101
|
Stein P, Savy V, Williams AM, Williams CJ. Modulators of calcium signalling at fertilization. Open Biol 2020; 10:200118. [PMID: 32673518 PMCID: PMC7574550 DOI: 10.1098/rsob.200118] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
Calcium (Ca2+) signals initiate egg activation across the animal kingdom and in at least some plants. These signals are crucial for the success of development and, in the case of mammals, health of the offspring. The mechanisms associated with fertilization that trigger these signals and the molecules that regulate their characteristic patterns vary widely. With few exceptions, a major contributor to fertilization-induced elevation in cytoplasmic Ca2+ is release from endoplasmic reticulum stores through the IP3 receptor. In some cases, Ca2+ influx from the extracellular space and/or release from alternative intracellular stores contribute to the rise in cytoplasmic Ca2+. Following the Ca2+ rise, the reuptake of Ca2+ into intracellular stores or efflux of Ca2+ out of the egg drive the return of cytoplasmic Ca2+ back to baseline levels. The molecular mediators of these Ca2+ fluxes in different organisms include Ca2+ release channels, uptake channels, exchangers and pumps. The functions of these mediators are regulated by their particular activating mechanisms but also by alterations in their expression and spatial organization. We discuss here the molecular basis for modulation of Ca2+ signalling at fertilization, highlighting differences across several animal phyla, and we mention key areas where questions remain.
Collapse
Affiliation(s)
- Paula Stein
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Virginia Savy
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Audrey M. Williams
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Carmen J. Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
102
|
Song C, Choi S, Oh KB, Sim T. Suppression of TRPM7 enhances TRAIL-induced apoptosis in triple-negative breast cancer cells. J Cell Physiol 2020; 235:10037-10050. [PMID: 32468675 DOI: 10.1002/jcp.29820] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 05/03/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022]
Abstract
Transient receptor potential cation channel subfamily M member 7 (TRPM7) composed of an ion channel and a kinase domain regulates triple-negative breast cancer (TNBC) cell migration, invasion, and metastasis, but it does not modulate TNBC proliferation. However, previous studies have shown that the combination treatment of nonselective TRPM7 channel inhibitors (2-aminoethoxydiphenyl borate and Gd3+ ) with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) increases antiproliferative effects and apoptosis in prostate cancer cells and hepatic stellate cells. We, therefore, investigated the potential role of TRPM7 in proliferation and apoptosis of TNBC cells (MDA-MB-231 and MDA-MB-468 cells) with TRAIL. We demonstrated that suppression of TRPM7 via TRPM7 knockdown or pharmacological inhibition synergistically increases TRAIL-induced antiproliferative effects and apoptosis in TNBC cells. Furthermore, we showed that the synergistic interaction might be associated with TRPM7 channel activities using combination treatments of TRAIL and TRPM7 inhibitors (NS8593 as a TRPM7 channel inhibitor and TG100-115 as a TRPM7 kinase inhibitor). We reveal that downregulation of cellular FLICE-inhibitory protein via inhibition of Ca2+ influx might be involved in the synergistic interaction. Our study would provide both a new role of TRPM7 in TNBC cell apoptosis and a potential combinatorial therapeutic strategy using TRPM7 inhibitors with TRAIL in the treatment of TNBC.
Collapse
Affiliation(s)
- Chiman Song
- Chemical Kinomics Research Center, Korea Institute of Science and Technology Seongbuk-gu, Seoul, Republic of Korea.,Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| | - Seunghye Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
| | - Ki-Bong Oh
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| | - Taebo Sim
- Chemical Kinomics Research Center, Korea Institute of Science and Technology Seongbuk-gu, Seoul, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seodaemun-gu, Seoul, Republic of Korea
| |
Collapse
|
103
|
Steroids and TRP Channels: A Close Relationship. Int J Mol Sci 2020; 21:ijms21113819. [PMID: 32471309 PMCID: PMC7325571 DOI: 10.3390/ijms21113819] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential (TRP) channels are remarkable transmembrane protein complexes that are essential for the physiology of the tissues in which they are expressed. They function as non-selective cation channels allowing for the signal transduction of several chemical, physical and thermal stimuli and modifying cell function. These channels play pivotal roles in the nervous and reproductive systems, kidney, pancreas, lung, bone, intestine, among others. TRP channels are finely modulated by different mechanisms: regulation of their function and/or by control of their expression or cellular/subcellular localization. These mechanisms are subject to being affected by several endogenously-produced compounds, some of which are of a lipidic nature such as steroids. Fascinatingly, steroids and TRP channels closely interplay to modulate several physiological events. Certain TRP channels are affected by the typical genomic long-term effects of steroids but others are also targets for non-genomic actions of some steroids that act as direct ligands of these receptors, as will be reviewed here.
Collapse
|
104
|
Manialawy Y, Khan SR, Bhattacharjee A, Wheeler MB. The magnesium transporter NIPAL1 is a pancreatic islet-expressed protein that conditionally impacts insulin secretion. J Biol Chem 2020; 295:9879-9892. [PMID: 32439805 DOI: 10.1074/jbc.ra120.013277] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/18/2020] [Indexed: 01/23/2023] Open
Abstract
Type 2 diabetes is a chronic metabolic disease characterized by pancreatic β-cell dysfunction and peripheral insulin resistance. Among individuals with type 2 diabetes, ∼30% exhibit hypomagnesemia. Hypomagnesemia has been linked to insulin resistance through reduced tyrosine kinase activity of the insulin receptor; however, its impact on pancreatic β-cell function is unknown. In this study, through analysis of several single-cell RNA-sequencing data sets in tandem with quantitative PCR validation in both murine and human islets, we identified NIPAL1 (NIPA-like domain containing 1), encoding a magnesium influx transporter, as an islet-enriched gene. A series of immunofluorescence experiments confirmed NIPAL1's magnesium-dependent expression and that it specifically localizes to the Golgi in Min6-K8 cells, a pancreatic β-cell-like cell line (mouse insulinoma 6 clone K8). Under varying magnesium concentrations, NIPAL1 knockdown decreased both basal insulin secretion and total insulin content; in contrast, its overexpression increased total insulin content. Although the expression, distribution, and magnesium responsiveness of NIPAL1 in α-TC6 glucagonoma cells (a pancreatic α-cell line) were similar to the observations in Min6-K8 cells, no effect was observed on glucagon secretion in α-TC6 cells under the conditions studied. Overall, these results suggest that NIPAL1 expression is regulated by extracellular magnesium and that down-regulation of this transporter decreases glucose-stimulated insulin secretion and intracellular insulin content, particularly under conditions of hypomagnesemia.
Collapse
Affiliation(s)
- Yousef Manialawy
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Saifur R Khan
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada .,Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Alpana Bhattacharjee
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Michael B Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada .,Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
105
|
Turlova E, Wong R, Xu B, Li F, Du L, Habbous S, Horgen FD, Fleig A, Feng ZP, Sun HS. TRPM7 Mediates Neuronal Cell Death Upstream of Calcium/Calmodulin-Dependent Protein Kinase II and Calcineurin Mechanism in Neonatal Hypoxic-Ischemic Brain Injury. Transl Stroke Res 2020; 12:164-184. [PMID: 32430797 DOI: 10.1007/s12975-020-00810-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/12/2020] [Accepted: 03/18/2020] [Indexed: 11/25/2022]
Abstract
Transient receptor potential melastatin 7 (TRPM7), a calcium-permeable, ubiquitously expressed ion channel, is critical for axonal development, and mediates hypoxic and ischemic neuronal cell death in vitro and in vivo. However, the downstream mechanisms underlying the TRPM7-mediated processes in physiology and pathophysiology remain unclear. In this study, we employed a mouse model of hypoxic-ischemic brain cell death which mimics the pathophysiology of hypoxic-ischemic encephalopathy (HIE). HIE is a major public health issue and an important cause of neonatal deaths worldwide; however, the available treatments for HIE remain limited. Its survivors face life-long neurological challenges including mental retardation, cerebral palsy, epilepsy and seizure disorders, motor impairments, and visual and auditory impairments. Through a proteomic analysis, we identified calcium/calmodulin-dependent protein kinase II (CaMKII) and phosphatase calcineurin as potential mediators of cell death downstream from TRPM7 activation. Further analysis revealed that TRPM7 mediates cell death through CaMKII, calmodulin, calcineurin, p38, and cofilin cascade. In vivo, we found a significant reduction of brain injury and improvement of short- and long-term functional outcomes after HI after administration of specific TRPM7 blocker waixenicin A. Our data demonstrate a molecular mechanism of TRPM7-mediated cell death and identifies TRPM7 as a promising therapeutic and drug development target for HIE.
Collapse
Affiliation(s)
- Ekaterina Turlova
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Raymond Wong
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Baofeng Xu
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Feiya Li
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Lida Du
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Steven Habbous
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI, 96744, USA
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96720, USA
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
| | - Hong-Shuo Sun
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
- Department of Pharmacology, University of Toronto, 1 King's College Circle, Toronto, M5S 1A8, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, University of Toronto, Toronto, Canada.
| |
Collapse
|
106
|
López-Romero AE, Hernández-Araiza I, Torres-Quiroz F, Tovar-Y-Romo LB, Islas LD, Rosenbaum T. TRP ion channels: Proteins with conformational flexibility. Channels (Austin) 2020; 13:207-226. [PMID: 31184289 PMCID: PMC6602575 DOI: 10.1080/19336950.2019.1626793] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Ion channels display conformational changes in response to binding of their agonists and antagonists. The study of the relationships between the structure and the function of these proteins has witnessed considerable advances in the last two decades using a combination of techniques, which include electrophysiology, optical approaches (i.e. patch clamp fluorometry, incorporation of non-canonic amino acids, etc.), molecular biology (mutations in different regions of ion channels to determine their role in function) and those that have permitted the resolution of their structures in detail (X-ray crystallography and cryo-electron microscopy). The possibility of making correlations among structural components and functional traits in ion channels has allowed for more refined conclusions on how these proteins work at the molecular level. With the cloning and description of the family of Transient Receptor Potential (TRP) channels, our understanding of several sensory-related processes has also greatly moved forward. The response of these proteins to several agonists, their regulation by signaling pathways as well as by protein-protein and lipid-protein interactions and, in some cases, their biophysical characteristics have been studied thoroughly and, recently, with the resolution of their structures, the field has experienced a new boom. This review article focuses on the conformational changes in the pores, concentrating on some members of the TRP family of ion channels (TRPV and TRPA subfamilies) that result in changes in their single-channel conductances, a phenomenon that may lead to fine-tuning the electrical response to a given agonist in a cell.
Collapse
Affiliation(s)
- Ana Elena López-Romero
- a Departamento de Neurociencia Cognitiva, División Neurociencias , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico , Mexico
| | - Ileana Hernández-Araiza
- a Departamento de Neurociencia Cognitiva, División Neurociencias , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico , Mexico
| | - Francisco Torres-Quiroz
- b Departamento de Bioquímica y Biología Estructural, División Investigación Básica , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - Luis B Tovar-Y-Romo
- c Departamento de Neuropatología Molecular, División Neurociencias , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - León D Islas
- d Departamento de Fisiología, Facultad de Medicina , Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - Tamara Rosenbaum
- a Departamento de Neurociencia Cognitiva, División Neurociencias , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico , Mexico
| |
Collapse
|
107
|
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
|
108
|
Affiliation(s)
- Valentina Trapani
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Federica I Wolf
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
| |
Collapse
|
109
|
Study of Magnesium Formulations on Intestinal Cells to Influence Myometrium Cell Relaxation. Nutrients 2020; 12:nu12020573. [PMID: 32098378 PMCID: PMC7071389 DOI: 10.3390/nu12020573] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/15/2020] [Accepted: 02/17/2020] [Indexed: 12/31/2022] Open
Abstract
Background: Magnesium is involved in a wide variety of physiological processes including direct relaxation of smooth muscle. A magnesium imbalance can be considered the primary cause or consequence of many pathophysiological conditions. The smooth muscle tissue of the uterus, i.e., the myometrium, undergoes numerous physiological changes during life, fundamental for uterine activities, and it receives proven benefits from magnesium supplementation. However, magnesium supplements have poor absorption and bioavailability. Furthermore, no data are available on the direct interaction between intestinal absorption of magnesium and relaxation of the myometrium. Methods: Permeability in human intestinal cells (Caco-2 cells) and direct effects on myometrial cells (PHM1-41 cells) of two different forms of magnesium, i.e., sucrosomial and bisglycinate, were studied in order to verify the magnesium capacity of modulate contractility. Cell viability, reactive oxygen species (ROS) and nitric oxide (NO) production, magnesium concentration, contractility, and pathways involved were analyzed. Results: Data showed a better influence of buffered chelate bisglycinate on intestinal permeability and myometrial relaxation over time with a maximum effect at 3 h and greater availability compared to the sucrosomial form. Conclusions: Magnesium-buffered bisglycinate chelate showed better intestinal absorption and myometrial contraction, indicating a better chance of effectiveness in human applications.
Collapse
|
110
|
Liu Y, Chen C, Liu Y, Li W, Wang Z, Sun Q, Zhou H, Chen X, Yu Y, Wang Y, Abumaria N. TRPM7 Is Required for Normal Synapse Density, Learning, and Memory at Different Developmental Stages. Cell Rep 2019; 23:3480-3491. [PMID: 29924992 DOI: 10.1016/j.celrep.2018.05.069] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 01/29/2018] [Accepted: 05/18/2018] [Indexed: 10/28/2022] Open
Abstract
The TRPM7 chanzyme contributes to several biological and pathological processes in different tissues. However, its role in the CNS under physiological conditions remains unclear. Here, we show that TRPM7 knockdown in hippocampal neurons reduces structural synapse density. The synapse density is rescued by the α-kinase domain in the C terminus but not by the ion channel region of TRPM7 or by increasing extracellular concentrations of Mg2+ or Zn2+. Early postnatal conditional knockout of TRPM7 in mice impairs learning and memory and reduces synapse density and plasticity. TRPM7 knockdown in the hippocampus of adult rats also impairs learning and memory and reduces synapse density and synaptic plasticity. In knockout mice, restoring expression of the α-kinase domain in the brain rescues synapse density/plasticity and memory, probably by interacting with and phosphorylating cofilin. These results suggest that brain TRPM7 is important for having normal synaptic and cognitive functions under physiological, non-pathological conditions.
Collapse
Affiliation(s)
- Yuqiang Liu
- Department of Neurology, Huashan Hospital, and Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Cui Chen
- Department of Neurology, Huashan Hospital, and Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Yunlong Liu
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wei Li
- Department of Neurology, Huashan Hospital, and Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Zhihong Wang
- Department of Neurology, Huashan Hospital, and Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Qifeng Sun
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Hang Zhou
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital, and Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Yongchun Yu
- Department of Neurology, Huashan Hospital, and Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Yun Wang
- Department of Neurology, Huashan Hospital, and Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Nashat Abumaria
- Department of Neurology, Huashan Hospital, and Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Department of Laboratory Animal Science, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
111
|
Zhou S, Zheng WJ, Liu BH, Zheng JC, Dong FS, Liu ZF, Wen ZY, Yang F, Wang HB, Xu ZS, Zhao H, Liu YW. Characterizing the Role of TaWRKY13 in Salt Tolerance. Int J Mol Sci 2019; 20:ijms20225712. [PMID: 31739570 PMCID: PMC6888956 DOI: 10.3390/ijms20225712] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 11/09/2019] [Accepted: 11/11/2019] [Indexed: 01/09/2023] Open
Abstract
The WRKY transcription factor superfamily is known to participate in plant growth and stress response. However, the role of this family in wheat (Triticum aestivum L.) is largely unknown. Here, a salt-induced gene TaWRKY13 was identified in an RNA-Seq data set from salt-treated wheat. The results of RT-qPCR analysis showed that TaWRKY13 was significantly induced in NaCl-treated wheat and reached an expression level of about 22-fold of the untreated wheat. Then, a further functional identification was performed in both Arabidopsis thaliana and Oryza sativa L. Subcellular localization analysis indicated that TaWRKY13 is a nuclear-localized protein. Moreover, various stress-related regulatory elements were predicted in the promoter. Expression pattern analysis revealed that TaWRKY13 can also be induced by polyethylene glycol (PEG), exogenous abscisic acid (ABA), and cold stress. After NaCl treatment, overexpressed Arabidopsis lines of TaWRKY13 have a longer root and a larger root surface area than the control (Columbia-0). Furthermore, TaWRKY13 overexpression rice lines exhibited salt tolerance compared with the control, as evidenced by increased proline (Pro) and decreased malondialdehyde (MDA) contents under salt treatment. The roots of overexpression lines were also more developed. These results demonstrate that TaWRKY13 plays a positive role in salt stress.
Collapse
Affiliation(s)
- Shuo Zhou
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
| | - Wei-Jun Zheng
- College of Agronomy, Northwest A&F University, Yangling 712100, China;
| | - Bao-Hua Liu
- Handan Academy of Agricultural Sciences, Handan 056001, China;
| | - Jia-Cheng Zheng
- College of Agronomy, Anhui Science and Technology University, Fengyang, Chuzhou 239000, China;
| | - Fu-Shuang Dong
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
| | | | - Zhi-Yu Wen
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
| | - Fan Yang
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
| | - Hai-Bo Wang
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
| | - Zhao-Shi Xu
- Institute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)/National Key Facility for Crop Gene Resources and Genetic Improvement, Key Laboratory of Biology and Genetic Improvement of Triticeae Crops, Ministry of Agriculture, Beijing 100081, China;
| | - He Zhao
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
- Correspondence: (H.Z.); (Y.-W.L.)
| | - Yong-Wei Liu
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
- Correspondence: (H.Z.); (Y.-W.L.)
| |
Collapse
|
112
|
Feng J, Armillei MK, Yu AS, Liang BT, Runnels LW, Yue L. Ca 2+ Signaling in Cardiac Fibroblasts and Fibrosis-Associated Heart Diseases. J Cardiovasc Dev Dis 2019; 6:E34. [PMID: 31547577 PMCID: PMC6956282 DOI: 10.3390/jcdd6040034] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiac fibrosis is the excessive deposition of extracellular matrix proteins by cardiac fibroblasts and myofibroblasts, and is a hallmark feature of most heart diseases, including arrhythmia, hypertrophy, and heart failure. This maladaptive process occurs in response to a variety of stimuli, including myocardial injury, inflammation, and mechanical overload. There are multiple signaling pathways and various cell types that influence the fibrogenesis cascade. Fibroblasts and myofibroblasts are central effectors. Although it is clear that Ca2+ signaling plays a vital role in this pathological process, what contributes to Ca2+ signaling in fibroblasts and myofibroblasts is still not wholly understood, chiefly because of the large and diverse number of receptors, transporters, and ion channels that influence intracellular Ca2+ signaling. Intracellular Ca2+ signals are generated by Ca2+ release from intracellular Ca2+ stores and by Ca2+ entry through a multitude of Ca2+-permeable ion channels in the plasma membrane. Over the past decade, the transient receptor potential (TRP) channels have emerged as one of the most important families of ion channels mediating Ca2+ signaling in cardiac fibroblasts. TRP channels are a superfamily of non-voltage-gated, Ca2+-permeable non-selective cation channels. Their ability to respond to various stimulating cues makes TRP channels effective sensors of the many different pathophysiological events that stimulate cardiac fibrogenesis. This review focuses on the mechanisms of Ca2+ signaling in fibroblast differentiation and fibrosis-associated heart diseases and will highlight recent advances in the understanding of the roles that TRP and other Ca2+-permeable channels play in cardiac fibrosis.
Collapse
Affiliation(s)
- Jianlin Feng
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Maria K Armillei
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Albert S Yu
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Bruce T Liang
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Loren W Runnels
- Department of Pharmacology, Rutgers, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - Lixia Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| |
Collapse
|
113
|
Gatica S, Eltit F, Santibanez JF, Varela D, Cabello-Verrugio C, Simon F. Expression Suppression and Activity Inhibition of TRPM7 Regulate Cytokine Production and Multiple Organ Dysfunction Syndrome During Endotoxemia: a New Target for Sepsis. Curr Mol Med 2019; 19:547-559. [PMID: 31288723 DOI: 10.2174/1566524019666190709181726] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 05/24/2019] [Accepted: 05/28/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Main pathological features detected during sepsis and endotoxemia include over-secretion of pro-inflammatory cytokines and multiorgan dysfunction syndrome (MODS). Unfortunately, current clinical efforts to treat sepsis are unsatisfactory, and mortality remains high. Interestingly, transient receptor potential (TRP) melastatin 7 (TRPM7) ion channel controlling Ca2+ and Mg2+ permeability is involved in cytokine production and inflammatory response. Furthermore, TRPM7 downregulation has been shown to alleviate local symptoms in some models of sepsis, but its effects at a systemic level remain to be explored. OBJECTIVE To test whether TRPM7 mediates cytokine production and MODS during endotoxemia. METHODS Endotoxemic and sham-endotoxemic rats were subjected to pharmacological inhibition of TRPM7 using carvacrol, or to expression suppression by adenovirus delivery of shRNA (AdVshTRPM7). Then, cytokine and MODS levels in the blood were measured. RESULTS Inhibition of TRPM7 with carvacrol and suppression with AdVshTRPM7 were both efficient in inhibiting the over-secretion of pro-inflammatory cytokines TNF-α, IL-1β, IL-6, and IL-12, in endotoxemic rats, without inducing downregulation in blood levels of antiinflammatory cytokines IL-10 and IL-4. Additionally, the use of carvacrol and AdVshTRPM7 significantly prevented liver and pancreas dysfunction, altered metabolic function, and hypoglycemia, induced by endotoxemia. Furthermore, muscle mass wasting and cardiac muscle damage were also significantly reduced by the use of carvacrol and AdVshTRPM7 in endotoxemic rats. CONCLUSION Our results indicate TRPM7 ion channel as a key protein regulating inflammatory responses and MODS during sepsis. Moreover, TRPM7 appears as a novel molecular target for the management of sepsis.
Collapse
Affiliation(s)
- Sebastian Gatica
- Departamento de Ciencias Biologicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370146, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, 8331150, Santiago, Chile
| | - Felipe Eltit
- Department of Materials Engineering, University of British Columbia, Vancouver, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Juan F Santibanez
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgrade, Serbia.,Centro Integrativo de Biología y Química Aplicada (CIBQA). Universidad Bernardo O'Higgins, Santiago, Chile
| | - Diego Varela
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Chile, Chile
| | - Claudio Cabello-Verrugio
- Departamento de Ciencias Biologicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370146, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, 8331150, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Felipe Simon
- Departamento de Ciencias Biologicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370146, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, 8331150, Santiago, Chile
| |
Collapse
|
114
|
Abumaria N, Li W, Clarkson AN. Role of the chanzyme TRPM7 in the nervous system in health and disease. Cell Mol Life Sci 2019; 76:3301-3310. [PMID: 31073743 PMCID: PMC11105578 DOI: 10.1007/s00018-019-03124-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 12/13/2022]
Abstract
The channel kinase (chanzyme) transient receptor potential melastatin-like 7 (TRPM7) has a unique dual protein structure composed of an ion channel with an α-kinase domain on its C-terminus. In the nervous system, under physiological conditions, TRPM7 contributes to critical neurobiological processes ranging from synaptic transmission to cognitive functions. Following certain pathological triggers, TRPM7 mediates neurotoxicity, neuro-injuries, and neuronal death. Here, we summarize the current knowledge of TRPM7 functions in neuronal systems in health and disease. The molecular mechanisms by which this chanzyme might regulate synaptic and cognitive functions are discussed. We also discuss the lack of knowledge regarding the molecular mechanisms responsible for turning TRPM7 into "a vicious tool" that mediates neuronal death following certain pathological triggers. Some synthetic and natural pharmacological modulators of the TRPM7 channel and its α-kinase are reviewed. We suggest that based on current knowledge, we should reshape our thinking regarding the implications of TRPM7 in neurological and neurodegenerative disorders. Moreover, we propose a paradigm shift concerning the targeting of TRPM7 as a therapeutic approach for treating certain neurological diseases. We agree that TRPM7 overexpression or overactivation may mediate neurodegenerative processes following certain triggers. However, TRPM7 dysfunction and/or downregulation might also be among the pathological changes leading to neurodegeneration. Consequently, further investigations are required before we decide whether blocking or activating the chanzyme is the correct therapeutic approach to treat certain neurological and/or neurodegenerative diseases.
Collapse
Affiliation(s)
- Nashat Abumaria
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
- Department of Laboratory Animal Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Wei Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Andrew N Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, PO Box 913, Dunedin, 9054, New Zealand
| |
Collapse
|
115
|
Scarlata S. The role of phospholipase Cβ on the plasma membrane and in the cytosol: How modular domains enable novel functions. Adv Biol Regul 2019; 73:100636. [PMID: 31409535 DOI: 10.1016/j.jbior.2019.100636] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 07/14/2019] [Accepted: 07/25/2019] [Indexed: 01/14/2023]
Abstract
Phospholipase Cβ (PLCβ) is a signaling enzyme activated by G proteins to generate calcium signals. The catalytic core of PLCβ is surrounded by modular domains that mediate the interaction of the enzyme with known protein partners on the plasma membrane. The C-terminal region PLCβ contains a novel coiled-coil domain that is required for Gαq binding and activation. Recent work has shown that this domain also binds a number of cytosolic proteins that regulate protein translation, and that these proteins compete with Gαq for PLCβ binding. The ability of PLCβ to shuttle between the cytosol to impact protein translation and the plasma membrane to mediate calcium signals puts PLCβ in a central role in cell function.
Collapse
Affiliation(s)
- Suzanne Scarlata
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, 100 Institute Rd., Worcester, MA, 01609, United States.
| |
Collapse
|
116
|
Chen YJ, Cheng FC, Chen CJ, Su HL, Sheu ML, Sheehan J, Pan HC. Down-Regulated Expression of Magnesium Transporter Genes Following a High Magnesium Diet Attenuates Sciatic Nerve Crush Injury. Neurosurgery 2019; 84:965-976. [PMID: 29672725 DOI: 10.1093/neuros/nyy120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/10/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Magnesium supplementation has potential for use in nerve regeneration. The expression of some magnesium transporter genes is reflective of the intracellular magnesium levels. OBJECTIVE To assess the expression of various magnesium transporter genes as they relate to neurological alterations in a sciatic nerve injury model. METHODS Sciatic nerve injury was induced in rats, which were then fed either basal or high magnesium diets. Magnesium concentrations and 5 magnesium transporter genes (SLC41A1, MAGT1, CNNM2, TRPM6, and TRPM7) were measured in the tissue samples. RESULTS The high magnesium diet attenuated cytoskeletal loss in a dose-dependent manner in isolated nerve explants. The high magnesium diet augmented nerve regeneration and led to the restoration of nerve structure, increased S-100, and neurofilaments. This increased regeneration was consistent with the improvement of neurobehavioral and electrophysiological assessment. The denervated muscle morphology was restored with the high magnesium diet, and that was also highly correlated with the increased expression of desmin and acetylcholine receptors in denervated muscle. The plasma magnesium levels were significantly elevated after the animals consumed a high magnesium diet and were reciprocally related to the down-regulation of CNNM2, MagT1, and SCL41A1 in the blood monocytes, nerves, and muscle tissues of the nerve crush injury model. CONCLUSION The increased plasma magnesium levels after consuming a high magnesium diet were highly correlated with the down-regulation of magnesium transporter genes in monocytes, nerves, and muscle tissues after sciatic nerve crush injury. The study findings suggest that there are beneficial effects of administering magnesium after a nerve injury.
Collapse
Affiliation(s)
- Ying-Ju Chen
- Department of food and nutrition, Providence University, Taichung, Taiwan
| | - Fu-Chou Cheng
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, Agriculture Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan
| | - Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, Virginia
| | - Hung-Chuan Pan
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
117
|
Comparative sequence analysis of patient-matched primary colorectal cancer, metastatic, and recurrent metastatic tumors after adjuvant FOLFOX chemotherapy. BMC Cancer 2019; 19:255. [PMID: 30898102 PMCID: PMC6429751 DOI: 10.1186/s12885-019-5479-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/14/2019] [Indexed: 12/22/2022] Open
Abstract
Background In the era of genome-guided personalized cancer treatment, we must understand chemotherapy-induced genomic changes in tumors. This study evaluated whether adjuvant FOLFOX chemotherapy modifies the mutational profile of recurrent colorectal cancer (CRC). Methods Whole exome sequencing was performed on samples from primary CRC tumors, untreated metastatic tumors, and recurrent tumors following adjuvant FOLFOX chemotherapy. The samples were resected from four patients. Results The number of mutations or the mutation spectrum in individual patients was nearly identical. Copy number variants persisted regardless of FOLFOX therapy administration. The genomic signature of oxaliplatin exposure (G > T/C > A, T > A/A > T) was not enriched after FOLFOX chemotherapy. Overlapping single nucleotide variants (SNVs) and indels remained in 26–65% of the patient-matched tumor samples. One patient harbored an AKT1 E17K mutation in the recurrent tumor, whereas PIK3CA E542K and E88Q mutations were detected in the primary and untreated metastatic tumor samples. Genes related to intracellular Ca2+ homeostasis were enriched among the genes uniquely mutated after FOLFOX chemotherapy. Conclusions We found that the mutation rates, mutation spectrum, and copy number variants were nearly identical regardless of the administration of FOLFOX therapy in the four CRC cases. The mutational discordance between the patient-matched tumor samples is likely caused by tumor heterogeneity and chemotherapy-induced clonal selection. These findings might be useful as pilot data for larger studies to clarify the changes in the mutational landscape induced by adjuvant FOLFOX chemotherapy. Electronic supplementary material The online version of this article (10.1186/s12885-019-5479-6) contains supplementary material, which is available to authorized users.
Collapse
|
118
|
Kühn FJP, Watt JM, Potter BVL, Lückhoff A. Different substrate specificities of the two ADPR binding sites in TRPM2 channels of Nematostella vectensis and the role of IDPR. Sci Rep 2019; 9:4985. [PMID: 30899048 PMCID: PMC6428886 DOI: 10.1038/s41598-019-41531-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 03/11/2019] [Indexed: 01/13/2023] Open
Abstract
NvTRPM2 (Nematostella vectensis Transient Receptor Potential Melastatin 2), the species variant of the human apoptosis-related cation channel hTRPM2, is gated by ADP-ribose (ADPR) independently of the C-terminal NUDT9H domain that mediates ADPR-directed gating in hTRPM2. The decisive binding site in NvTRPM2 is likely to be identical with the N-terminal ADPR binding pocket in zebra fish DrTRPM2. Our aim was a characterization of this binding site in NvTRPM2 with respect to its substrate specificity, in comparison to the classical ADPR interaction site within NUDT9H that is highly homologous in hTRPM2 and NvTRPM2, although only in NvTRPM2, catalytic (ADPRase) activity is conserved. With various ADPR analogues, key differences of the two sites were identified. Particularly, two reported antagonists on hTRPM2 were agonists on NvTRPM2. Moreover, IDP-ribose (IDPR) induced currents both in hTRPM2 and NvTRPM2 but not in NvTRPM2 mutants in which NUDT9H was absent. Thus, IDPR acts on NUDT9H rather than N-terminally, revealing a regulatory function of NUDT9H in NvTRPM2 opposed to that in hTRPM2. We propose that IDPR competitively inhibits the ADPRase function of NUDT9H and evokes ADPR accumulation. The findings provide important insights into the structure-function relationship of NvTRPM2 and will allow further characterization of the novel ADPR interaction site.
Collapse
Affiliation(s)
- Frank J P Kühn
- Institute of Physiology, Medical Faculty, RWTH Aachen, D52057, Aachen, Germany.
| | - Joanna M Watt
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Barry V L Potter
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Andreas Lückhoff
- Institute of Physiology, Medical Faculty, RWTH Aachen, D52057, Aachen, Germany
| |
Collapse
|
119
|
TRPM7 is the central gatekeeper of intestinal mineral absorption essential for postnatal survival. Proc Natl Acad Sci U S A 2019; 116:4706-4715. [PMID: 30770447 PMCID: PMC6410795 DOI: 10.1073/pnas.1810633116] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Zn2+, Mg2+, and Ca2+ are the most abundant divalent metals in mammals. Different categories of cation-selective channels and transporters are thought to control the levels of individual metals in a cell-specific manner. However, the mechanisms responsible for the organismal balance of these minerals are poorly understood. Using genetic mouse models together with biophysical and biochemical analysis, we show that the channel-kinase TRPM7 is a master regulator of the organismal balance of divalent cations. TRPM7 activity is primarily required in the intestine, while TRPM7 function in the kidney—commonly thought to be essential—is expendable. Hence, against current thinking, organismal balance of multiple divalent cations predominantly relies on a common gatekeeper, TRPM7, rather than on individual specialized channels/transporters. Zn2+, Mg2+, and Ca2+ are essential minerals required for a plethora of metabolic processes and signaling pathways. Different categories of cation-selective channels and transporters are therefore required to tightly control the cellular levels of individual metals in a cell-specific manner. However, the mechanisms responsible for the organismal balance of these essential minerals are poorly understood. Herein, we identify a central and indispensable role of the channel-kinase TRPM7 for organismal mineral homeostasis. The function of TRPM7 was assessed by single-channel analysis of TRPM7, phenotyping of TRPM7-deficient cells in conjunction with metabolic profiling of mice carrying kidney- and intestine-restricted null mutations in Trpm7 and animals with a global “kinase-dead” point mutation in the gene. The TRPM7 channel reconstituted in lipid bilayers displayed a similar permeability to Zn2+ and Mg2+. Consistently, we found that endogenous TRPM7 regulates the total content of Zn2+ and Mg2+ in cultured cells. Unexpectedly, genetic inactivation of intestinal rather than kidney TRPM7 caused profound deficiencies specifically of Zn2+, Mg2+, and Ca2+ at the organismal level, a scenario incompatible with early postnatal growth and survival. In contrast, global ablation of TRPM7 kinase activity did not affect mineral homeostasis, reinforcing the importance of the channel activity of TRPM7. Finally, dietary Zn2+ and Mg2+ fortifications significantly extended the survival of offspring lacking intestinal TRPM7. Hence, the organismal balance of divalent cations critically relies on one common gatekeeper, the intestinal TRPM7 channel.
Collapse
|
120
|
Liu J, Hu G, Gong Y, Yu Q, He B, Li W, He Z, Hao W, He Z, Liu Y. Silencing of TRPM8 inhibits aggressive tumor phenotypes and enhances gemcitabine sensitivity in pancreatic cancer. Pancreatology 2018; 18:935-944. [PMID: 30316690 DOI: 10.1016/j.pan.2018.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 07/04/2018] [Accepted: 08/21/2018] [Indexed: 12/11/2022]
Abstract
The transient receptor potential TRPM8 ion channel is required for cellular proliferation in pancreatic epithelia and adenocarcinoma. To elucidate the mechanism that mediates the function of TRPM8, we examined its role in the proliferation and invasion of pancreatic cancer (PC) cells. TRPM8 expression increased in both the PC tissues and cell lines; a high TRPM8 expression was correlated with poorer prognosis in patients with PC. In PC cell lines, PACN-1 and BxPC-3, Ca2+ influxes could be evoked by TRPM8; the sensitivity of PC cells to gemcitabine was increased, while the proliferation and invasion of PC cells were suppressed after RNA interference-mediated silencing of TRPM8. The mechanism of TRPM8 in gemcitabine-based chemotherapy was then investigated. The expression and activity of multidrug resistance-associated proteins, P-gp, MRP-2, LRP, was significantly reduced in response to TRPM8 silence. Moreover, TRPM8 knockdown significantly increased hENT1 protein levels and the ratio of Bax/Bcl-2 while decreased the protein levels of RRM1. Thus, TRPM8 is required for PC cell proliferation and invasion and was closely related to the gemcitabine sensitivity of PC. The modulation of TRPM8 expression may help improve treatment response of PC by combining with traditional chemotherapy.
Collapse
Affiliation(s)
- JieFeng Liu
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - GuoHuang Hu
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - YuJing Gong
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - QianLe Yu
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - Bin He
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - WangHao Li
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - ZhiGuo He
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - WenJie Hao
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - ZiChao He
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - YiPing Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410078, People's Republic of China.
| |
Collapse
|
121
|
Abstract
Since its discovery, aldosterone and ion modulation have been entwined. While scientific investigations throughout the decades have emphasized aldosterone's connection to Na+, K+, and H+ homeostasis, more recent research has demonstrated a relationship between aldosterone and Mg2+, Ca2+, and Cl- homeostasis. The mechanisms connecting aldosterone to ion regulation frequently involve ion channels; the membrane localized proteins containing at least one aqueous pore for ion conduction. In order to precisely control intracellular or intraorganelle ion concentrations, ion channels have evolved highly specific regions within the conduction pore that select ions by charge, size, and/or dehydration energy requirement, meaning aldosterone must be able to modulate multiple ion channels to regulate the many ions described above. The list of ion channels presently connected to aldosterone includes ENaC (Na+), ROMK/BK (K+), TRPV4/5/6 (Ca2+), TRPM7/6 (Mg2+), and ClC-K/CFTR (Cl-), among others. This list is only expected to grow over time, as the promiscuity of aldosterone becomes more understood.
Collapse
Affiliation(s)
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Alvin Shrier
- Department of Physiology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
122
|
Zhang H, Wang Y, Lv Q, Gao J, Hu L, He Z. MicroRNA-21 Overexpression Promotes the Neuroprotective Efficacy of Mesenchymal Stem Cells for Treatment of Intracerebral Hemorrhage. Front Neurol 2018; 9:931. [PMID: 30459705 PMCID: PMC6233525 DOI: 10.3389/fneur.2018.00931] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/15/2018] [Indexed: 12/25/2022] Open
Abstract
Intracerebral hemorrhage (ICH) has high morbidity and mortality, with no effective treatment at present. One possible therapeutic strategy involves the use of mesenchymal stem cells (MSCs), which have shown promise in experimental models and have great potential for treating nervous illnesses in humans. However, many deficiencies in MSC treatment still need to be addressed, including their poor survival rate post-transplantation. Previously, we reported that the microRNA-21 (miR-21) is downregulated in ICH patients' blood and brain tissue. In this study, we aimed to examine its role and therapeutic efficacy in ICH using miR-21-overexpressing MSCs. We found that this microRNA can enhance MSC survival and recovery of neurological function in ICH rats. Its mechanism of action involves reduced neuronal apoptosis. In addition, we demonstrated that miR-21 can be transported to neurons through exosomes derived from MSCs and that it can target transient receptor potential melastatin 7 (TRPM7) to alleviate neuronal injury following ICH. We also observed that the NF-κB pathway is involved in the regulation of miR-21 in neural cells. In conclusion, miR-21 significantly enhances the survival of MSCs in ICH, and miR-21-overexpressing MSCs clearly improved neurological function in ICH rats. Transplantation of miR-21-overexpressing MSCs may, therefore, provide an effective strategy for neuroprotection and treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Heyu Zhang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Yanzhe Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Qing Lv
- Department of Neurology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Jun Gao
- Department of Neurology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Liuting Hu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Zhiyi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, Liaoning, China
| |
Collapse
|
123
|
Zhang X, Hu M, Yang Y, Xu H. Organellar TRP channels. Nat Struct Mol Biol 2018; 25:1009-1018. [PMID: 30374082 DOI: 10.1038/s41594-018-0148-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 09/28/2018] [Indexed: 02/08/2023]
Abstract
Mammalian transient receptor potential (TRP) channels mediate Ca2+ flux and voltage changes across membranes in response to environmental and cellular signals. At the plasma membrane, sensory TRPs act as neuronal detectors of physical and chemical environmental signals, and receptor-operated (metabotropic) TRPs decode extracellular neuroendocrine cues to control body homeostasis. In intracellular membranes, such as those in lysosomes, organellar TRPs respond to compartment-derived signals to control membrane trafficking, signal transduction, and organelle function. Complementing mouse and human genetics and high-resolution structural approaches, physiological studies employing natural agonists and synthetic inhibitors have become critical in resolving the in vivo functions of metabotropic, sensory, and organellar TRPs.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Meiqin Hu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.,Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yexin Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
124
|
Tian Y, Yang T, Yu S, Liu C, He M, Hu C. Prostaglandin E2 increases migration and proliferation of human glioblastoma cells by activating transient receptor potential melastatin 7 channels. J Cell Mol Med 2018; 22:6327-6337. [PMID: 30338939 PMCID: PMC6237613 DOI: 10.1111/jcmm.13931] [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: 03/05/2018] [Accepted: 08/30/2018] [Indexed: 12/26/2022] Open
Abstract
Recent studies showed that both prostaglandin E2 (PGE2) and transient receptor potential melastatin 7 (TRPM7) play important roles in migration and proliferation of human glioblastoma cells. In this study, we tested the association between PGE2 and TRPM7. We found that PGE2 increased TRPM7 currents in HEK293 and human glioblastoma A172 cells. The PGE2 EP3 receptor antagonist L‐798106 abrogated the PGE2 stimulatory effect, while EP3 agonist 17‐phenyl trinor prostaglandin E2 (17‐pt‐PGE2) mimicked the effect of PEG2 on TRPM7. The TRPM7 phosphotransferase activity‐deficient mutation, K1646R had no effect on PGE2 induced increase of TRPM7 currents. Inhibition of protein kinase A (PKA) activity by Rp‐cAMP increased TRPM7 currents. TRPM7 PKA phosphorylation site mutation S1269A abolished the PGE2 effect on TRPM7 currents. PGE2 increased both mRNA and membrane protein expression of TRPM7 in A172 cells. Knockdown of TRPM7 by shRNA abrogated the PGE2 stimulated migration and proliferation of A172 cells. Blockage of TRPM7 with 2‐aminoethoxydiphenyl borate (2‐APB) or NS8593 had a similar effect as TRPM7‐shRNA. In conclusion, our results demonstrate that PGE2 activates TRPM7 via EP3/PKA signalling pathway, and that PGE2 enhances migration and proliferation of human glioblastoma cells by up‐regulation of the TRPM7 channel.
Collapse
Affiliation(s)
- Yafei Tian
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Tingting Yang
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Shuntai Yu
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Cuiyun Liu
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Min He
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Changlong Hu
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
125
|
Schubert R. The second life of ion transporters as signal transducers. Acta Physiol (Oxf) 2018; 224:e13155. [PMID: 29938912 DOI: 10.1111/apha.13155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- R Schubert
- Cardiovascular Physiology, Centre for Biomedicine and Medical Technology Mannheim (CBTM), European Center of Angioscience (ECAS), Ruprecht-Karls-University Heidelberg, Mannheim, Germany
| |
Collapse
|
126
|
Duan J, Li Z, Li J, Hulse RE, Santa-Cruz A, Valinsky WC, Abiria SA, Krapivinsky G, Zhang J, Clapham DE. Structure of the mammalian TRPM7, a magnesium channel required during embryonic development. Proc Natl Acad Sci U S A 2018; 115:E8201-E8210. [PMID: 30108148 PMCID: PMC6126765 DOI: 10.1073/pnas.1810719115] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The transient receptor potential ion channel subfamily M, member 7 (TRPM7), is a ubiquitously expressed protein that is required for mouse embryonic development. TRPM7 contains both an ion channel and an α-kinase. The channel domain comprises a nonselective cation channel with notable permeability to Mg2+ and Zn2+ Here, we report the closed state structures of the mouse TRPM7 channel domain in three different ionic conditions to overall resolutions of 3.3, 3.7, and 4.1 Å. The structures reveal key residues for an ion binding site in the selectivity filter, with proposed partially hydrated Mg2+ ions occupying the center of the conduction pore. In high [Mg2+], a prominent external disulfide bond is found in the pore helix, which is essential for ion channel function. Our results provide a structural framework for understanding the TRPM1/3/6/7 subfamily and extend the knowledge base upon which to study the diversity and evolution of TRP channels.
Collapse
Affiliation(s)
- Jingjing Duan
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Zongli Li
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115
| | - Jian Li
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330031 Jiangxi, China
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536
| | - Raymond E Hulse
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Ana Santa-Cruz
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - William C Valinsky
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Sunday A Abiria
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115
| | | | - Jin Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115;
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330031 Jiangxi, China
| | - David E Clapham
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115;
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
127
|
Nadolni W, Zierler S. The Channel-Kinase TRPM7 as Novel Regulator of Immune System Homeostasis. Cells 2018; 7:cells7080109. [PMID: 30126133 PMCID: PMC6115979 DOI: 10.3390/cells7080109] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/13/2018] [Accepted: 08/13/2018] [Indexed: 12/16/2022] Open
Abstract
The enzyme-coupled transient receptor potential channel subfamily M member 7, TRPM7, has been associated with immunity and immune cell signalling. Here, we review the role of this remarkable signalling protein in lymphocyte proliferation, differentiation, activation and survival. We also discuss its role in mast cell, neutrophil and macrophage function and highlight the potential of TRPM7 to regulate immune system homeostasis. Further, we shed light on how the cellular signalling cascades involving TRPM7 channel and/or kinase activity culminate in pathologies as diverse as allergic hypersensitivity, arterial thrombosis and graft versus host disease (GVHD), stressing the need for TRPM7 specific pharmacological modulators.
Collapse
Affiliation(s)
- Wiebke Nadolni
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Goethestr. 33, 80336 Munich, Germany.
| | - Susanna Zierler
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Goethestr. 33, 80336 Munich, Germany.
| |
Collapse
|
128
|
Huang Y, Leng TD, Inoue K, Yang T, Liu M, Horgen FD, Fleig A, Li J, Xiong ZG. TRPM7 channels play a role in high glucose-induced endoplasmic reticulum stress and neuronal cell apoptosis. J Biol Chem 2018; 293:14393-14406. [PMID: 30076216 DOI: 10.1074/jbc.ra117.001032] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 06/29/2018] [Indexed: 12/12/2022] Open
Abstract
High-glucose (HG) levels and hyperglycemia associated with diabetes are known to cause neuronal damage. The detailed molecular mechanisms, however, remain to be elucidated. Here, we investigated the role of transient receptor potential melastatin 7 (TRPM7) channels in HG-mediated endoplasmic reticulum stress (ERS) and injury of NS20Y neuronal cells. The cells were incubated in the absence or presence of HG for 48 h. We found that mRNA and protein levels of TRPM7 and of ERS-associated proteins, such as C/EBP homologous protein (CHOP), 78-kDa glucose-regulated protein (GRP78), and inducible nitric-oxide synthase (iNOS), increased in HG-treated cells, along with significantly increased TRPM7-associated currents in these cells. Similar results were obtained in cerebral cortical tissue from an insulin-deficiency model of diabetic mice. Moreover, HG treatment of cells activated ERS-associated proapoptotic caspase activity and induced cellular injury. Interestingly, a NOS inhibitor, l-NAME, suppressed the HG-induced increase of TRPM7 expression and cellular injury. siRNA-mediated TRPM7 knockdown or chemical inhibition of TRPM7 activity also suppressed HG-induced ERS and decreased cleaved caspase-12/caspase-3 levels and cell injury. Of note, TRPM7 overexpression increased ERS and cell injury independently of its kinase activity. Taken together, our findings suggest that TRPM7 channel activities play a key role in HG-associated ERS and cytotoxicity through an apoptosis-inducing signaling cascade involving HG, iNOS, TRPM7, ERS proteins, and caspases.
Collapse
Affiliation(s)
- Yan Huang
- From the School of Pharmacy, Anhui Medical University, Hefei 230032, China.,the Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia, 30310
| | - Tian-Dong Leng
- the Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia, 30310,
| | - Koichi Inoue
- the Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia, 30310.,the Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Tao Yang
- the Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia, 30310
| | - Mingli Liu
- the Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia, 30310
| | - F David Horgen
- the Department of Natural Sciences, Hawaii Pacific University, Kaneohe, Hawaii 96744, and
| | - Andrea Fleig
- the Laboratory of Cell and Molecular Signaling, Center for Biomedical Research at The Queen's Medical Center and University of Hawaii John A. Burns School of Medicine and Cancer Center, Honolulu, Hawaii 96813
| | - Jun Li
- From the School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zhi-Gang Xiong
- the Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia, 30310,
| |
Collapse
|
129
|
Suzuki S, Lis A, Schmitz C, Penner R, Fleig A. The TRPM7 kinase limits receptor-induced calcium release by regulating heterotrimeric G-proteins. Cell Mol Life Sci 2018; 75:3069-3078. [PMID: 29500477 PMCID: PMC6033657 DOI: 10.1007/s00018-018-2786-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 02/06/2018] [Accepted: 02/26/2018] [Indexed: 12/17/2022]
Abstract
The melastatin-related transient receptor potential member 7 (TRPM7) is a unique fusion protein with both ion channel function and enzymatic α-kinase activity. TRPM7 is essential for cellular systemic magnesium homeostasis and early embryogenesis; it promotes calcium transport during global brain ischemia and emerges as a key player in cancer growth. TRPM7 channels are negatively regulated through G-protein-coupled receptor-stimulation, either by reducing cellular cyclic adenosine monophosphate (cAMP) or depleting phosphatidylinositol bisphosphate (PIP2) levels in the plasma membrane. We here identify that heterologous overexpression of human TRPM7-K1648R mutant will lead to disruption of protease or purinergic receptor-induced calcium release. The disruption occurs at the level of Gq, which requires intact TRPM7 kinase phosphorylation activity for orderly downstream signal transduction to activate phospholipase (PLC)β and cause calcium release. We propose that this mechanism may support limiting GPCR-mediated calcium signaling in times of insufficient cellular ATP supply.
Collapse
Affiliation(s)
- Sayuri Suzuki
- Center for Biomedical Research, The Queen's Medical Center, 1301 Punchbowl St., Honolulu, HI, 96813, USA
- John A. Burns School of Medicine, University of Hawaii Cancer Center, University of Hawaii, 651 Ilalo St, Honolulu, HI, 96813, USA
| | - Annette Lis
- Center for Biomedical Research, The Queen's Medical Center, 1301 Punchbowl St., Honolulu, HI, 96813, USA
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, 66421, Homburg/Saar, Germany
| | - Carsten Schmitz
- Integrated Department of Immunology, National Jewish Health, University of Colorado Denver, Denver, CO, 80206, USA
| | - Reinhold Penner
- Center for Biomedical Research, The Queen's Medical Center, 1301 Punchbowl St., Honolulu, HI, 96813, USA
- John A. Burns School of Medicine, University of Hawaii Cancer Center, University of Hawaii, 651 Ilalo St, Honolulu, HI, 96813, USA
| | - Andrea Fleig
- Center for Biomedical Research, The Queen's Medical Center, 1301 Punchbowl St., Honolulu, HI, 96813, USA.
- John A. Burns School of Medicine, University of Hawaii Cancer Center, University of Hawaii, 651 Ilalo St, Honolulu, HI, 96813, USA.
| |
Collapse
|
130
|
Ha YS, Kim YY, Yu NH, Chun SY, Choi SH, Lee JN, Kim BS, Yoo ES, Kwon TG. Down-regulation of transient receptor potential melastatin member 7 prevents migration and invasion of renal cell carcinoma cells via inactivation of the Src and Akt pathway. Investig Clin Urol 2018; 59:263-274. [PMID: 29984342 PMCID: PMC6028469 DOI: 10.4111/icu.2018.59.4.263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/17/2018] [Indexed: 11/18/2022] Open
Abstract
Purpose Transient receptor potential melastatin member 7 (TRPM7), an ion channel and serine/threonine protein kinase, has been linked with distinct human malignancies. However, the role of TRPM7 in renal cell carcinoma (RCC) has not been investigated. The aim of this study is to determine whether TRPM7 regulates the migration and invasion of RCC cells. Its relationship with signal transduction pathways was also studied. Materials and Methods The human RCC cell lines ACHN and SN12C were chosen for this study. The molecular mechanisms of TRPM7 action were studied using Western blot analysis and small interfering RNA (siRNA)-based knockdown. The effect of TRPM7 knockdown on RCC cells was measured by using Transwell invasion and wound healing migration assays. Results siRNA-induced silencing of TRPM7 notably decreased the migration and invasion of ACHN and SN12C RCC cells. The phosphorylation levels of Src in both cells were obviously reduced after TRPM7 silencing compared with that of the control ACHN and SN12C cells. Furthermore, the phosphorylation levels of Akt were greatly decreased in ACHN cells after siRNA-induced knockdown of TRPM7. Additionally, the treatment of cells with Src and Akt inhibitors clearly limited the migration and invasion of RCC cells. Conclusions Our data show that TRPM7 regulated ACHN and SN12C RCC cell invasion via the Src/Akt signaling pathway. Therefore, targeting the Src/Akt signaling pathway and/or the expression or function of TRPM7 could be a potential beneficial treatment for patients with RCC.
Collapse
Affiliation(s)
- Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Korea
| | - Yeon-Yong Kim
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu, Korea
- Immunoregulatory Material Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Korea
| | - Na Hee Yu
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Korea
| | - So Young Chun
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Korea
| | - Seock Hwan Choi
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Korea
| | - Jun Nyung Lee
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Korea
| | - Bum Soo Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Korea
| | - Eun Sang Yoo
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Korea
| | - Tae Gyun Kwon
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Korea
| |
Collapse
|
131
|
Hantute-Ghesquier A, Haustrate A, Prevarskaya N, Lehen'kyi V. TRPM Family Channels in Cancer. Pharmaceuticals (Basel) 2018; 11:ph11020058. [PMID: 29875336 PMCID: PMC6027338 DOI: 10.3390/ph11020058] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/28/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022] Open
Abstract
Members of the TRPM ("Melastatin") family fall into the subclass of the TRP channels having varying permeability to Ca2+ and Mg2+, with three members of the TRPM family being chanzymes, which contain C-terminal enzyme domains. The role of different TRPM members has been shown in various cancers such as prostate cancer for mostly TRPM8 and TRPM2, breast cancer for mostly TRPM2 and TRPM7, and pancreatic cancer for TRPM2/7/8 channels. The role of TRPM5 channels has been shown in lung cancer, TRPM1 in melanoma, and TRPM4 channel in prostate cancer as well. Thus, the TRPM family of channels may represent an appealing target for the anticancer therapy.
Collapse
Affiliation(s)
- Aline Hantute-Ghesquier
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d'Ascq, France.
| | - Aurélien Haustrate
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d'Ascq, France.
- FONDATION ARC, 9 rue Guy Môquet 94830 Villejuif, France.
| | - Natalia Prevarskaya
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d'Ascq, France.
| | - V'yacheslav Lehen'kyi
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d'Ascq, France.
- FONDATION ARC, 9 rue Guy Môquet 94830 Villejuif, France.
| |
Collapse
|
132
|
Krishnamoorthy M, Wasim L, Buhari FHM, Zhao T, Mahtani T, Ho J, Kang S, Deason-Towne F, Perraud AL, Schmitz C, Treanor B. The channel-kinase TRPM7 regulates antigen gathering and internalization in B cells. Sci Signal 2018; 11:11/533/eaah6692. [PMID: 29871912 DOI: 10.1126/scisignal.aah6692] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Members of the transient receptor potential (TRP) family of ion channels are cellular sensors involved in numerous physiological and pathological processes. We identified the TRP subfamily M member 7 (TRPM7) channel-kinase as a previously uncharacterized regulator of B cell activation. We showed that TRPM7 played a critical role in the early events of B cell activation through both its ion channel and kinase functions. DT40 B cells deficient in TRPM7 or expressing a kinase-deficient mutant of TRPM7 showed defective gathering of antigen and prolonged B cell receptor (BCR) signaling. We showed that lipid metabolism was altered in TRPM7-deficient cells and in cells expressing a kinase-deficient mutant of TRPM7 and suggest that PLC-γ2 may be a target of the kinase activity of TRPM7. Primary B cells that expressed less TRPM7 or were treated with a pharmacological inhibitor of TRPM7 also displayed defective antigen gathering and increased BCR signaling. Finally, we demonstrated that blocking TRPM7 function compromised antigen internalization and presentation to T cells. These data suggest that TRPM7 controls an essential process required for B cell affinity maturation and the production of high-affinity antibodies.
Collapse
Affiliation(s)
- Mithunah Krishnamoorthy
- Department of Cell and Systems Biology, University of Toronto, 24 Harbord Street, Toronto, Ontario M5S 3G5, Canada
| | - Laabiah Wasim
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Fathima Hifza Mohamed Buhari
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario M1C 1A4, Canada
| | - Tiantian Zhao
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Trisha Mahtani
- Department of Cell and Systems Biology, University of Toronto, 24 Harbord Street, Toronto, Ontario M5S 3G5, Canada
| | - Josephine Ho
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario M1C 1A4, Canada
| | - Sohee Kang
- Department of Computer and Mathematical Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada
| | - Francina Deason-Towne
- Department of Immunology and Microbiology, University of Colorado, Denver, CO 80206, USA
| | - Anne-Laure Perraud
- Department of Immunology and Microbiology, University of Colorado, Denver, CO 80206, USA
| | - Carsten Schmitz
- Department of Immunology and Microbiology, University of Colorado, Denver, CO 80206, USA.,Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA
| | - Bebhinn Treanor
- Department of Cell and Systems Biology, University of Toronto, 24 Harbord Street, Toronto, Ontario M5S 3G5, Canada. .,Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.,Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario M1C 1A4, Canada
| |
Collapse
|
133
|
Cai N, Lou L, Al-Saadi N, Tetteh S, Runnels LW. The kinase activity of the channel-kinase protein TRPM7 regulates stability and localization of the TRPM7 channel in polarized epithelial cells. J Biol Chem 2018; 293:11491-11504. [PMID: 29866880 DOI: 10.1074/jbc.ra118.001925] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/25/2018] [Indexed: 12/13/2022] Open
Abstract
The channel-kinase transient receptor potential melastatin 7 (TRPM7) is a bifunctional protein with ion channel and kinase domains. The kinase activity of TRPM7 has been linked to the regulation of a broad range of cellular activities, but little is understood as to how the channel itself is regulated by its own kinase activity. Here, using several mammalian cell lines expressing WT TRPM7 or kinase-inactive variants, we discovered that compared with the cells expressing WT TRPM7, cells in which TRPM7's kinase activity was inactivated had faster degradation, elevated ubiquitination, and increased intracellular retention of the channel. Mutational analysis of TRPM7 autophosphorylation sites further revealed a role for Ser-1360 of TRPM7 as a key residue mediating both TRPM7 stability and intracellular trafficking. Additional trafficking roles were uncovered for Ser-1403 and Ser-1567, whose phosphorylation by TRPM7's kinase activity mediated the interaction of the channel with the signaling protein 14-3-3θ. In summary, our results point to a critical role for TRPM7's kinase activity in regulating proteasome-mediated turnover of the TRPM7 channel and controlling its cellular localization in polarized epithelial cells. Overall, these findings improve our understanding of the significance of TRPM7's kinase activity for functional regulation of its channel activity.
Collapse
Affiliation(s)
- Na Cai
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Liping Lou
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Namariq Al-Saadi
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854; University of Misan, Amarah 62001, Iraq
| | - Sandra Tetteh
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Loren W Runnels
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854.
| |
Collapse
|
134
|
mRNA expression of transient receptor potential melastatin (TRPM) channels 2 and 7 in perinatal brain development. Int J Dev Neurosci 2018; 69:23-31. [DOI: 10.1016/j.ijdevneu.2018.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/04/2018] [Accepted: 05/18/2018] [Indexed: 12/31/2022] Open
|
135
|
Zhang J, Chen X, Xue Y, Gamper N, Zhang X. Beyond voltage-gated ion channels: Voltage-operated membrane proteins and cellular processes. J Cell Physiol 2018; 233:6377-6385. [PMID: 29667735 DOI: 10.1002/jcp.26555] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 02/20/2018] [Indexed: 12/26/2022]
Abstract
Voltage-gated ion channels were believed to be the only voltage-sensitive proteins in excitable (and some non-excitable) cells for a long time. Emerging evidence indicates that the voltage-operated model is shared by some other transmembrane proteins expressed in both excitable and non-excitable cells. In this review, we summarize current knowledge about voltage-operated proteins, which are not classic voltage-gated ion channels as well as the voltage-dependent processes in cells for which single voltage-sensitive proteins have yet to be identified. Particularly, we will focus on the following. (1) Voltage-sensitive phosphoinositide phosphatases (VSP) with four transmembrane segments homologous to the voltage sensor domain (VSD) of voltage-gated ion channels; VSPs are the first family of proteins, other than the voltage-gated ion channels, for which there is sufficient evidence for the existence of the VSD domain; (2) Voltage-gated proton channels comprising of a single voltage-sensing domain and lacking an identified pore domain; (3) G protein coupled receptors (GPCRs) that mediate the depolarization-evoked potentiation of Ca2+ mobilization; (4) Plasma membrane (PM) depolarization-induced but Ca2+ -independent exocytosis in neurons. (5) Voltage-dependent metabolism of phosphatidylinositol 4,5-bisphosphate (PtdIns[4,5]P2 , PIP2 ) in the PM. These recent discoveries expand our understanding of voltage-operated processes within cellular membranes.
Collapse
Affiliation(s)
- Jianping Zhang
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xingjuan Chen
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Beijing Key Laboratory of Diabetes Prevention and Research, Lu He Hospital, Capital Medical University, Beijing, China
| | - Yucong Xue
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Nikita Gamper
- Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Xuan Zhang
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
136
|
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
|
137
|
|
138
|
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]
|
139
|
Won J, Vang H, Kim JH, Lee PR, Kang Y, Oh SB. TRPM7 Mediates Mechanosensitivity in Adult Rat Odontoblasts. J Dent Res 2018; 97:1039-1046. [PMID: 29489440 DOI: 10.1177/0022034518759947] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Odontoblasts, with their strategic arrangement along the outermost compartment of the dentin-pulp complex, have been suggested to have sensory function. In addition to their primary role in dentin formation, growing evidence shows that odontoblasts are capable of sensing mechanical stimulation. Previously, we found that most odontoblasts express TRPM7, the nonselective mechanosensitive ion channel reported to be critical in Mg2+ homeostasis and dentin mineralization. In line with this finding, we sought to elucidate the functional expression of TRPM7 in odontoblasts by pharmacological approaches and mechanical stimulation. Naltriben, a TRPM7-specific agonist, induced calcium transient in the majority of odontoblasts, which was blocked by TRPM7 blockers such as extracellular Mg2+ and FTY720 in a dose-dependent manner. Mechanical stretch of the odontoblastic membrane with hypotonic solution also induced calcium transient, which was blocked by Gd3+, a nonselective mechanosensitive channel blocker. Calcium transient induced by hypotonic solution was also blocked by high extracellular Mg2+ or FTY720. When TRPM7-mediated calcium transients in odontoblasts were analyzed on the subcellular level, remarkably larger transients were detected in the distal odontoblastic process compared with the soma, which was further verified with comparable immunocytochemical analysis. Our results demonstrate that TRPM7 in odontoblasts can serve as a mechanical sensor, with its distribution to facilitate intracellular Ca2+ signaling in the odontoblastic process. These findings suggest TRPM7 as a mechanical transducer in odontoblasts to mediate intracellular calcium dynamics under diverse pathophysiological conditions of the dentin.
Collapse
Affiliation(s)
- J Won
- 1 Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - H Vang
- 2 Dental Research Institute and Department of Neurobiology & Physiology, School of Dentistry, Seoul National University, Seoul, Republic of Korea.,3 Department of Basic Science in Dentistry, University of Health Sciences, Vientiane, Laos
| | - J H Kim
- 1 Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - P R Lee
- 1 Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Y Kang
- 2 Dental Research Institute and Department of Neurobiology & Physiology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - S B Oh
- 1 Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea.,2 Dental Research Institute and Department of Neurobiology & Physiology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
140
|
Inactivation of TRPM7 kinase in mice results in enlarged spleens, reduced T-cell proliferation and diminished store-operated calcium entry. Sci Rep 2018; 8:3023. [PMID: 29445164 PMCID: PMC5813043 DOI: 10.1038/s41598-018-21004-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/29/2018] [Indexed: 12/20/2022] Open
Abstract
T lymphocytes enlarge (blast) and proliferate in response to antigens in a multistep program that involves obligatory cytosolic calcium elevations. Store-operated calcium entry (SOCE) pathway is the primary source of Ca2+ in these cells. Here, we describe a novel modulator of blastogenesis, proliferation and SOCE: the TRPM7 channel kinase. TRPM7 kinase-dead (KD) K1646R knock-in mice exhibited splenomegaly and impaired blastogenic responses elicited by PMA/ionomycin or anti-CD3/CD28 antibodies. Splenic T-cell proliferation in vitro was weaker in the mutant compared to wildtype littermates. TRPM7 current magnitudes in WT and KD mouse T cells were, however, similar. We tested the dependence of T-cell proliferation on external Ca2+ and Mg2+ concentrations. At a fixed [Mg2+o] of ~0.4 mM, Ca2+o stimulated proliferation with a steep concentration dependence and vice versa, at a fixed [Ca2+o] of ~0.4 mM, Mg2+o positively regulated proliferation but with a shallower dependence. Proliferation was significantly lower in KD mouse than in wildtype at all Ca2+ and Mg2+ concentrations. Ca2+ elevations elicited by anti-CD3 antibody were diminished in KD mutant T cells and SOCE measured in activated KD splenocytes was reduced. These results demonstrate that a functional TRPM7 kinase supports robust SOCE, blastogenesis and proliferation, whereas its inactivation suppresses these cellular events.
Collapse
|
141
|
Gotru SK, Chen W, Kraft P, Becker IC, Wolf K, Stritt S, Zierler S, Hermanns HM, Rao D, Perraud AL, Schmitz C, Zahedi RP, Noy PJ, Tomlinson MG, Dandekar T, Matsushita M, Chubanov V, Gudermann T, Stoll G, Nieswandt B, Braun A. TRPM7 Kinase Controls Calcium Responses in Arterial Thrombosis and Stroke in Mice. Arterioscler Thromb Vasc Biol 2018; 38:344-352. [DOI: 10.1161/atvbaha.117.310391] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/30/2017] [Indexed: 11/16/2022]
Affiliation(s)
- Sanjeev K. Gotru
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Wenchun Chen
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Peter Kraft
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Isabelle C. Becker
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Karen Wolf
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Simon Stritt
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Susanna Zierler
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Heike M. Hermanns
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Deviyani Rao
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Anne-Laure Perraud
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Carsten Schmitz
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - René P. Zahedi
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Peter J. Noy
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Michael G. Tomlinson
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Thomas Dandekar
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Masayuki Matsushita
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Vladimir Chubanov
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Thomas Gudermann
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Guido Stoll
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Bernhard Nieswandt
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| | - Attila Braun
- From the Institute of Experimental Biomedicine, University Hospital of Würzburg (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), Rudolf Virchow Center (S.K.G., W.C., I.C.B., K.W., S.S., B.N., A.B.), and Institute of Clinical Epidemiology and Biometry, Comprehensive Heart Failure Center (P.K.), University of Würzburg, Germany; Department of Hepatology (H.M.H.) and Department of Neurology (P.K., G.S.), University Hospital of Würzburg, Germany; Walther-Straub-Institute for Pharmacology and Toxicology,
| |
Collapse
|
142
|
Aldosterone, SGK1, and ion channels in the kidney. Clin Sci (Lond) 2018; 132:173-183. [PMID: 29352074 PMCID: PMC5817097 DOI: 10.1042/cs20171525] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/15/2017] [Accepted: 12/19/2017] [Indexed: 12/14/2022]
Abstract
Hyperaldosteronism, a common cause of hypertension, is strongly connected to Na+, K+, and Mg2+ dysregulation. Owing to its steroidal structure, aldosterone is an active transcriptional modifier when bound to the mineralocorticoid receptor (MR) in cells expressing the enzyme 11β-hydroxysteroid dehydrogenase 2, such as those comprising the aldosterone-sensitive distal nephron (ASDN). One such up-regulated protein, the ubiquitous serum and glucocorticoid regulated kinase 1 (SGK1), has the capacity to modulate the surface expression and function of many classes of renal ion channels, including those that transport Na+ (ENaC), K+ (ROMK/BK), Ca2+ (TRPV4/5/6), Mg2+ (TRPM7/6), and Cl− (ClC-K, CFTR). Here, we discuss the mechanisms by which ASDN expressed channels are up-regulated by SGK1, while highlighting newly discovered pathways connecting aldosterone to nonselective cation channels that are permeable to Mg2+ (TRPM7) or Ca2+ (TRPV4).
Collapse
|
143
|
Guo J, She J, Zeng W, Chen Q, Bai XC, Jiang Y. Structures of the calcium-activated, non-selective cation channel TRPM4. Nature 2017; 552:205-209. [PMID: 29211714 PMCID: PMC5901961 DOI: 10.1038/nature24997] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/03/2017] [Indexed: 12/12/2022]
Abstract
TRPM4 is a calcium-activated, phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2) -modulated, non-selective cation channel that belongs to the family of melastatin-related transient receptor potential (TRPM) channels. Here we present the electron cryo-microscopy structures of the mouse TRPM4 channel with and without ATP. TRPM4 consists of multiple transmembrane and cytosolic domains, which assemble into a three-tiered architecture. The N-terminal nucleotide-binding domain and the C-terminal coiled-coil participate in the tetrameric assembly of the channel; ATP binds at the nucleotide-binding domain and inhibits channel activity. TRPM4 has an exceptionally wide filter but is only permeable to monovalent cations; filter residue Gln973 is essential in defining monovalent selectivity. The S1-S4 domain and the post-S6 TRP domain form the central gating apparatus that probably houses the Ca2+- and PtdIns(4,5)P2-binding sites. These structures provide an essential starting point for elucidating the complex gating mechanisms of TRPM4 and reveal the molecular architecture of the TRPM family.
Collapse
Affiliation(s)
- Jiangtao Guo
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA
| | - Ji She
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA
| | - Weizhong Zeng
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA
| | - Qingfeng Chen
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA
| | - Xiao-Chen Bai
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA.,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9039, USA
| | - Youxing Jiang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA
| |
Collapse
|
144
|
Kondratskyi A, Kondratska K, Skryma R, Klionsky DJ, Prevarskaya N. Ion channels in the regulation of autophagy. Autophagy 2017; 14:3-21. [PMID: 28980859 PMCID: PMC5846505 DOI: 10.1080/15548627.2017.1384887] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 09/07/2017] [Accepted: 09/21/2017] [Indexed: 12/18/2022] Open
Abstract
Autophagy is a cellular process in which the cell degrades and recycles its own constituents. Given the crucial role of autophagy in physiology, deregulation of autophagic machinery is associated with various diseases. Hence, a thorough understanding of autophagy regulatory mechanisms is crucially important for the elaboration of efficient treatments for different diseases. Recently, ion channels, mediating ion fluxes across cellular membranes, have emerged as important regulators of both basal and induced autophagy. However, the mechanisms by which specific ion channels regulate autophagy are still poorly understood, thus underscoring the need for further research in this field. Here we discuss the involvement of major types of ion channels in autophagy regulation.
Collapse
Affiliation(s)
- Artem Kondratskyi
- Inserm, U-1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille 1, Villeneuve d'Ascq, France
| | - Kateryna Kondratska
- Inserm, U-1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille 1, Villeneuve d'Ascq, France
| | - Roman Skryma
- Inserm, U-1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille 1, Villeneuve d'Ascq, France
| | - Daniel J. Klionsky
- Life Sciences Institute, and Department of Molecular, Cellular and Developmental Biology; University of Michigan, Ann Arbor, MI, USA
| | - Natalia Prevarskaya
- Inserm, U-1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille 1, Villeneuve d'Ascq, France
| |
Collapse
|
145
|
Lange I, Espinoza-Fuenzalida I, Ali MW, Serrano LE, Koomoa DLT. FTY-720 induces apoptosis in neuroblastoma via multiple signaling pathways. Oncotarget 2017; 8:109985-109999. [PMID: 29299124 PMCID: PMC5746359 DOI: 10.18632/oncotarget.22452] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/17/2017] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma (NB) is the most common extra-cranial pediatric solid tumor. High-risk NB is difficult to treat due to the lack of response to current therapies and aggressive disease progression. Despite novel drugs, alternative treatments and multi-modal treatments, finding an effective treatment strategy for these patients continues to be a major challenge. The current study focuses on examining the effects of FTY-720 or fingolimod, a drug that is FDA-approved for refractory multiple sclerosis, in NB. The results showed that FTY-720 regulates multiple pathways that result in various effects on calcium signaling, ion channel activation and cell survival/death pathways. FTY-720 rapidly inhibits TRPM7 channel activity, and inhibited TRPM7 kinase activity, modulates calcium signaling, induces a loss of mitochondrial membrane potential and opening of the mitochondrial permeability transition pore, and ultimately leads to cell death. Interestingly, the data also showed that low concentrations of FTY-720 sensitized drug-resistant NB cells to antineoplastic drugs. These results suggest that FTY-720 may be an attractive alternative for the treatment of NB.
Collapse
Affiliation(s)
- Ingo Lange
- University of Hawaii at Hilo, The Daniel K. Inouye College of Pharmacy, Hilo, HI 96720, USA
| | | | - Mourad Wagdy Ali
- University of Hawaii at Hilo, The Daniel K. Inouye College of Pharmacy, Hilo, HI 96720, USA
| | - Laura Espana Serrano
- University of Hawaii at Hilo, The Daniel K. Inouye College of Pharmacy, Hilo, HI 96720, USA
| | - Dana-Lynn T Koomoa
- University of Hawaii at Hilo, The Daniel K. Inouye College of Pharmacy, Hilo, HI 96720, USA
| |
Collapse
|
146
|
Ion channel functional protein kinase TRPM7 regulates Mg ions to promote the osteoinduction of human osteoblast via PI3K pathway: In vitro simulation of the bone-repairing effect of Mg-based alloy implant. Acta Biomater 2017; 63:369-382. [PMID: 28882757 DOI: 10.1016/j.actbio.2017.08.051] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/31/2017] [Accepted: 08/31/2017] [Indexed: 01/29/2023]
Abstract
Mg-based alloys, as the potential orthopaedic implant, can self-degrade to avoid second operation for its remove, and enable to promote bone repair; however, the underlying molecular mechanisms remain unclear. In the present study, we examined the effect of Mg ions on osteogenesis, chemotaxis and anti-alkaline stress in hFOB1.19 human osteoblast cells to simulate bone-repairing effect of a biodegradable Mg-based alloy implant in vitro, and explored the regulatory role of the transient receptor potential melastatin 7 (TRPM7)/phosphoinositide 3-kinase (PI3K) signalling pathway in the process of Mg ion-induced bone repair by knockdown of TRPM7 and antagonizing PI3K activity. Results indicate that Mg ions up-regulated the expression of Runx2 and alkaline phosphatase (ALP) through TRPM7/PI3K signalling pathway, which could significantly enhance the osteogenic activity of human osteoblasts. Furthermore, the expression levels of MMP2, MMP9 and vascular endothelial growth factor (VEGF) were increased by TRPM7/PI3K signalling pathway, which recruits osteoblasts from low- to high-Mg ion environments by inducing cell migration. Although an alkaline environment has antibacterial effects, alkaline stress can cause cytotoxicity and induce cell death. Finally, we found that Mg ions could activate PI3K phosphorylation to promote cell growth and survival, protecting cells against the alkaline-stress-induced cytotoxicity caused by the degradation of Mg-based alloy implants. Our study not only revealed the molecular mechanism of Mg in promoting bone repair but also explained the protective effects of Mg ions on osteoblasts in an alkaline environment, which provides a theoretical basis and new directions for the application of Mg-based alloy implant material in orthopaedics fixations and osteosarcoma treatment. STATEMENTS OF SIGNIFICANCE As a potential biomaterial for orthopaedic implant, biodegradable magnesium has several advantages including self-degradation and bone repair promotion; however, the underlying mechanisms and effective concentration by which molecular regulates the bone repair remain unclear. The present study revealed that Mg ion and its effective concentration for activating PI3K phosphorylation via TRPM7, which causes three processes affecting bone repair, namely, osteoblast recruitment, osteogenesis and resistance to alkaline stress in human osteoblast. Therefore, our results have provided insight into the underlying molecular biological basis, and guidance for manipulating degradation rate, such as surface modification, of orthopaedic Mg-based implants.
Collapse
|
147
|
Assessment of TRPM7 functions by drug-like small molecules. Cell Calcium 2017; 67:166-173. [DOI: 10.1016/j.ceca.2017.03.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 12/28/2022]
|
148
|
Kühn F, Kühn C, Lückhoff A. Different Principles of ADP-Ribose-Mediated Activation and Opposite Roles of the NUDT9 Homology Domain in the TRPM2 Orthologs of Man and Sea Anemone. Front Physiol 2017; 8:879. [PMID: 29163217 PMCID: PMC5671594 DOI: 10.3389/fphys.2017.00879] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/18/2017] [Indexed: 01/31/2023] Open
Abstract
A decisive element in the human cation channel TRPM2 is a region in its cytosolic C-terminus named NUDT9H because of its homology to the NUDT9 enzyme, a pyrophosphatase degrading ADP-ribose (ADPR). In hTRPM2, however, the NUDT9H domain has lost its enzymatic activity but serves as a binding domain for ADPR. As consequence of binding, gating of the channel is initiated. Since ADPR is produced after oxidative DNA damage, hTRPM2 mediates Ca2+ influx in response to oxidative stress which may lead to cell death. In the genome of the sea anemone Nematostella vectensis (nv), a preferred model organism for the evolution of key bilaterian features, a TRPM2 ortholog has been identified that contains a NUDT9H domain as well. Heterologous expression of nvTRPM2 in HEK-293 cells reveals a cation channel with many close similarities to the human counterpart. Most notably, nvTRPM2 is activated by ADPR, and Ca2+ is a co-agonist. However, the intramolecular mechanisms of ADPR gating as well as the role of NUDT9H are strikingly different in the two species. Whereas already subtle changes of NUDT9H abolish ADPR gating in hTRPM2, the region can be completely removed from nvTRPM2 without loss of responses to ADPR. An alternative ADPR binding site seems to be present but has not yet been characterized. The ADP-ribose pyrophosphatase (ADPRase) function of nvNUDT9H has been preserved but can be abolished by numerous genetic manipulations. All these manipulations create channels that are sensitive to hydrogen peroxide which fails to induce channel activity in wild-type nvTRPM2. Therefore, the function of NUDT9H in nvTRPM2 is the degradation of ADPR, thereby reducing agonist concentration in the presence of oxidative stress. Thus, the two TRPM2 orthologs have evolved divergently but nevertheless gained analogous functional properties, i.e., gating by ADPR with Ca2+ as co-factor. Opposite roles are played by the respective NUDT9H domains, either binding of ADPR and mediating channel activity, or controlling the availability of ADPR at the binding site located in a different domain.
Collapse
Affiliation(s)
- Frank Kühn
- Medical Faculty, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| | - Cornelia Kühn
- Medical Faculty, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| | - Andreas Lückhoff
- Medical Faculty, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
149
|
Pérez-García MT, Cidad P, López-López JR. The secret life of ion channels: Kv1.3 potassium channels and proliferation. Am J Physiol Cell Physiol 2017; 314:C27-C42. [PMID: 28931540 DOI: 10.1152/ajpcell.00136.2017] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Kv1.3 channels are involved in the switch to proliferation of normally quiescent cells, being implicated in the control of cell cycle in many different cell types and in many different ways. They modulate membrane potential controlling K+ fluxes, sense changes in potential, and interact with many signaling molecules through their intracellular domains. From a mechanistic point of view, we can describe the role of Kv1.3 channels in proliferation with at least three different models. In the "membrane potential model," membrane hyperpolarization resulting from Kv1.3 activation provides the driving force for Ca2+ influx required to activate Ca2+-dependent transcription. This model explains most of the data obtained from several cells from the immune system. In the "voltage sensor model," Kv1.3 channels serve mainly as sensors that transduce electrical signals into biochemical cascades, independently of their effect on membrane potential. Kv1.3-dependent proliferation of vascular smooth muscle cells (VSMCs) could fit this model. Finally, in the "channelosome balance model," the master switch determining proliferation may be related to the control of the Kv1.3 to Kv1.5 ratio, as described in glial cells and also in VSMCs. Since the three mechanisms cannot function independently, these models are obviously not exclusive. Nevertheless, they could be exploited differentially in different cells and tissues. This large functional flexibility of Kv1.3 channels surely gives a new perspective on their functions beyond their elementary role as ion channels, although a conclusive picture of the mechanisms involved in Kv1.3 signaling to proliferation is yet to be reached.
Collapse
Affiliation(s)
- M Teresa Pérez-García
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas , Valladolid , Spain
| | - Pilar Cidad
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas , Valladolid , Spain
| | - José R López-López
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas , Valladolid , Spain
| |
Collapse
|
150
|
Oh HG, Chung S. Activation of transient receptor potential melastatin 7 (TRPM7) channel increases basal autophagy and reduces amyloid β-peptide. Biochem Biophys Res Commun 2017; 493:494-499. [PMID: 28870815 DOI: 10.1016/j.bbrc.2017.08.163] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 11/24/2022]
Abstract
Cerebral accumulation of amyloid β-peptide (Aβ), which is produced from amyloid precursor protein (APP), is the primary cause of Alzheimer's disease (AD). Autophagy recycles cellular components and digests intracellular components including Aβ. The Ca2+- and Mg2+-permeable transient receptor potential melastatin 7 (TRPM7) channel underlies the constitutive Ca2+ influx in some cells. Since we already reported that TRPM7 channel-mediated Ca2+ influx regulates basal autophagy, we hypothesize that the activation of TRPM7 channel could increase basal autophagy and consequently decrease Aβ. In this study, we showed that naltriben (NTB), a specific TRPM7 channel activator, induced Ca2+ influx and activated autophagic signaling in neuroblastoma SH-SY5Y cells. NTB also promoted co-localization of LC3 and APP, and reduced Aβ. Furthermore, we found that an early-onset familial AD-associated presenilin1 ΔE9 (PS1 ΔE9) mutant cells had attenuated basal autophagy. NTB was able to recover autophagy and decrease Aβ in PS1 ΔE9 cells. Our results show that the activating TRPM7 channel may prevent AD-related Aβ neuropathology via modulating Ca2+-regulated basal autophagy.
Collapse
Affiliation(s)
- Hyun Geun Oh
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon 440-746, South Korea
| | - Sungkwon Chung
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon 440-746, South Korea.
| |
Collapse
|