity. Channels (Austin) 2023; 17:2200874. [PMID: 37040321 PMCID: PMC10761173 DOI: 10.1080/19336950.2023.2200874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 04/04/2023] [Indexed: 04/12/2023] Open
Abstract
Transient receptor potential melastatin-like 7 (TRPM7) is a key player in various physiological and pathological processes. TRPM7 channel activity is regulated by different factors. The effects of cleavage of different domains on channel activity remain unknown. Here, we constructed several TRPM7 clones and explored the effects of truncating the mouse TRPM7 at different locations on the ion channel activity in two cell lines. We compared the clones' activity with the full-length TRPM7 and the native TRPM7 in transfected and untransfected cells. We also expressed fluorescently tagged truncated clones to examine their protein stability and membrane targeting. We found that truncating the kinase domain induced reduction in TRPM7 channel activity. Further truncations beyond the kinase (serine/threonine rich domain and/or coiled-coil domain) did not result in further reductions in channel activity. Two truncated clones lacking the TRP domain or the melastatin homology domain had a completely nonfunctional channel apparently due to disruption of protein stability. We identified the shortest structure of TRPM7 with measurable channel activity. We found that the truncated TRPM7 containing only S5 and S6 domains retained some channel activity. Adding the TRP domain to the S5-S6 resulted in a significant increase in channel activity. Finally, our analysis showed that TRPM7 outward currents are more sensitive to truncations than inward currents. Our data provide insights on the effects of truncating TRPM7 at different locations on the channel functions, highlighting the importance of different domains in impacting channel activity, protein stability, and/or membrane targeting.
Collapse
Affiliation(s)
- Zhuqing Xie
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Nashat Abumaria
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Deng F, Fu M, Zhao C, Lei J, Xu T, Ji B, Ding H, Zhang Y, Chen J, Qiu J, Gao Q. Calcium signals and potential therapy targets in ovarian cancer (Review). Int J Oncol 2023; 63:125. [PMID: 37711071 PMCID: PMC10552713 DOI: 10.3892/ijo.2023.5573] [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: 03/23/2023] [Accepted: 08/22/2023] [Indexed: 09/16/2023] Open
Abstract
Ovarian cancer (OC) is a deadly disease. The poor prognosis and high lethality of OC are attributed to its high degrees of aggressiveness, resistance to chemotherapy and recurrence rates. Calcium ion (Ca2+) signaling has received attention in recent years, as it appears to form an essential part of various aspects of cancer pathophysiology and is a potential therapeutic target for OC treatment. Disruption of normal Ca2+ signaling pathways can induce changes in cell cycle progression, apoptosis, proliferation and migration and invasion, leading to the development of the malignant phenotype of tumors. In the present review, the main roles of ion channel/receptor/pump‑triggered Ca2+ signaling pathways located at the plasma membrane and organelle Ca2+ transport in OC are summarized. In addition, the potential of Ca2+ signaling as a novel target for the development of effective treatment strategies for OC was discussed. Furthering the understanding into the role of Ca2+ signaling in OC is expected to facilitated the identification of novel therapeutic targets and improved clinical outcomes for patients.
Collapse
Affiliation(s)
- Fengying Deng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Mengyu Fu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Chenxuan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jiahui Lei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Ting Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Bingyu Ji
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongmei Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yueming Zhang
- Department of Gynecology and Obstetrics, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215100, P.R. China
| | - Jie Chen
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Junlan Qiu
- Department of Oncology and Hematology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu 215153, P.R. China
| | - Qinqin Gao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
7
|
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
|
8
|
Otero-Sobrino Á, Blanco-Carlón P, Navarro-Aguadero MÁ, Gallardo M, Martínez-López J, Velasco-Estévez M. Mechanosensitive Ion Channels: Their Physiological Importance and Potential Key Role in Cancer. Int J Mol Sci 2023; 24:13710. [PMID: 37762011 PMCID: PMC10530364 DOI: 10.3390/ijms241813710] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Mechanosensitive ion channels comprise a broad group of proteins that sense mechanical extracellular and intracellular changes, translating them into cation influx to adapt and respond to these physical cues. All cells in the organism are mechanosensitive, and these physical cues have proven to have an important role in regulating proliferation, cell fate and differentiation, migration and cellular stress, among other processes. Indeed, the mechanical properties of the extracellular matrix in cancer change drastically due to high cell proliferation and modification of extracellular protein secretion, suggesting an important contribution to tumor cell regulation. In this review, we describe the physiological significance of mechanosensitive ion channels, emphasizing their role in cancer and immunity, and providing compelling proof of the importance of continuing to explore their potential as new therapeutic targets in cancer research.
Collapse
Affiliation(s)
- Álvaro Otero-Sobrino
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Pablo Blanco-Carlón
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Miguel Ángel Navarro-Aguadero
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Miguel Gallardo
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Joaquín Martínez-López
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - María Velasco-Estévez
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| |
Collapse
|
9
|
Liu Q, Li S, Qiu Y, Zhang J, Rios FJ, Zou Z, Touyz RM. Cardiovascular toxicity of tyrosine kinase inhibitors during cancer treatment: Potential involvement of TRPM7. Front Cardiovasc Med 2023; 10:1002438. [PMID: 36818331 PMCID: PMC9936099 DOI: 10.3389/fcvm.2023.1002438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) are a class of membrane spanning cell-surface receptors that transmit extracellular signals through the membrane to trigger diverse intracellular signaling through tyrosine kinases (TKs), and play important role in cancer development. Therapeutic approaches targeting RTKs such as vascular endothelial growth factor receptor (VEGFR), epidermal growth factor receptor (EGFR), and platelet-derived growth factor receptor (PDGFR), and TKs, such as c-Src, ABL, JAK, are widely used to treat human cancers. Despite favorable benefits in cancer treatment that prolong survival, these tyrosine kinase inhibitors (TKIs) and monoclonal antibodies targeting RTKs are also accompanied by adverse effects, including cardiovascular toxicity. Mechanisms underlying TKI-induced cardiovascular toxicity remain unclear. The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed chanzyme consisting of a membrane-based ion channel and intracellular α-kinase. TRPM7 is a cation channel that regulates transmembrane Mg2+ and Ca2+ and is involved in a variety of (patho)physiological processes in the cardiovascular system, contributing to hypertension, cardiac fibrosis, inflammation, and atrial arrhythmias. Of importance, we and others demonstrated significant cross-talk between TRPM7, RTKs, and TK signaling in different cell types including vascular smooth muscle cells (VSMCs), which might be a link between TKIs and their cardiovascular effects. In this review, we summarize the implications of RTK inhibitors (RTKIs) and TKIs in cardiovascular toxicities during anti-cancer treatment, with a focus on the potential role of TRPM7/Mg2+ as a mediator of RTKI/TKI-induced cardiovascular toxicity. We also describe the important role of TRPM7 in cancer development and cardiovascular diseases, and the interaction between TRPM7 and RTKs, providing insights for possible mechanisms underlying cardiovascular disease in cancer patients treated with RTKI/TKIs.
Collapse
Affiliation(s)
- Qing Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suyao Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuran Qiu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayu Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Francisco J. Rios
- Research Institute of McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Zhiguo Zou
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Zhiguo Zou ✉
| | - Rhian M. Touyz
- Research Institute of McGill University Health Centre, McGill University, Montreal, QC, Canada,*Correspondence: Rhian M. Touyz ✉
| |
Collapse
|
10
|
The Response of the Human Umbilical Vein Endothelial Cell Transcriptome to Variation in Magnesium Concentration. Nutrients 2022; 14:nu14173586. [PMID: 36079843 PMCID: PMC9460622 DOI: 10.3390/nu14173586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular endothelial cells have a critical role in the maintenance of cardiovascular function. Evidence suggests that endothelial function may be compromised under conditions of magnesium deficiency, which increases vulnerability to inflammation. Whole genome transcription analysis was used to explore the acute (24 h) effects of magnesium on human umbilical vascular endothelial cells (HUVEC) cultured in low (0.1 mM) or high (5 mM) concentrations. With low magnesium 2728 transcripts were differentially expressed compared to the 1 mM control cultures and 3030 were differentially expressed with high magnesium. 615 transcripts were differentially expressed under both conditions, of which only 34 showed a concentration-dependent response. Analysis indicated that cellular organisation and biogenesis and key cellular processes such as apoptosis were impacted by both low and high conditions. High magnesium also influenced protein binding functions, intracellular signal transduction, metabolic and catalytic processes. Both conditions impacted on stress-related processes, in particular the inflammatory response. Key mediators of calcium-dependent regulation of gene expression were responsive to both high and low magnesium conditions. The HUVEC transcriptome is highly sensitive to acute changes in the concentration of magnesium in culture medium. The findings of this study support the view that whilst inflammation is an important process that is responsive to magnesium, the function of the endothelium may be impacted by other magnesium-induced changes including maintenance of cellular integrity, receptor expression and metabolic functions. The high proportion of transcripts that did not show a concentration-dependent response suggests variation in magnesium may elicit indirect changes, possibly mediated by other ions.
Collapse
|
11
|
Chen TM, Huang CM, Hsieh MS, Lin CS, Lee WH, Yeh CT, Liu SC. TRPM7 via calcineurin/NFAT pathway mediates metastasis and chemotherapeutic resistance in head and neck squamous cell carcinoma. Aging (Albany NY) 2022; 14:5250-5270. [PMID: 35771152 PMCID: PMC9271301 DOI: 10.18632/aging.204154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 06/23/2022] [Indexed: 12/18/2022]
Abstract
The exact mechanisms of Head and neck squamous carcinoma (HNSCC) chemoresistance and metastatic transformation remain unclear. In recent decades, members of the transient receptor potential (TRP) channel family have been proposed as potential biomarkers and/or drug targets in cancer treatment. First, in a TCGA cohort of HNSCC, TRPM7 is highly expressed in cancer tissues, especially the expression in invasive cancer tissues is statistically significant (p>0.001). In GEO and TCGA cohort, patients with high expression of TRPM7 and NFATC2 have poor overall survival rates. The expression of TRPM7 and NFATC2 showed a positive correlation. Compared to human normal oral keratinocytes (hNOK), TRPM7 is overexpressed in FaDU, SAS, and TW2.6 cell lines. Similarly, patients with HNSCC exhibited higher TRPM7 expression than non-HNSCC subjects, and this high TRPM7 expression was associated with worse 5-year overall survival. Furthermore, TRPM7 inversely correlated with E-cadherin, but positively correlated with Vimentin, NANOG, and BMI-1 mRNA levels. Consistent with this, we demonstrated the overexpression of TRPM7 in cisplatin-resistant subjects, compared to the cisplatin-sensitive counterparts. Moreover, shRNA-mediated silencing of TRPM7 significantly suppressed the migration, invasion, colony formation, and tumorsphere formation of SAS cells, with associated downregulation of Snail, c-Myc, cyclin D1, SOX2, OCT4, and NANOG proteins expression. Finally, compared with the untreated wild-type SAS cells or cisplatin-treated cells, shTRPM7 alone or in combination with cisplatin significantly inhibited tumorsphere and colony formation. These findings serving as the basis for development of novel therapeutic strategies against metastasis and chemoresistance, while providing new insights into TRPM7 biology and activity in HNSCC.
Collapse
Affiliation(s)
- Tsung-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan.,Department of Otolaryngology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
| | - Chih-Ming Huang
- Department of Otolaryngology, Taitung Mackay Memorial Hospital, Taitung City 950408, Taiwan.,Department of Nursing, Tajen University, Yanpu 90741, Pingtung County, Taiwan
| | - Ming-Shou Hsieh
- Department of Medical Research and Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Chun-Shu Lin
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan
| | - Wei-Hwa Lee
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Chi-Tai Yeh
- Department of Medical Research and Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City 235, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu City 30015, Taiwan
| | - Shao-Cheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan
| |
Collapse
|
12
|
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
|
13
|
Yeung BHY, Griffiths K, Berger L, Paudel O, Shin MK, Rui L, Sham JSK, Polotsky VY, Tang WY. Leptin Induces Epigenetic Regulation of Transient Receptor Potential Melastatin 7 in Rat Adrenal Pheochromocytoma Cells. Am J Respir Cell Mol Biol 2021; 65:214-221. [PMID: 33891828 DOI: 10.1165/rcmb.2020-0374oc] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Obesity elevates the plasma level of leptin, which has been associated with hypertension. Our recent studies in mice demonstrated that leptin increases blood pressure by activating the carotid sinus nerve, which transmits the chemosensory input from carotid bodies (CBs) to the medullary centers, and that the effect of leptin is mediated via Trpm7 (TRP [transient receptor potential] melastatin 7) channels in CB glomus cells. We also found that Trpm7 overexpression and Trpm7 promoter demethylation in CBs correlate positively with the hyperleptinemia and leptin receptor overexpression in CBs. Hence, we postulated that leptin epigenetically regulates Trpm7 expression in CBs. We addressed our hypothesis by using rat adrenal pheochromocytoma (PC12) cells as a model of CB glomus cells. PC12 cells expressing LEPRb (long, active form of leptin receptor) showed dramatic induction of the promoter activity and expression of Trpm7 upon leptin treatment. The increased Trpm7 expression coincided with the reduction of CpG site-specific methylation and trimethylation of H3K27 (H3 [histone 3] K27 [lysine 27]) and the increase of acetylation of H3K27 and trimethylation of H3K4 (H3 lysine 4) at the Trpm7 promoter. The inhibitor of STAT3 (signal transducer and activator of transcription 3) signaling, SD1008, reversed the leptin-induced Trpm7 promoter activity via modulations of the binding of pSTAT3 (phosphorylated STAT3) and DNMT3B (DNA methyltransferase 3B) and modifications of H3K27 and H3K4 at the Trpm7 promoter. Our results suggest that leptin-activated pSTAT3 epigenetically regulates the transcription of Trpm7 through DNA methylation and histone modifications. Because epigenetic changes are reversible, targeting epigenetic modifications of Trpm7 may serve as a new therapeutic approach for the treatment of hypertension in obesity.
Collapse
Affiliation(s)
- Bonnie Ho-Yee Yeung
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, and.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Kelly Griffiths
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, and
| | - Liron Berger
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Omkar Paudel
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Mi-Kyung Shin
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Liangyou Rui
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, University of Michigan, Ann Arbor, Michigan
| | - James S K Sham
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, and.,Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Vsevolod Y Polotsky
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Wan-Yee Tang
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, and.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
14
|
Jeong JH, Lee SH, Kho AR, Hong DK, Kang DH, Kang BS, Park MK, Choi BY, Choi HC, Lim MS, Suh SW. The Transient Receptor Potential Melastatin 7 (TRPM7) Inhibitors Suppress Seizure-Induced Neuron Death by Inhibiting Zinc Neurotoxicity. Int J Mol Sci 2020; 21:ijms21217897. [PMID: 33114331 PMCID: PMC7663745 DOI: 10.3390/ijms21217897] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/25/2022] Open
Abstract
Transient receptor potential melastatin 7 (TRPM7) is an ion channel that mediates monovalent cations out of cells, as well as the entry of divalent cations, such as zinc, magnesium, and calcium, into the cell. It has been reported that inhibitors of TRPM7 are neuroprotective in various neurological diseases. Previous studies in our lab suggested that seizure-induced neuronal death may be caused by the excessive release of vesicular zinc and the subsequent accumulation of zinc in the neurons. However, no studies have evaluated the effects of carvacrol and 2-aminoethoxydiphenyl borate (2-APB), both inhibitors of TRPM7, on the accumulation of intracellular zinc in dying neurons following seizure. Here, we investigated the therapeutic efficacy of carvacrol and 2-APB against pilocarpine-induced seizure. Carvacrol (50 mg/kg) was injected once per day for 3 or 7 days after seizure. 2-APB (2 mg/kg) was also injected once per day for 3 days after seizure. We found that inhibitors of TRPM7 reduced seizure-induced TRPM7 overexpression, intracellular zinc accumulation, and reactive oxygen species production. Moreover, there was a suppression of oxidative stress, glial activation, and the blood–brain barrier breakdown. In addition, inhibitors of TRPM7 remarkably decreased apoptotic neuron death following seizure. Taken together, the present study demonstrates that TRPM7-mediated zinc translocation is involved in neuron death after seizure. The present study suggests that inhibitors of TRPM7 may have high therapeutic potential to reduce seizure-induced neuron death.
Collapse
Affiliation(s)
- Jeong Hyun Jeong
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Song Hee Lee
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - A Ra Kho
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Dae Ki Hong
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Dong Hyeon Kang
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Beom Seok Kang
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Min Kyu Park
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Bo Young Choi
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
- Correspondence: (B.Y.C.); (H.C.C.); (M.-S.L.); (S.W.S.); Tel.: +82-10-8573-6364 (S.W.S.)
| | - Hui Chul Choi
- Department of Neurology, Hallym University, College of Medicine, Chuncheon 24252, Korea
- Correspondence: (B.Y.C.); (H.C.C.); (M.-S.L.); (S.W.S.); Tel.: +82-10-8573-6364 (S.W.S.)
| | - Man-Sup Lim
- Department of Medical Education, Hallym University, College of Medicine, Chuncheon 24252, Korea
- Correspondence: (B.Y.C.); (H.C.C.); (M.-S.L.); (S.W.S.); Tel.: +82-10-8573-6364 (S.W.S.)
| | - Sang Won Suh
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
- Correspondence: (B.Y.C.); (H.C.C.); (M.-S.L.); (S.W.S.); Tel.: +82-10-8573-6364 (S.W.S.)
| |
Collapse
|
15
|
A Review of the Action of Magnesium on Several Processes Involved in the Modulation of Hematopoiesis. Int J Mol Sci 2020; 21:ijms21197084. [PMID: 32992944 PMCID: PMC7582682 DOI: 10.3390/ijms21197084] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 12/18/2022] Open
Abstract
Magnesium (Mg2+) is an essential mineral for the functioning and maintenance of the body. Disturbances in Mg2+ intracellular homeostasis result in cell-membrane modification, an increase in oxidative stress, alteration in the proliferation mechanism, differentiation, and apoptosis. Mg2+ deficiency often results in inflammation, with activation of inflammatory pathways and increased production of proinflammatory cytokines by immune cells. Immune cells and others that make up the blood system are from hematopoietic tissue in the bone marrow. The hematopoietic tissue is a tissue with high indices of renovation, and Mg2+ has a pivotal role in the cell replication process, as well as DNA and RNA synthesis. However, the impact of the intra- and extracellular disturbance of Mg2+ homeostasis on the hematopoietic tissue is little explored. This review deals specifically with the physiological requirements of Mg2+ on hematopoiesis, showing various studies related to the physiological requirements and the effects of deficiency or excess of this mineral on the hematopoiesis regulation, as well as on the specific process of erythropoiesis, granulopoiesis, lymphopoiesis, and thrombopoiesis. The literature selected includes studies in vitro, in animal models, and in humans, giving details about the impact that alterations of Mg2+ homeostasis can have on hematopoietic cells and hematopoietic tissue.
Collapse
|
16
|
Magnesium and calciprotein particles in vascular calcification: the good cop and the bad cop. Curr Opin Nephrol Hypertens 2020; 28:368-374. [PMID: 31045659 DOI: 10.1097/mnh.0000000000000509] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Vascular calcification is a major contributor to increased cardiovascular mortality in chronic kidney disease (CKD). Recently, calciprotein particles (CPP) were identified to drive the calcification process. CPP may explain the effects of high phosphate on vascular calcification. Magnesium is a promising novel therapeutic approach to halt vascular calcification, because it inhibits CPP maturation and is associated with reduced cardiovascular mortality in CKD. We aim to examine the current evidence for the role of CPP in the calcification process and to explain how magnesium prevents calcification. RECENT FINDINGS A recent meta-analysis concluded that reducing high phosphate levels in CKD patients does not associate with lowering cardiovascular mortality. Inhibition of CPP formation prevents phosphate-induced calcification in vitro. Consequently, delaying CPP formation and maturation may be a clinical approach to reduce calcification. Magnesium inhibits CPP maturation and vascular calcification. Clinical pilot studies suggest that magnesium is a promising intervention strategy against calcification in CKD patients. SUMMARY CPP induce vascular calcification and are modulated by serum phosphate and magnesium concentrations. Magnesium is a strong inhibitor of CPP maturation and therefore, a promising therapeutic approach to reduce vascular calcification in CKD. Currently, several studies are being performed to determine the clinical outcomes of magnesium supplementation in CKD.
Collapse
|
17
|
Lee S, Lee S, Lee A, Sim HJ, Kim GA, Kang BJ, Kim WH. The Presence and Distribution of TRPM7 in the Canine Mammary Glands. Animals (Basel) 2020; 10:ani10030466. [PMID: 32168794 PMCID: PMC7142925 DOI: 10.3390/ani10030466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
The transient receptor potential melastatin-subfamily member 7 (TRPM7) cation channel is a bifunctional ion channel with intrinsic kinase activity and is ubiquitously expressed in the animal/human body. Accumulated knowledge of TRPM7 suggests that it plays an essential role in normal physiological processes, including the development, survival, proliferation, differentiation, and migration of cells. The aim of this study was to demonstrate the presence and expression patterns of TRPM7 in normal canine mammary glands using reverse transcription-polymerase chain reaction (RT-PCR), Western blotting, and immunohistochemistry. Normal mammary gland tissue samples were obtained from five female beagle dogs. RT-PCR and sequencing of the amplified PCR products demonstrated the presence of TRPM7 mRNA in normal mammary glands, and the presence of TRPM7 protein was confirmed by Western blotting. Immunohistochemical investigations demonstrated the expression of TRPM7 in the apical membrane of acinar and ductal epithelial cells in the canine mammary glands. These results provide the first evidence of the presence and distribution of TRPM7 in the canine mammary gland and could help explain the physiological and pathological roles of TRPM7 in the canine mammary gland; however, additional studies are required to elucidate these roles.
Collapse
Affiliation(s)
- Sungin Lee
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (S.L.); (S.L.); (H.J.S.); (G.A.K.); (B.-J.K.)
| | - Seulji Lee
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (S.L.); (S.L.); (H.J.S.); (G.A.K.); (B.-J.K.)
| | - Aeri Lee
- Seeu Animal Medical Center, 24, Ichon-ro 64 gil, Younsan-gu, Seoul 04427, Korea;
| | - Hun Ju Sim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (S.L.); (S.L.); (H.J.S.); (G.A.K.); (B.-J.K.)
| | - Geon A. Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (S.L.); (S.L.); (H.J.S.); (G.A.K.); (B.-J.K.)
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (S.L.); (S.L.); (H.J.S.); (G.A.K.); (B.-J.K.)
| | - Wan Hee Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (S.L.); (S.L.); (H.J.S.); (G.A.K.); (B.-J.K.)
- Correspondence:
| |
Collapse
|
18
|
Zhou DM, Sun LL, Zhu J, Chen B, Li XQ, Li WD. MiR-9 promotes angiogenesis of endothelial progenitor cell to facilitate thrombi recanalization via targeting TRPM7 through PI3K/Akt/autophagy pathway. J Cell Mol Med 2020; 24:4624-4632. [PMID: 32147957 PMCID: PMC7176881 DOI: 10.1111/jcmm.15124] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/11/2019] [Accepted: 01/06/2020] [Indexed: 12/13/2022] Open
Abstract
Endothelial progenitor cells (EPCs) have emerged as a promising therapeutic choice for thrombi recanalization. However, this role of EPCs is confined by some detrimental factors. The aim of this study was to explore the role of the miR‐9‐5p in regulation of the proliferation, migration and angiogenesis of EPCs and the subsequent therapeutic role in thrombosis event. Wound healing, transwell assay, tube formation assay and in vivo angiogenesis assay were carried out to measure cell migration, invasion and angiogenic abilities, respectively. Western blot was performed to elucidate the relationship between miR‐9‐5p and TRPM7 in the autophagy pathway. It was found that miR‐9‐5p could promote migration, invasion and angiogenesis of EPCs by attenuating TRPM7 expression via activating PI3K/Akt/autophagy pathway. In conclusion, miR‐9‐5p, targets TRPM7 via the PI3K/Ak/autophagy pathway, thereby mediating cell proliferation, migration and angiogenesis in EPCs. Acting as a potential therapeutic target, miR‐9‐5p may play an important role in the prognosis of DVT.
Collapse
Affiliation(s)
- Dong-Ming Zhou
- Department of Hematology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Li-Li Sun
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.,Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Zhu
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Bing Chen
- Department of Hematology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Wen-Dong Li
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| |
Collapse
|
19
|
Inhibition of TRPM7 blocks MRTF/SRF-dependent transcriptional and tumorigenic activity. Oncogene 2019; 39:2328-2344. [PMID: 31844251 DOI: 10.1038/s41388-019-1140-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 11/24/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022]
Abstract
Myocardin-related transcription factors A and B (MRTFs) are coactivators of Serum Response Factor (SRF) that mediates the expression of genes involved in cell proliferation, migration and differentiation. There is mounting evidence that MRTFs and SRF represent promising targets for hepatocellular carcinoma (HCC) growth. Since MRTF-A nuclear localization is a prerequisite for its transcriptional activity and oncogenic properties, we searched for pharmacologically active compounds able to redistribute MRTF-A to the cytoplasm. We identified NS8593, a negative gating modulator of the transient receptor potential cation channel TRPM7, as a novel inhibitor of MRTF-A nuclear localization and transcriptional activity. Using a pharmacological approach and targeted genome editing, we investigated the functional contribution of TRPM7, a unique ion channel containing a serine-threonine kinase domain, to MRTF transcriptional and tumorigenic activity. We found that TRPM7 function regulates RhoA activity and subsequently actin polymerization, MRTF-A-Filamin A complex formation and MRTF-A/SRF target gene expression. Mechanistically, TRPM7 signaling relies on TRPM7 channel-mediated Mg2+ influx and phosphorylation of RhoA by TRPM7 kinase. Pharmacological blockade of TRPM7 results in oncogene-induced senescence of hepatocellular carcinoma (HCC) cells in vitro and in vivo in HCC xenografts. Hence, inhibition of the TRPM7/MRTF axis emerges as a promising strategy to curb HCC growth.
Collapse
|
20
|
Redox TRPs in diabetes and diabetic complications: Mechanisms and pharmacological modulation. Pharmacol Res 2019; 146:104271. [PMID: 31096011 DOI: 10.1016/j.phrs.2019.104271] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/04/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022]
Abstract
Transient receptor potential (TRP) channels have shown to be involved in a wide variety of physiological functions and pathophysiological conditions. Modulation of TRP channels reported to play a major role in number of disorders starting from central nervous system related disorders to cardiovascular, inflammatory, cancer, gastrointestinal and metabolic diseases. Recently, a subset of TRP ion channels called redox TRPs gained importance on account of their ability to sense the cellular redox environment and respond accordingly to such redox stimuli. Diabetes, the silent epidemic of the world is increasing at an alarming rate in spite of novel therapeutic interventions. Moreover, diabetes and its associated complications are reported to arise due to a change in oxidative status of cell induced by hyperglycemia. Such a change in cellular oxidative status can modulate the activities of various redox TRP channels (TRPA1, TRPC5, TRPMs and TRPV1). Targeting redox TRPs have potential in diabetes and diabetic complications like neuropathy, cardiomyopathy, retinopathy, cystopathy, and encephalopathy. Thus in this review, we have discussed the activities of different redox sensing TRPs in diabetes and diabetic complications and how they can be modulated pharmacologically, so as to consider them a potential novel therapeutic target in treating diabetes and its comorbidity.
Collapse
|
21
|
YALÇIN E, PALA Ş, ATILGAN* R, KULOĞLU T, ÖNALAN E, ARTAŞ G, BURAN İ. Is there any difference between endometrial hyperplasia and endometrial carcinoma in terms of expression of TRPM2 and TRPM7 ion channels? Turk J Med Sci 2019; 49:653-660. [PMID: 30997980 PMCID: PMC7018370 DOI: 10.3906/sag-1810-176] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Background/aim This study compared TRPM2 and TRPM7 ion channel gene expression and immunohistochemical staining in endometrial hyperplasia and endometrium adenocarcinoma. Materials and methods Sections were taken from paraffin blocks of 120 patients who were divided into 6 groups as follows: G1 (n = 20), proliferative endometrium (PE); G2 (n = 20), EH without atypia; G3 (n = 20), EH with atypia; G4 (n = 20), stage 1A, grade 1 EC; G5 (n = 20), stage 1A, grade 2 EC; and G6 (n = 20), stage 1A, grade 3 EC. TRPM2 and TRPM7 genes were analyzed with qRT-PCR in paraffin-embedded tissue samples. Under light microscopy, TRPM2 and TRPM7 immunostaining scores of the samples taken from polylysine slides were evaluated. Results Compared to G1, TRPM2 mRNA gene expression was significantly downregulated in G3 and G5. TRPM2 immunoreactivity scores were similar in all groups. TRPM7 mRNA gene expression was significantly downregulated in G2, G3, and G6 when compared to G1. TRPM7 immunoreactivity scores were similar in G1, G2, and G3, but significantly decreased in G4, G5, and G6 Conclusion Reduction in TRPM7 ion channel activity may be a progression marker for endometrial hyperplasia regardless of the atypical criteria.
Collapse
Affiliation(s)
- Emre YALÇIN
- Department of Obstetrics and Gynecology, School of Medicine, Fırat University, ElazığTurkey
| | - Şehmus PALA
- Department of Obstetrics and Gynecology, School of Medicine, Fırat University, ElazığTurkey
| | - Remzi ATILGAN*
- Department of Obstetrics and Gynecology, School of Medicine, Fırat University, ElazığTurkey
| | - Tuncay KULOĞLU
- Department of Histology and Embryology, School of Medicine, Fırat University, ElazığTurkey
| | - Ebru ÖNALAN
- Department of Department of Medical Biology, School of Medicine, Fırat University, ElazığTurkey
| | - Gökhan ARTAŞ
- Department of Pathology, School of Medicine, Fırat University, ElazığTurkey
| | - İlay BURAN
- Department of Department of Medical Biology, School of Medicine, Fırat University, ElazığTurkey
| |
Collapse
|
22
|
Qiao W, Lan XM, Ma HX, Chan JYK, Lui VWY, Yeung KWK, Kwong DLW, Hu Z, Tsoi JKH, Matinlinna JP, Su YX. Effects of Salivary Mg on Head and Neck Carcinoma via TRPM7. J Dent Res 2019; 98:304-312. [PMID: 30513244 DOI: 10.1177/0022034518813359] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Magnesium (Mg) has been known to play vital roles in regulating growth and various metabolic processes. In recent years, the association between Mg and tumorigenesis has raised more and more attention. However, the effects of Mg on the progression of head and neck carcinoma (HNC), as well as the mechanism behind it, remain undefined. In this study, the roles of Mg in tumorigenic activities were tested in CAL27 and FaDu cells as well as in a xenograft tumor model in nude mice. We demonstrated that a moderate increase in extracellular Mg contributed to the proliferation, migration, and invasion of 2 HNC cell lines, while the addition of Mg in drinking water promoted the growth of xenograft tumors in mice without altering their serum Mg levels. Moreover, TRPM7, a major Mg transporter, was shown to be essential for the tumorigenic activities of HNC and the Mg-induced promotive effects on HNC cells and was further shown to be associated with the activation of AKT/mTOR (mammalian target of rapamycin) signaling. In a preliminary clinical study, we determined the Mg ion concentrations in the stimulated saliva from 72 patients with nasopharynx carcinoma and 12 healthy individuals. Our data revealed that the salivary Mg levels of subjects with nasopharynx carcinoma were significantly higher than those of the healthy controls. This is correlated with our finding showing TRPM7 to be overexpressed in tumor tissues harvested from 9 patients with HNC. Therefore, we can conclude that salivary Mg level, within a certain range, could act as a risk factor for the progression of HNC, which involves the activation of AKT/mTOR signaling pathways through the TRPM7 channel. The control of salivary Mg level and the intervention of TRPM7 should not be ignored during the study of HNC.
Collapse
Affiliation(s)
- W Qiao
- 1 Dental Materials Science, Applied Oral Sciences, Faculty of Dentistry, University of Hong Kong, Hong Kong, China
- 2 Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - X M Lan
- 3 Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Hong Kong, Hong Kong, China
| | - H X Ma
- 4 Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - J Y K Chan
- 5 Department of Otorhinolaryngology-Head and Neck Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - V W Y Lui
- 6 School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - K W K Yeung
- 2 Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - D L W Kwong
- 7 Department of Clinical Oncology, University of Hong Kong, Hong Kong, China
| | - Z Hu
- 4 Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - J K H Tsoi
- 1 Dental Materials Science, Applied Oral Sciences, Faculty of Dentistry, University of Hong Kong, Hong Kong, China
| | - J P Matinlinna
- 1 Dental Materials Science, Applied Oral Sciences, Faculty of Dentistry, University of Hong Kong, Hong Kong, China
| | - Y X Su
- 3 Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Hong Kong, Hong Kong, China
| |
Collapse
|
23
|
Broertjes J, Klarenbeek J, Habani Y, Langeslag M, Jalink K. TRPM7 residue S1269 mediates cAMP dependence of Ca2+ influx. PLoS One 2019; 14:e0209563. [PMID: 30615643 PMCID: PMC6322742 DOI: 10.1371/journal.pone.0209563] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023] Open
Abstract
The nonspecific divalent cation channel TRPM7 (transient receptor potential-melastatin-like 7) is involved in many Ca2+ and Mg2+-dependent cellular processes, including survival, proliferation and migration. TRPM7 expression predicts metastasis and recurrence in breast cancer and several other cancers. In cultured cells, it can induce an invasive phenotype by promoting Ca2+-mediated epithelial-mesenchymal transition. We previously showed that in neuroblastoma cells that overexpress TRPM7 moderately, stimulation with Ca2+-mobilizing agonists leads to a characteristic sustained influx of Ca2+. Here we report that sustained influx through TRPM7 is abruptly abrogated by elevating intracellular levels of cyclic adenosine monophosphate (cAMP). Using pharmacological inhibitors and overexpression studies we show that this blockage is mediated by the cAMP effector Protein Kinase A (PKA). Mutational analysis demonstrates that the Serine residue S1269, which is present proximal to the coiled-coil domain within the protein c-terminus, is responsible for sensitivity to cAMP.
Collapse
Affiliation(s)
- Jorrit Broertjes
- Division of Cell Biology I, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeffrey Klarenbeek
- Division of Cell Biology I, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Yasmin Habani
- Division of Cell Biology I, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michiel Langeslag
- Division of Cell Biology I, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kees Jalink
- Division of Cell Biology I, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
24
|
Zhelay T, Wieczerzak KB, Beesetty P, Alter GM, Matsushita M, Kozak JA. Depletion of plasma membrane-associated phosphoinositides mimics inhibition of TRPM7 channels by cytosolic Mg 2+, spermine, and pH. J Biol Chem 2018; 293:18151-18167. [PMID: 30305398 PMCID: PMC6254349 DOI: 10.1074/jbc.ra118.004066] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/04/2018] [Indexed: 12/20/2022] Open
Abstract
Transient receptor potential cation channel subfamily M member 7 (TRPM7) is an ion channel/protein kinase belonging to the TRP melastatin and eEF2 kinase families. Under physiological conditions, most native TRPM7 channels are inhibited by cytoplasmic Mg2+, protons, and polyamines. Currents through these channels (ITRPM7) are robustly potentiated when the cell interior is exchanged with low Mg2+-containing buffers. ITRPM7 is also potentiated by phosphatidyl inositol bisphosphate (PI(4,5)P2) and suppressed by its hydrolysis. Here we characterized internal Mg2+- and pH-mediated inhibition of TRPM7 channels in HEK293 cells overexpressing WT voltage-sensing phospholipid phosphatase (VSP) or its catalytically inactive variant VSP-C363S. VSP-mediated depletion of membrane phosphoinositides significantly increased channel sensitivity to Mg2+ and pH. Proton concentrations that were too low to inhibit ITRPM7 when the VSP-C363S variant was expressed (pH 8.2) became inhibitory in WT VSP-expressing cells. At pH 6.5, protons inhibited ITRPM7 both in WT and VSP C363S-expressing cells but with a faster time course in the WT VSP-expressing cells. Inhibition by 150 μm Mg2+ was also significantly faster in the WT VSP-expressing cells. Cellular PI(4,5)P2 depletion increased the sensitivity of TRPM7 channels to the inhibitor 2-aminoethyl diphenyl borinate, which acidifies the cytosol. Single substitutions at Ser-1107 of TRPM7, reducing its sensitivity to Mg2+, also decreased its inhibition by spermine and acidic pH. Furthermore, these channel variants were markedly less sensitive to VSP-mediated PI(4,5)P2 depletion than the WT. We conclude that the internal Mg2+-, polyamine-, and pH-mediated inhibition of TRPM7 channels is not direct but, rather, reflects electrostatic screening and resultant disruption of PI(4,5)P2-channel interactions.
Collapse
Affiliation(s)
- Tetyana Zhelay
- From the Departments of Neuroscience, Cell Biology, and Physiology and
| | | | - Pavani Beesetty
- From the Departments of Neuroscience, Cell Biology, and Physiology and
| | - Gerald M Alter
- Biochemistry and Molecular Biology, Wright State University, Dayton, Ohio 45435 and
| | - Masayuki Matsushita
- the Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - J Ashot Kozak
- From the Departments of Neuroscience, Cell Biology, and Physiology and.
| |
Collapse
|
25
|
Liu A, Wu J, Yang C, Wu Y, Zhang Y, Zhao F, Wang H, Yuan L, Song L, Zhu T, Fan Y, Yang B. TRPM7 in CHBP-induced renoprotection upon ischemia reperfusion-related injury. Sci Rep 2018; 8:5510. [PMID: 29615639 PMCID: PMC5882857 DOI: 10.1038/s41598-018-22852-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/01/2018] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential melastatin 7 (TRPM7) is a membrane ion channel and kinase. TRPM7 was abundantly expressed in the kidney, and up-regulated by ischemia reperfusion (IR) injury. Our previous studies showed that cyclic helix B peptide (CHBP) improved renal IR-related injury, but its underlying mechanism is not well defined. IR-related injury was established in renal tubular epithelial cells (TCMK-1 and HK-2) via 12 to 24-h hypoxia (H) followed by 2-24 h reoxygenation (R), and in mouse kidneys subjected to 30-min ischemia and 12-h to 7-day reperfusion. TRPM7-like current in TCMK-1 cells, TRPM7 mRNA and protein in the in vitro and in vivo models were increased, but reversed by CHBP. TRPM7 was also positively associated with LDH, HMGB1, caspase-3, Bax/Bcl-2, inflammation, apoptosis, tubulointerstitial damage and renal function respectively. Furthermore, silencing TRPM7 improved injury parameters, renal histology and function in the both models. Specific TRPM7 agonist, bradykinin, exaggerated HR induced injury in TCMK-1 cells, and partially blocked the renoprotection of CHBP as well. In conclusion, TRPM7 is involved not only in IR-related injury, but also CHBP-induced renoprotection, which are through its ion channel and subsequent affects inflammation and apoptosis. Therefore, TRPM7 could be a potential biomarker for IR-induced acute kidney injury.
Collapse
Affiliation(s)
- Aifen Liu
- Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, Jiangsu, 226001, China
| | - Jing Wu
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University; Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Yuanyuan Wu
- Department of Pathology, Medical College of Nantong University, Nantong, Jiangsu, 226001, China
| | - Yufang Zhang
- Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, Jiangsu, 226001, China
| | - Fengbo Zhao
- Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, Jiangsu, 226001, China
| | - Hui Wang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Li Yuan
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Lirui Song
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University; Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Yaping Fan
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Bin Yang
- Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, Jiangsu, 226001, China. .,Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China. .,Department of Infection, Immunity and Inflammation, University of Leicester, Leicester General Hospital, University Hospital of Leicester, Leicester, LE1 9HN, United Kingdom.
| |
Collapse
|
26
|
|
27
|
Zhao Z, Zhang M, Duan X, Chen Y, Li E, Luo L, Wu W, Peng Z, Qiu H, Zeng G. TRPM7 Regulates AKT/FOXO1–Dependent Tumor Growth and Is an Independent Prognostic Indicator in Renal Cell Carcinoma. Mol Cancer Res 2018; 16:1013-1023. [PMID: 29545479 DOI: 10.1158/1541-7786.mcr-17-0767] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/15/2018] [Accepted: 03/06/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Zhijian Zhao
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China.
| | - Mengping Zhang
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaolu Duan
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Yiwen Chen
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Ermao Li
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Lianmin Luo
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Wenqi Wu
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Zhenwei Peng
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Huijuan Qiu
- Department of VIP, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Guohua Zeng
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China.
| |
Collapse
|
28
|
Sander P, Mostafa H, Soboh A, Schneider JM, Pala A, Baron AK, Moepps B, Wirtz CR, Georgieff M, Schneider M. Vacquinol-1 inducible cell death in glioblastoma multiforme is counter regulated by TRPM7 activity induced by exogenous ATP. Oncotarget 2018; 8:35124-35137. [PMID: 28410232 PMCID: PMC5471040 DOI: 10.18632/oncotarget.16703] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/15/2017] [Indexed: 12/29/2022] Open
Abstract
Glioblastomas (GBM) are the most malignant brain tumors in humans and have a very poor prognosis. New therapeutic options are urgently needed. A novel drug, Vacquinol-1 (Vac), a quinolone derivative, displays promising properties by inducing rapid cell death in GBM but not in non-transformed tissues. Features of this type of cell death are compatible with a process termed methuosis. Here we tested Vac on a highly malignant glioma cell line observed by long-term video microscopy. Human dental-pulp stem cells (DPSCs) served as controls. A major finding was that an exogenous ATP concentration of as little as 1 μM counter regulated the Vac-induced cell death. Studies using carvacrol, an inhibitor of transient receptor potential cation channel, subfamily M, member 7 (TRPM7), demonstrated that the ATP-inducible inhibitory effect is likely to be via TRPM7. Exogenous ATP is of relevance in GBM with large necrotic areas. Our results support the use of GBM cultures with different grades of malignancy to address their sensitivity to methuosis. The video-microscopy approach presented here allows decoding of signaling pathways as well as mechanisms of chemotherapeutic resistance by long-term observation. Before implementing Vac as a novel therapeutic drug in GBM, cells from each individual patient need to be assessed for their ATP sensitivity. In summary, the current investigation supports the concept of methuosis, described as non-apoptotic cell death and a promising approach for GBM treatment. Tissue-resident ATP/necrosis may interfere with this cell-death pathway but can be overcome by a natural compound, carvacrol that even penetrates the blood-brain barrier.
Collapse
Affiliation(s)
- Philip Sander
- Division of Experimental Anesthesiology, University Hospital Ulm, 89081 Ulm, Germany
| | - Haouraa Mostafa
- Division of Experimental Anesthesiology, University Hospital Ulm, 89081 Ulm, Germany
| | - Ayman Soboh
- Division of Experimental Anesthesiology, University Hospital Ulm, 89081 Ulm, Germany
| | - Julian M Schneider
- Division of Experimental Anesthesiology, University Hospital Ulm, 89081 Ulm, Germany
| | - Andrej Pala
- Department of Neurosurgery, Bezirkskrankenhaus Guenzburg, 89312 Guenzburg, Germany
| | - Ann-Kathrin Baron
- Department of Operative Dentistry and Periodontology, University Hospital Ulm, 89081 Ulm, Germany
| | - Barbara Moepps
- Institute of Pharmacology and Toxicology, University Hospital Ulm, 89081 Ulm, Germany
| | - C Rainer Wirtz
- Department of Neurosurgery, Bezirkskrankenhaus Guenzburg, 89312 Guenzburg, Germany
| | - Michael Georgieff
- Department of Anesthesiology, University Hospital Ulm, 89081 Ulm, Germany
| | - Marion Schneider
- Division of Experimental Anesthesiology, University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
29
|
Abumaria N, Li W, Liu Y. TRPM7 functions in non-neuronal and neuronal systems: Perspectives on its role in the adult brain. Behav Brain Res 2018; 340:81-86. [DOI: 10.1016/j.bbr.2016.08.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/17/2016] [Accepted: 08/19/2016] [Indexed: 12/30/2022]
|
30
|
Ogata K, Tsumuraya T, Oka K, Shin M, Okamoto F, Kajiya H, Katagiri C, Ozaki M, Matsushita M, Okabe K. The crucial role of the TRPM7 kinase domain in the early stage of amelogenesis. Sci Rep 2017; 7:18099. [PMID: 29273814 PMCID: PMC5741708 DOI: 10.1038/s41598-017-18291-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 12/07/2017] [Indexed: 12/15/2022] Open
Abstract
Transient receptor potential melastatin-7 (TRPM7) is a bi-functional protein containing a kinase domain fused to an ion channel. TRPM7 is highly expressed in ameloblasts during tooth development. Here we show that TRPM7 kinase-inactive knock-in mutant mice (TRPM7 KR mice) exhibited small enamel volume with opaque white-colored incisors. The TRPM7 channel function of ameloblast-lineage cells from TRPM7 KR mice was normal. Interestingly, phosphorylation of intracellular molecules including Smad1/5/9, p38 and cAMP response element binding protein (CREB) was inhibited in ameloblasts from TRPM7 KR mice at the pre-secretory stage. An immunoprecipitation assay showed that CREB was bound to TRPM7, suggesting that direct phosphorylation of CREB by TRPM7 was inhibited in ameloblast-lineage cells from TRPM7 KR mice. These results indicate that the function of the TRPM7 kinase domain plays an important role in ameloblast differentiation, independent of TRPM7 channel activity, via phosphorylation of CREB.
Collapse
Affiliation(s)
- Kayoko Ogata
- Section of Cellular Physiology, Department of Physiological Sciences and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan.,Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan
| | - Tomoyuki Tsumuraya
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Kyoko Oka
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan.
| | - Masashi Shin
- Section of Cellular Physiology, Department of Physiological Sciences and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Fujio Okamoto
- Section of Cellular Physiology, Department of Physiological Sciences and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Hiroshi Kajiya
- Section of Cellular Physiology, Department of Physiological Sciences and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Chiaki Katagiri
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Masao Ozaki
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan
| | - Masayuki Matsushita
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Koji Okabe
- Section of Cellular Physiology, Department of Physiological Sciences and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
31
|
Gao SL, Kong CZ, Zhang Z, Li ZL, Bi JB, Liu XK. TRPM7 is overexpressed in bladder cancer and promotes proliferation, migration, invasion and tumor growth. Oncol Rep 2017; 38:1967-1976. [PMID: 28791418 PMCID: PMC5652943 DOI: 10.3892/or.2017.5883] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 06/23/2017] [Indexed: 12/22/2022] Open
Abstract
Recent findings suggest that the melastatin transient receptor potential channel 7 (TRPM7) is overexpressed in many types of cancers and is involved in tumorigenesis. However, its expression pattern and the potential role in bladder cancer remain unclear. The aim of the present study was to investigate the expression status of TRPM7 and its relationship with the development of bladder cancer. In the present study, we observed that the expression of TRPM7 was significantly elevated in bladder cancer tissues compared with that noted in the adjacent non-tumor tissues. Furthermore, increased TRPM7 expression was significantly associated with recurrence, metastasis and prognosis. In addition, after knockdown of the expression of TRPM7 by siRNA, the proliferation and the motility of T24 and 5637 cells were obviously inhibited, and downregulation of TRPM7 was found to play an important role in bladder cancer cell apoptosis. In conclusion, our findings suggest that TRPM7 plays an important role in bladder cancer, and TRPM7 may serve as a potentially unfavorable factor and novel target for human bladder cancer.
Collapse
Affiliation(s)
- Sheng-Lin Gao
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Chui-Ze Kong
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Zhe Zhang
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Ze-Liang Li
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Jian-Bin Bi
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Xian-Kui Liu
- Institute of Urology, Department of Urology, The First Affiliated Hospital of China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
32
|
Yee NS. Role of TRPM7 in Cancer: Potential as Molecular Biomarker and Therapeutic Target. Pharmaceuticals (Basel) 2017; 10:39. [PMID: 28379203 PMCID: PMC5490396 DOI: 10.3390/ph10020039] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 02/22/2017] [Accepted: 03/29/2017] [Indexed: 12/31/2022] Open
Abstract
The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed ion channel with intrinsic kinase activity. Molecular and electrophysiological analyses of the structure and activity of TRPM7 have revealed functional coupling of its channel and kinase activity. Studies have indicated the important roles of TRPM7 channel-kinase in fundamental cellular processes, physiological responses, and embryonic development. Accumulating evidence has shown that TRPM7 is aberrantly expressed and/or activated in human diseases including cancer. TRPM7 plays a variety of functional roles in cancer cells including survival, cell cycle progression, proliferation, growth, migration, invasion, and epithelial-mesenchymal transition (EMT). Data from a study using mouse xenograft of human cancer show that TRPM7 is required for tumor growth and metastasis. The aberrant expression of TRPM7 and its genetic mutations/polymorphisms have been identified in various types of carcinoma. Chemical modulators of TRPM7 channel produced inhibition of proliferation, growth, migration, invasion, invadosome formation, and markers of EMT in cancer cells. Taken together, these studies suggest the potential value of exploiting TRPM7 channel-kinase as a molecular biomarker and therapeutic target in human malignancies.
Collapse
Affiliation(s)
- Nelson S Yee
- Division of Hematology-Oncology, Department of Medicine, PennState Health Milton S. Hershey Medical Center, Program of Experimental Therapeutics, PennState Cancer Institute, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA.
| |
Collapse
|
33
|
Huang T, Ji Y, Hu D, Chen B, Zhang H, Li C, Chen G, Luo X, Zheng XW, Lin X. SNHG8 is identified as a key regulator of epstein-barr virus(EBV)-associated gastric cancer by an integrative analysis of lncRNA and mRNA expression. Oncotarget 2016; 7:80990-81002. [PMID: 27835598 PMCID: PMC5348371 DOI: 10.18632/oncotarget.13167] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 10/31/2016] [Indexed: 12/26/2022] Open
Abstract
The Epstein-Barr virus (EBV) is associated with a variety of cancers, including gastric cancer, which has one of the highest mortality rates of all human cancers. Long non-coding RNAs (lncRNAs) have been suggested to have important causal roles in gastric cancer. However, the interaction between lncRNAs and EBV has not yet been studied. To this end, we sequenced 11,311 lncRNAs and 144,826 protein-coding transcripts from four types of tissue: one non-EBV-infected gastric carcinoma (EBVnGC) and its adjacent normal tissue, and one EBV-associated gastric carcinoma (EBVaGC) and its adjacent normal tissue. Five lncRNAs showed EBVaGC-specific expression; of those, one (SNHG8) was validated using real-time PCR in an independent cohort with 88 paired gastric cancer and adjacent tissue samples. To explore the functions of SNHG8, we identified its mRNA targets on the lncRNA-mRNA co-expression network of the Illumina Body Map, which contains the RNA sequencing data of mRNAs and lncRNAs from 16 normal human tissues. SNHG8 lncRNA was found to affect several gastric cancer-specific pathways and target genes of EBV. Our results reveal the intertwined tumorigenesis mechanisms of lncRNA and EBV and identify SNHG8 as a highly possible candidate biomarker and drug target of gastric cancer.
Collapse
Affiliation(s)
- Tao Huang
- Department of Pathology, Fujian Provincial Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Ji
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Hu
- Department of Pathology, Fujian Provincial Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Baozheng Chen
- Department of Pathology, Fujian Provincial Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Hejun Zhang
- Department of Pathology, Fujian Provincial Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Chao Li
- Department of Pathology, Fujian Provincial Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Gang Chen
- Department of Pathology, Fujian Provincial Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Xiong-wei Zheng
- Department of Pathology, Fujian Provincial Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
| | - Xiandong Lin
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
| |
Collapse
|
34
|
Chen J, Dou Y, Zheng X, Leng T, Lu X, Ouyang Y, Sun H, Xing F, Mai J, Gu J, Lu B, Yan G, Lin J, Zhu W. TRPM7 channel inhibition mediates midazolam-induced proliferation loss in human malignant glioma. Tumour Biol 2016; 37:14721-14731. [PMID: 27629139 DOI: 10.1007/s13277-016-5317-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/05/2016] [Indexed: 01/09/2023] Open
Abstract
The melastatin-like transient receptor potential 7 (TRPM7) has been implicated in proliferation or apoptosis of some cancers, indicating the potential of TRPM7 as an anti-anaplastic target. Here, we identified the characteristic TRPM7 channel currents in human malignant glioma MGR2 cells, which could be blocked by a pharmacologic inhibitor Gd3+. We mined the clinical sample data from Oncomine Database and found that human malignant glioma tissues expressed higher TRPM7 mRNA than normal brain ones. Importantly, we identified a widely used clinical anesthetic midazolam as a TRPM7 inhibitor. Midazolam treatment for seconds suppressed the TRPM7 currents and calcium influx, and treatment for 48 h inhibited the TRPM7 expression. The inhibitory effect on TRPM7 accounts for the proliferation loss and G0/G1 phase cell cycle arrest induced by midazolam. Our data demonstrates that midazolam represses proliferation of human malignant glioma cells through inhibiting TRPM7 currents, which may be further potentiated by suppressing the expression of TRPM7. Our result indicates midazolam as a pharmacologic lead compound with brain-blood barrier permeability for targeting TRPM7 in the glioma.
Collapse
Affiliation(s)
- Jingkao Chen
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China.,School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Yunling Dou
- Department of Anesthesiology, Department of Pathology and Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Xiaoke Zheng
- Department of Anesthesiology, Department of Pathology and Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Tiandong Leng
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, 30329, USA
| | - Xiaofang Lu
- Department of Anesthesiology, Department of Pathology and Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Ying Ouyang
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Huawei Sun
- Department of Anesthesiology, Department of Pathology and Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Fan Xing
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Jialuo Mai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Jiayu Gu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Bingzheng Lu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China.,South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center and Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Jun Lin
- Department of Anesthesiology, Stony Brook University Health Science Center, Stony Brook, NY, 11794-8480, USA.
| | - Wenbo Zhu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China. .,South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center and Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.
| |
Collapse
|
35
|
Nakano Y, Le MH, Abduweli D, Ho SP, Ryazanova LV, Hu Z, Ryazanov AG, Den Besten PK, Zhang Y. A Critical Role of TRPM7 As an Ion Channel Protein in Mediating the Mineralization of the Craniofacial Hard Tissues. Front Physiol 2016; 7:258. [PMID: 27458382 PMCID: PMC4934143 DOI: 10.3389/fphys.2016.00258] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/13/2016] [Indexed: 01/24/2023] Open
Abstract
Magnesium ion (Mg(2+)) is the fourth most common cation in the human body, and has a crucial role in many physiological functions. Mg(2+) homeostasis is an important contributor to bone development, however, its roles in the development of dental mineralized tissues have not yet been well known. We identified that transient receptor potential cation channel, subfamily M, member 7 (TRPM7), was significantly upregulated in the mature ameloblasts as compared to other ameloblasts through our whole transcript microarray analyses of the ameloblasts. TRPM7, an ion channel for divalent metal cations with an intrinsic serine/threonine protein kinase activity, has been characterized as a key regulator of whole body Mg(2+) homeostasis. Semi-quantitative PCR and immunostaining for TRMP7 confirmed its upregulation during the maturation stage of enamel formation, at which ameloblasts direct rapid mineralization of the enamel matrix. The significantly hypomineralized craniofacial structures, including incisors, molars, and cranial bones were demonstrated by microCT analysis, von Kossa and trichrome staining in Trpm7 (Δkinase∕+) mice. A previously generated heterozygous mouse model with the deletion of the TRPM7 kinase domain. Interestingly, the skeletal phenotype of Trpm7 (Δkinase∕+) mice resembled those found in the tissue-nonspecific alkaline phosphatase (Alpl) KO mice, thus we further examined whether ALPL protein content and alkaline phosphatase (ALPase) activity in ameloblasts, odontoblasts and osteoblasts were affected in those mice. While ALPL protein in Trpm7 (Δkinase∕+) mice remained at the similar level as that in wt mice, ALPase activities in the Trpm7 (Δkinase∕+) mice were almost nonexistent. Supplemented magnesium successfully rescued the activities of ALPase in ameloblasts, odontoblasts and osteoblasts of Trpm7 (Δkinase∕+) mice. These results suggested that TRPM7 is essential for mineralization of enamel as well as dentin and bone by providing sufficient Mg(2+) for the ALPL activity, underlining the key importance of ALPL for biomineralization.
Collapse
Affiliation(s)
- Yukiko Nakano
- Department of Orofacial Sciences, University of California, San FranciscoSan Francisco, CA, USA; Center for Children's Oral Health Research, University of California, San FranciscoSan Francisco, CA, USA
| | - Michael H Le
- Department of Orofacial Sciences, University of California, San Francisco San Francisco, CA, USA
| | - Dawud Abduweli
- Department of Orofacial Sciences, University of California, San Francisco San Francisco, CA, USA
| | - Sunita P Ho
- Preventive and Restorative Dental Sciences, University of California, San Francisco San Francisco, CA, USA
| | - Lillia V Ryazanova
- Department of Pharmacology, Robert Wood Johnson Medical School Piscataway, NJ, USA
| | - Zhixian Hu
- Department of Pharmacology, Robert Wood Johnson Medical School Piscataway, NJ, USA
| | - Alexey G Ryazanov
- Department of Pharmacology, Robert Wood Johnson Medical School Piscataway, NJ, USA
| | - Pamela K Den Besten
- Department of Orofacial Sciences, University of California, San FranciscoSan Francisco, CA, USA; Center for Children's Oral Health Research, University of California, San FranciscoSan Francisco, CA, USA
| | - Yan Zhang
- Department of Orofacial Sciences, University of California, San FranciscoSan Francisco, CA, USA; Center for Children's Oral Health Research, University of California, San FranciscoSan Francisco, CA, USA
| |
Collapse
|
36
|
LIN XIAOLING, YANG CHENG, HUANG LINJIE, CHEN MING, SHI JIANTING, OUYANG LIHUA, TANG TIANTIAN, ZHANG WEI, LI YIQUN, LIANG RUIYUN, JIANG SHANPING. Upregulation of TRPM7 augments cell proliferation and interleukin-8 release in airway smooth muscle cells of rats exposed to cigarette smoke. Mol Med Rep 2016; 13:4995-5004. [PMID: 27108806 PMCID: PMC4878570 DOI: 10.3892/mmr.2016.5161] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 03/10/2016] [Indexed: 12/26/2022] Open
Abstract
Proliferation and synthetic function (i.e. the capacity to release numerous chemokines and cytokines) of airway smooth muscle cells (ASMCs) are important in airway remodeling induced by cigarette smoke exposure. However, the molecular mechanism has not been clarified. Transient receptor potential cation channel subfamily M member 7 (TRPM7) is expressed ubiquitously and is crucial for the cellular physiological function of many cell types. The present study aimed to detect the expression of TRPM7 in ASMCs from smoke‑exposed rats and determine the importance of TRPM7 in proliferation and interleukin‑8 (IL‑8) release. ASMCs were isolated and cultured from smoke‑exposed rats. Expression levels of TRPM7 were determined by reverse transcription‑polymerase chain reaction, western blot analysis and immunofluorescence. TRPM7 was silenced with TRPM7‑short hairpin RNA lentivirus vector. DNA synthesis, cell number and IL‑8 release of ASMCs induced by cigarette smoke extract (CSE) and tumor necrosis factor‑α (TNF‑α) were assessed using [3H]-thymidine incorporation assay, hemocytometer and enzyme‑linked immunosorbent assay, respectively. It was determined that mRNA and protein expression levels of TRPM7 were increased in ASMCs from smoke‑exposed rats. Stimulation with CSE or TNF‑α elevated DNA synthesis, cell number and IL‑8 release were more marked in ASMCs from smoke‑exposed rats. Silencing of TRPM7 reduced DNA synthesis, cell number and IL‑8 release induced by CSE or TNF‑α in ASMCs from smoke-exposed rats. In conclusion, expression of TRPM7 increased significantly in ASMCs from smoke‑exposed rats and the upregulation of TRPM7 led to augmented cell proliferation and IL-8 release in ASMCs from rats exposed to cigarette smoke.
Collapse
Affiliation(s)
- XIAOLING LIN
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - CHENG YANG
- Department of Respiratory Medicine, Meizhou People's Hospital, Meizhou Affiliated Hospital of Sun Yat-Sen University, Meizhou, Guangdong 514031, P.R. China
| | - LINJIE HUANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - MING CHEN
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - JIANTING SHI
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - LIHUA OUYANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - TIANTIAN TANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - WEI ZHANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - YIQUN LI
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - RUIYUN LIANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - SHANPING JIANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
37
|
Chen WL, Barszczyk A, Turlova E, Deurloo M, Liu B, Yang BB, Rutka JT, Feng ZP, Sun HS. Inhibition of TRPM7 by carvacrol suppresses glioblastoma cell proliferation, migration and invasion. Oncotarget 2016; 6:16321-40. [PMID: 25965832 PMCID: PMC4599272 DOI: 10.18632/oncotarget.3872] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 04/02/2015] [Indexed: 01/27/2023] Open
Abstract
Glioblastomas are progressive brain tumors with devastating proliferative and invasive characteristics. Ion channels are the second largest target class for drug development. In this study, we investigated the effects of the TRPM7 inhibitor carvacrol on the viability, resistance to apoptosis, migration, and invasiveness of the human U87 glioblastoma cell line. The expression levels of TRPM7 mRNA and protein in U87 cells were detected by RT-PCR, western blotting and immunofluorescence. TRPM7 currents were recorded using whole-cell patch-clamp techniques. An MTT assay was used to assess cell viability and proliferation. Wound healing and transwell experiments were used to evaluate cell migration and invasion. Protein levels of p-Akt/t-Akt, p-ERK1/2/t-ERK1/2, cleaved caspase-3, MMP-2 and phosphorylated cofilin were also detected. TRPM7 mRNA and protein expression in U87 cells is higher than in normal human astrocytes. Whole-cell patch-clamp recording showed that carvacrol blocks recombinant TRPM7 current in HEK293 cells and endogenous TRPM7-like current in U87 cells. Carvacrol treatment reduced the viability, migration and invasion of U87 cells. Carvacrol also decreased MMP-2 protein expression and promoted the phosphorylation of cofilin. Furthermore, carvacrol inhibited the Ras/MEK/MAPK and PI3K/Akt signaling pathways. Therefore, carvacrol may have therapeutic potential for the treatment of glioblastomas through its inhibition of TRPM7 channels.
Collapse
Affiliation(s)
- Wen-Liang Chen
- Department of Surgery, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - Andrew Barszczyk
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Ekaterina Turlova
- Department of Surgery, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - Marielle Deurloo
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Baosong Liu
- Department of Surgery, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - Burton B Yang
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - James T Rutka
- Department of Surgery, University of Toronto, Toronto, Canada
| | - Zhong-Ping Feng
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Hong-Shuo Sun
- Department of Surgery, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada.,Department of Pharmacology, University of Toronto, Toronto, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
38
|
Middelbeek J, Visser D, Henneman L, Kamermans A, Kuipers AJ, Hoogerbrugge PM, Jalink K, van Leeuwen FN. TRPM7 maintains progenitor-like features of neuroblastoma cells: implications for metastasis formation. Oncotarget 2016; 6:8760-76. [PMID: 25797249 PMCID: PMC4496182 DOI: 10.18632/oncotarget.3315] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/08/2015] [Indexed: 12/18/2022] Open
Abstract
Neuroblastoma is an embryonal tumor derived from poorly differentiated neural crest cells. Current research is aimed at identifying the molecular mechanisms that maintain the progenitor state of neuroblastoma cells and to develop novel therapeutic strategies that induce neuroblastoma cell differentiation. Mechanisms controlling neural crest development are typically dysregulated during neuroblastoma progression, and provide an appealing starting point for drug target discovery. Transcriptional programs involved in neural crest development act as a context dependent gene regulatory network. In addition to BMP, Wnt and Notch signaling, activation of developmental gene expression programs depends on the physical characteristics of the tissue microenvironment. TRPM7, a mechanically regulated TRP channel with kinase activity, was previously found essential for embryogenesis and the maintenance of undifferentiated neural crest progenitors. Hence, we hypothesized that TRPM7 may preserve progenitor-like, metastatic features of neuroblastoma cells. Using multiple neuroblastoma cell models, we demonstrate that TRPM7 expression closely associates with the migratory and metastatic properties of neuroblastoma cells in vitro and in vivo. Moreover, microarray-based expression profiling on control and TRPM7 shRNA transduced neuroblastoma cells indicates that TRPM7 controls a developmental transcriptional program involving the transcription factor SNAI2. Overall, our data indicate that TRPM7 contributes to neuroblastoma progression by maintaining progenitor-like features.
Collapse
Affiliation(s)
- Jeroen Middelbeek
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Daan Visser
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Linda Henneman
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Arthur J Kuipers
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Peter M Hoogerbrugge
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Princes Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Kees Jalink
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frank N van Leeuwen
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
39
|
Yee NS. TRPM8 Ion Channels as Potential Cancer Biomarker and Target in Pancreatic Cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016:127-155. [DOI: 10.1016/bs.apcsb.2016.01.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
40
|
Liu C, Niu Y, Zhou X, Xu X, Yang Y, Zhang Y, Zheng L. Cell cycle control, DNA damage repair, and apoptosis-related pathways control pre-ameloblasts differentiation during tooth development. BMC Genomics 2015; 16:592. [PMID: 26265206 PMCID: PMC4534026 DOI: 10.1186/s12864-015-1783-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 07/16/2015] [Indexed: 02/05/2023] Open
Abstract
Background Ameloblast differentiation is the most critical stepwise process in amelogenesis, and it is controlled by precise molecular events. To better understand the mechanism controlling pre-ameloblasts (PABs) differentiation into secretory ameloblasts (SABs), a more precise identification of molecules and signaling networks will elucidate the mechanisms governing enamel formation and lay a foundation for enamel regeneration. Results We analyzed transcriptional profiles of human PABs and SABs. From a total of 28,869 analyzed transcripts, we identified 923 differentially expressed genes (DEGs) with p < 0.05 and Fold-change > 2. Among the DEGs, 647 genes showed elevated expression in PABs compared to SABs. Notably, 38 DEGs displayed greater than eight-fold changes. Comparative analysis revealed that highly expressed genes in PABs were involved in cell cycle control, DNA damage repair and apoptosis, while highly expressed genes in SABs were related to cell adhesion and extracellular matrix. Moreover, coexpression network analysis uncovered two highly conserved sub-networks contributing to differentiation, containing transcription regulators (RUNX2, ETV1 and ETV5), solute carrier family members (SLC15A1 and SLC7A11), enamel matrix protein (MMP20), and a polymodal excitatory ion channel (TRPA1). Conclusions By combining comparative analysis and coexpression networks, this study provides novel biomarkers and research targets for ameloblast differentiation and the potential for their application in enamel regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1783-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chengcheng Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China.
| | - Yulong Niu
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, PR China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China.
| | - Xin Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China.
| | - Yi Yang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, PR China.
| | - Yan Zhang
- Department of Orofacial Sciences, University of California, San Francisco, CA, 94143, USA.
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China.
| |
Collapse
|
41
|
Xyloketal B suppresses glioblastoma cell proliferation and migration in vitro through inhibiting TRPM7-regulated PI3K/Akt and MEK/ERK signaling pathways. Mar Drugs 2015; 13:2505-25. [PMID: 25913706 PMCID: PMC4413223 DOI: 10.3390/md13042505] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/03/2015] [Accepted: 04/08/2015] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma, the most common and aggressive type of brain tumors, has devastatingly proliferative and invasive characteristics. The need for finding a novel and specific drug target is urgent as the current approaches have limited therapeutic effects in treating glioblastoma. Xyloketal B is a marine compound obtained from mangrove fungus Xylaria sp. (No. 2508) from the South China Sea, and has displayed antioxidant activity and protective effects on endothelial and neuronal oxidative injuries. In this study, we used a glioblastoma U251 cell line to (1) explore the effects of xyloketal B on cell viability, proliferation, and migration; and (2) investigate the underlying molecular mechanisms and signaling pathways. MTT assay, colony formation, wound healing, western blot, and patch clamp techniques were employed. We found that xyloketal B reduced cell viability, proliferation, and migration of U251 cells. In addition, xyloketal B decreased p-Akt and p-ERK1/2 protein expressions. Furthermore, xyloketal B blocked TRPM7 currents in HEK-293 cells overexpressing TRPM7. These effects were confirmed by using a TRPM7 inhibitor, carvacrol, in a parallel experiment. Our findings indicate that TRPM7-regulated PI3K/Akt and MEK/ERK signaling is involved in anti-proliferation and migration effects of xyloketal B on U251 cells, providing in vitro evidence for the marine compound xyloketal B to be a potential drug for treating glioblastoma.
Collapse
|
42
|
Yee NS, Kazi AA, Li Q, Yang Z, Berg A, Yee RK. Aberrant over-expression of TRPM7 ion channels in pancreatic cancer: required for cancer cell invasion and implicated in tumor growth and metastasis. Biol Open 2015; 4:507-514. [PMID: 25770184 PMCID: PMC4400593 DOI: 10.1242/bio.20137088] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 12/13/2014] [Indexed: 01/12/2023] Open
Abstract
Our previous studies in zebrafish development have led to identification of the novel roles of the transient receptor potential melastatin-subfamily member 7 (TRPM7) ion channels in human pancreatic cancer. However, the biological significance of TRPM7 channels in pancreatic neoplasms was mostly unexplored. In this study, we determined the expression levels of TRPM7 in pancreatic tissue microarrays and correlated these measurements in pancreatic adenocarcinoma with the clinicopathological features. We also investigated the role of TRPM7 channels in pancreatic cancer cell invasion using the Matrigel(TM)-coated transwell assay. In normal pancreas, TRPM7 is expressed at a discernable level in the ductal cells and centroacinar cells and at a relatively high level in the islet endocrine cells. In chronic pancreatitis, pre-malignant tissues, and malignant neoplasms, there is variable expression of TRPM7. In the majority of pancreatic adenocarcinoma specimens examined, TRPM7 is expressed at either moderate-level or high-level. Anti-TRPM7 immunoreactivity in pancreatic adenocarcinoma significantly correlates with the size and stages of tumors. In human pancreatic adenocarcinoma cells in which TRPM7 is highly expressed, short hairpin RNA-mediated suppression of TRPM7 impairs cell invasion. The results demonstrate that TRPM7 channels are over-expressed in a proportion of the pre-malignant lesions and malignant tumors of the pancreas, and they are necessary for invasion by pancreatic cancer cells. We propose that TRPM7 channels play important roles in development and progression of pancreatic neoplasm, and they may be explored as clinical biomarkers and targets for its prevention and treatment.
Collapse
Affiliation(s)
- Nelson S Yee
- Division of Hematology-Oncology, Department of Medicine, Penn State College of Medicine, Program of Experimental Therapeutics, Penn State Hershey Cancer Institute, Penn State Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, PA 17033, USA
| | - Abid A Kazi
- Division of Hematology-Oncology, Department of Medicine, Penn State College of Medicine, Program of Experimental Therapeutics, Penn State Hershey Cancer Institute, Penn State Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, PA 17033, USA
| | - Qin Li
- Division of Hematology-Oncology, Department of Medicine, Penn State College of Medicine, Program of Experimental Therapeutics, Penn State Hershey Cancer Institute, Penn State Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, PA 17033, USA
| | - Zhaohai Yang
- Division of Anatomic Pathology, Department of Pathology, Penn State College of Medicine, Penn State Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, PA 17033, USA
| | - Arthur Berg
- Division of Biostatistics and Bioinformatics, Department of Public Health, Penn State College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Rosemary K Yee
- Schreyer Honors College, Pennsylvania State University, University Park, PA 16802, USA, Penn State Harrisburg School of Humanities, Pennsylvania State University, Middletown, PA 17057, USA
| |
Collapse
|
43
|
Wang J, Liao QJ, Zhang Y, Zhou H, Luo CH, Tang J, Wang Y, Tang Y, Zhao M, Zhao XH, Zhang QY, Xiao L. TRPM7 is required for ovarian cancer cell growth, migration and invasion. Biochem Biophys Res Commun 2014; 454:547-53. [DOI: 10.1016/j.bbrc.2014.10.118] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 10/24/2014] [Indexed: 02/02/2023]
|