1
|
Rajamanickam G, Hu Z, Liao P. Targeting the TRPM4 Channel for Neurologic Diseases: Opportunity and Challenge. Neuroscientist 2025:10738584251318979. [PMID: 40012174 DOI: 10.1177/10738584251318979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
As a monovalent cation channel, the transient receptor potential melastatin 4 (TRPM4) channel is a unique member of the transient receptor potential family. Abnormal TRPM4 activity has been identified in various neurologic disorders, such as stroke, spinal cord injury, traumatic brain injury, multiple sclerosis, amyotrophic lateral sclerosis, pathologic pain, and epilepsy. Following brain hypoxia/ischemia and inflammation, TRPM4 up-regulation and enhanced activity contribute to the cell death of neurons, vascular endothelial cells, and astrocytes. Enhanced ionic influx via TRPM4 leads to cell volume increase and oncosis. Depolarization of membrane potential following TRPM4 activation and interaction between TRPM4 and N-methyl-d-aspartate receptors exacerbate excitotoxicity during hypoxia. Importantly, TRPM4 expression and activity remain low in healthy neurons, making it an ideal drug target. Current approaches to inhibit or modulate the TRPM4 channel have various limitations that hamper the interpretation of TRPM4 physiology in the nervous system and potentially hinder their translation into therapy. In this review, we discuss the pathophysiologic roles of TRPM4 and the different inhibitors that modulate TRPM4 activity for potential treatment of neurologic diseases.
Collapse
Affiliation(s)
| | - Zhenyu Hu
- Calcium Signalling Laboratory, National Neuroscience Institute, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ping Liao
- Calcium Signalling Laboratory, National Neuroscience Institute, Singapore
- Health and Social Sciences Cluster, Singapore Institute of Technology, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
2
|
Bochen F, Subedi S, La Manna F, Jarrin S, Papapostolou I, Kruithof-de Julio M, Peinelt C. TRPM4 contributes to cell death in prostate cancer tumor spheroids, and to extravasation and metastasis in a zebrafish xenograft model system. Mol Oncol 2025. [PMID: 39821469 DOI: 10.1002/1878-0261.13795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/29/2024] [Accepted: 12/15/2024] [Indexed: 01/19/2025] Open
Abstract
Transient receptor potential melastatin-4 (TRPM4) ion channel expression is upregulated in prostate cancer (PCa), contributing to increased cell proliferation, migration, adhesion, epithelial-to-mesenchymal transition, cell cycle shift, and alterations of intracellular Ca2+ signaling. GEO2R platform analysis of messenger RNA (mRNA) expression of ~ 6350 genes in normal and malignant prostate tissue samples from 15 PCa patients demonstrates that TRPM4 expression is upregulated sixfold and is among the most significantly upregulated genes in PCa. We find that absence of TRPM4 reduced PCa tumor spheroid size and decreased PCa tumor spheroid outgrowth. In addition, lack of TRPM4 increased cell death in PCa tumor spheroids, a phenotype that is absent in two-dimensional (2D) cancer cell systems. Lastly, absence of TRPM4 in PCa cells reduced extravasation and metastatic burden in a preclinical zebrafish cancer model. Taken together, our findings show that TRPM4 is an attractive therapeutic target in PCa and highlights the need for future development of pharmacological tools.
Collapse
Affiliation(s)
- Florian Bochen
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Saurav Subedi
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Federico La Manna
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Sofia Jarrin
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Irida Papapostolou
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Binkle-Ladisch L, Pironet A, Zaliani A, Alcouffe C, Mensching D, Haferkamp U, Willing A, Woo MS, Erdmann A, Jessen T, Hess SD, Gribbon P, Pless O, Vennekens R, Friese MA. Identification and development of TRPM4 antagonists to counteract neuronal excitotoxicity. iScience 2024; 27:111425. [PMID: 39687019 PMCID: PMC11648915 DOI: 10.1016/j.isci.2024.111425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/21/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration in central nervous system disorders is linked to dysregulated neuronal calcium. Direct inhibition of glutamate-induced neuronal calcium influx, particularly via N-methyl-D-aspartate receptors (NMDAR), has led to adverse effects and clinical trial failures. A more feasible approach is to modulate NMDAR activity or calcium signaling indirectly. In this respect, the calcium-activated non-selective cation channel transient receptor potential melastatin 4 (TRPM4) has been identified as a promising target. However, high affinity and specific antagonists are lacking. Here, we conducted high-throughput screening of a compound library to identify high affinity TRPM4 antagonists. This yielded five lead compound series with nanomolar half-maximal inhibitory concentration values. Through medicinal chemistry optimization of two series, we established detailed structure-activity relationships and inhibition of excitotoxicity in neurons. Moreover, we identified their potential binding site supported by electrophysiological measurements. These potent TRPM4 antagonists are promising drugs for treating neurodegenerative disorders and TRPM4-related pathologies, potentially overcoming previous therapeutic challenges.
Collapse
Affiliation(s)
- Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andy Pironet
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Campus Gasthuisberg O/N1, Herestraat 49-Bus 802, 3000 Leuven, Belgium
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Chantal Alcouffe
- Department of Chemistry, Evotec SE, 195 Route D'Espagne, 31036 Toulouse, France
| | - Daniel Mensching
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Anne Willing
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marcel S. Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Alexandre Erdmann
- Department of Chemistry, Evotec SE, 195 Route D'Espagne, 31036 Toulouse, France
| | | | - Stephen D. Hess
- Evotec Asia Pte Ltd, 79 Science Park Drive, #04-05 Cintech IV, Singapore 118264, Singapore
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Campus Gasthuisberg O/N1, Herestraat 49-Bus 802, 3000 Leuven, Belgium
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
4
|
Van Kessel ATM, Cosa G. Lipid-derived electrophiles inhibit the function of membrane channels during ferroptosis. Proc Natl Acad Sci U S A 2024; 121:e2317616121. [PMID: 38743627 PMCID: PMC11127018 DOI: 10.1073/pnas.2317616121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
The therapeutic targeting of ferroptosis requires full understanding of the molecular mechanism of this regulated cell death pathway. While lipid-derived electrophiles (LDEs), including 4-hydroxy-2-nonenal (4-HNE), are important biomarkers of ferroptosis, a functional role for these highly reactive species in ferroptotic cell death execution has not been established. Here, through mechanistic characterization of LDE-detoxification impairment, we demonstrate that LDEs mediate altered protein function during ferroptosis. Applying live cell fluorescence imaging, we first identified that export of glutathione-LDE-adducts through multidrug resistance-associated protein (MRP) channels is inhibited following exposure to a panel of ferroptosis inducers (FINs) with different modes of action (type I-IV FINs erastin, RSL3, FIN56, and FINO2). This channel inhibition was recreated by both initiation of lipid peroxidation and treatment with 4-HNE. Importantly, treatment with radical-trapping antioxidants prevented impaired LDE-adduct export when working with both FINs and lipid peroxidation initiators but not 4-HNE, pinpointing LDEs as the cause of this inhibited MRP activity observed during ferroptosis. Our findings, when combined with reports of widespread LDE alkylation of key proteins following ferroptosis induction, including MRP1, set a precedent for LDEs as critical mediators of ferroptotic cell damage. Lipid hydroperoxide breakdown to form truncated phospholipids and LDEs may fully explain membrane permeabilization and modified protein function downstream of lipid peroxidation, offering a unified explanation of the molecular cell death mechanism of ferroptosis.
Collapse
Affiliation(s)
- Antonius T. M. Van Kessel
- Department of Chemistry, Centre for Structural Biology Research (CRBS) and Quebec Centre for Advanced Materials (QCAM), McGill University, Montreal, QCH3A 0B8, Canada
| | - Gonzalo Cosa
- Department of Chemistry, Centre for Structural Biology Research (CRBS) and Quebec Centre for Advanced Materials (QCAM), McGill University, Montreal, QCH3A 0B8, Canada
| |
Collapse
|
5
|
Poore CP, Wei S, Chen B, Low SW, Tan JSQ, Lee ATH, Nilius B, Liao P. In vivo evaluation of monoclonal antibody M4M using a humanised rat model of stroke demonstrates attenuation of reperfusion injury via blocking human TRPM4 channel. J Drug Target 2024; 32:413-422. [PMID: 38345028 DOI: 10.1080/1061186x.2024.2313522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/23/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Blocking Transient Receptor Potential Melastatin 4 (TRPM4) in rodents by our antibody M4P has shown to attenuate cerebral ischaemia-reperfusion injury. Since M4P does not interact with human TRPM4, the therapeutic potential of blocking human TRPM4 remains unclear. We developed a monoclonal antibody M4M that inhibited human TRPM4 in cultured cells. However, M4M has no effect on stroke outcome in wild-type rats. Therefore, M4M needs to be evaluated on animal models expressing human TRPM4. METHODS We generated a humanised rat model using the CRISPR/Cas technique to knock-in (KI) the human TRPM4 antigen sequence. RESULTS In primary neurons from human TRPM4 KI rats, M4M binds to hypoxic neurons, but not normoxic nor wild-type neurons. Electrophysiological studies showed that M4M blocked ATP depletion-induced activation of TRPM4 and inhibited hypoxia-associated cell volume increase. In a stroke model, administration of M4M reduced infarct volume in KI rats. Rotarod test and Neurological deficit score revealed improvement following M4M treatment. CONCLUSION M4M selectively binds and inhibits hypoxia-induced human TRPM4 channel activation in neurons from the humanised rat model, with no effect on healthy neurons. Use of M4M in stroke rats showed functional improvements, suggesting the potential for anti-human TRPM4 antibodies in treating acute ischaemic stroke patients.
Collapse
Affiliation(s)
- Charlene Priscilla Poore
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Shunhui Wei
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Bo Chen
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - See Wee Low
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Jeslyn Si Qi Tan
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Andy Thiam-Huat Lee
- Health and Social Sciences, Singapore Institute of Technology, Singapore, Singapore
| | - Bernd Nilius
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ping Liao
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
- Health and Social Sciences, Singapore Institute of Technology, Singapore, Singapore
- Graduate Medical School, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
6
|
Poore CP, Hazalin NAMN, Wei S, Low SW, Chen B, Nilius B, Hassan Z, Liao P. TRPM4 blocking antibody reduces neuronal excitotoxicity by specifically inhibiting glutamate-induced calcium influx under chronic hypoxia. Neurobiol Dis 2024; 191:106408. [PMID: 38199274 DOI: 10.1016/j.nbd.2024.106408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 01/12/2024] Open
Abstract
Excitotoxicity arises from unusually excessive activation of excitatory amino acid receptors such as glutamate receptors. Following an energy crisis, excitotoxicity is a major cause for neuronal death in neurological disorders. Many glutamate antagonists have been examined for their efficacy in mitigating excitotoxicity, but failed to generate beneficial outcome due to their side effects on healthy neurons where glutamate receptors are also blocked. In this study, we found that during chronic hypoxia there is upregulation and activation of a nonselective cation channel TRPM4 that contributes to the depolarized neuronal membrane potential and enhanced glutamate-induced calcium entry. TRPM4 is involved in modulating neuronal membrane excitability and calcium signaling, with a complex and multifaceted role in the brain. Here, we inhibited TRPM4 using a newly developed blocking antibody M4P, which could repolarize the resting membrane potential and ameliorate calcium influx upon glutamate stimulation. Importantly, M4P did not affect the functions of healthy neurons as the activity of TRPM4 channel is not upregulated under normoxia. Using a rat model of chronic hypoxia with both common carotid arteries occluded, we found that M4P treatment could reduce apoptosis in the neurons within the hippocampus, attenuate long-term potentiation impairment and improve the functions of learning and memory in this rat model. With specificity to hypoxic neurons, TRPM4 blocking antibody can be a novel way of controlling excitotoxicity with minimal side effects that are common among direct blockers of glutamate receptors.
Collapse
Affiliation(s)
- Charlene P Poore
- Calcium Signaling Laboratory, National Neuroscience Institute, 308433, Singapore
| | - Nurul A M N Hazalin
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Puncak Alam, 42300, Selangor, Malaysia; Centre for Drug Research, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Shunhui Wei
- Calcium Signaling Laboratory, National Neuroscience Institute, 308433, Singapore
| | - See Wee Low
- Calcium Signaling Laboratory, National Neuroscience Institute, 308433, Singapore
| | - Bo Chen
- Calcium Signaling Laboratory, National Neuroscience Institute, 308433, Singapore
| | - Bernd Nilius
- Department Molecular Cell Biology, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, 11800 Penang, Malaysia.
| | - Ping Liao
- Calcium Signaling Laboratory, National Neuroscience Institute, 308433, Singapore.
| |
Collapse
|
7
|
Wei S, Chen B, Low SW, Poore CP, Gao Y, Nilius B, Liao P. SLC26A11 Inhibition Reduces Oncotic Neuronal Death and Attenuates Stroke Reperfusion Injury. Mol Neurobiol 2023; 60:5931-5943. [PMID: 37380823 PMCID: PMC10471688 DOI: 10.1007/s12035-023-03453-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Neuronal swelling is a pathological feature of stroke which contributes to the formation of cytotoxic edema. Under hypoxic condition, aberrant accumulation of sodium and chloride ions inside neurons increases osmotic pressure, leading to cell volume increase. Sodium entry pathway in neurons has been studied extensively. Here, we determine whether SLC26A11 is the major chloride entry pathway under hypoxia and could be the target for protection against ischemic stroke. In this study, electrophysiological properties of chloride current in primary cultured neurons were characterized using low chloride solution, 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid, and SLC26A11-specific siRNA under physiological conditions or ATP-depleted conditions. In vivo effect of SLC26A11 was evaluated on a rat stroke reperfusion model. We found that SLC26A11 mRNA in primary cultured neurons was upregulated as early as 6 h after oxygen glucose deprivation, and later, the protein level was elevated accordingly. Blockade of SLC26A11 activity could reduce chloride entry and attenuate hypoxia-induced neuronal swelling. In the animal stroke model, SLC26A11 upregulation was mainly located in surviving neurons close to the infarct core. SLC26A11 inhibition ameliorates infarct formation and improves functional recovery. These findings demonstrate that SLC26A11 is a major pathway for chloride entry in stroke, contributing to neuronal swelling. Inhibition of SLC26A11 could be a novel therapeutic strategy for stroke.
Collapse
Affiliation(s)
- Shunhui Wei
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Bo Chen
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - See Wee Low
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Charlene Priscilla Poore
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Yahui Gao
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Present Address: Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 119077 Singapore
| | - Bernd Nilius
- Department of Cellular and Molecular Medicine, KU Leuven, 3000 Louvain, Belgium
| | - Ping Liao
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Duke-NUS Medical School, Singapore, 169857 Singapore
- Health and Social Sciences, Singapore Institute of Technology, Singapore, 138683 Singapore
| |
Collapse
|
8
|
Zhang M, Ma Y, Ye X, Zhang N, Pan L, Wang B. TRP (transient receptor potential) ion channel family: structures, biological functions and therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:261. [PMID: 37402746 DOI: 10.1038/s41392-023-01464-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/26/2023] [Accepted: 04/25/2023] [Indexed: 07/06/2023] Open
Abstract
Transient receptor potential (TRP) channels are sensors for a variety of cellular and environmental signals. Mammals express a total of 28 different TRP channel proteins, which can be divided into seven subfamilies based on amino acid sequence homology: TRPA (Ankyrin), TRPC (Canonical), TRPM (Melastatin), TRPML (Mucolipin), TRPN (NO-mechano-potential, NOMP), TRPP (Polycystin), TRPV (Vanilloid). They are a class of ion channels found in numerous tissues and cell types and are permeable to a wide range of cations such as Ca2+, Mg2+, Na+, K+, and others. TRP channels are responsible for various sensory responses including heat, cold, pain, stress, vision and taste and can be activated by a number of stimuli. Their predominantly location on the cell surface, their interaction with numerous physiological signaling pathways, and the unique crystal structure of TRP channels make TRPs attractive drug targets and implicate them in the treatment of a wide range of diseases. Here, we review the history of TRP channel discovery, summarize the structures and functions of the TRP ion channel family, and highlight the current understanding of the role of TRP channels in the pathogenesis of human disease. Most importantly, we describe TRP channel-related drug discovery, therapeutic interventions for diseases and the limitations of targeting TRP channels in potential clinical applications.
Collapse
Affiliation(s)
- Miao Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yueming Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xianglu Ye
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ning Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lei Pan
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
9
|
Chen B, Wei S, Low SW, Poore CP, Lee ATH, Nilius B, Liao P. TRPM4 Blocking Antibody Protects Cerebral Vasculature in Delayed Stroke Reperfusion. Biomedicines 2023; 11:biomedicines11051480. [PMID: 37239151 DOI: 10.3390/biomedicines11051480] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Reperfusion therapy for acute ischemic stroke aims to restore the blood flow of occluded blood vessels. However, successful recanalization is often associated with disruption of the blood-brain barrier, leading to reperfusion injury. Delayed recanalization increases the risk of severe reperfusion injury, including severe cerebral edema and hemorrhagic transformation. The TRPM4-blocking antibody M4P has been shown to alleviate reperfusion injury and improve functional outcomes in animal models of early stroke reperfusion. In this study, we examined the role of M4P in a clinically relevant rat model of delayed stroke reperfusion in which the left middle cerebral artery was occluded for 7 h. To mimic the clinical scenario, M4P or control IgG was administered 1 h before recanalization. Immunostaining showed that M4P treatment improved vascular morphology after stroke. Evans blue extravasation demonstrated attenuated vascular leakage following M4P treatment. With better vascular integrity, cerebral perfusion was improved, leading to a reduction of infarct volume and animal mortality rate. Functional outcome was evaluated by the Rotarod test. As more animals with severe injuries died during the test in the control IgG group, we observed no difference in functional outcomes in the surviving animals. In conclusion, we identified the potential of TRPM4 blocking antibody M4P to ameliorate vascular injury during delayed stroke reperfusion. If combined with reperfusion therapy, M4P has the potential to improve current stroke management.
Collapse
Affiliation(s)
- Bo Chen
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore 308433, Singapore
| | - Shunhui Wei
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore 308433, Singapore
| | - See Wee Low
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore 308433, Singapore
| | - Charlene Priscilla Poore
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore 308433, Singapore
| | - Andy Thiam-Huat Lee
- Health and Social Sciences, Singapore Institute of Technology, Singapore 138683, Singapore
| | - Bernd Nilius
- Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Ping Liao
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, Singapore 308433, Singapore
- Health and Social Sciences, Singapore Institute of Technology, Singapore 138683, Singapore
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
10
|
Boukenna M, Rougier JS, Aghagolzadeh P, Pradervand S, Guichard S, Hämmerli AF, Pedrazzini T, Abriel H. Multiomics uncover the proinflammatory role of Trpm4 deletion after myocardial infarction in mice. Am J Physiol Heart Circ Physiol 2023; 324:H504-H518. [PMID: 36800508 DOI: 10.1152/ajpheart.00671.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Upon myocardial infarction (MI), ischemia-induced cell death triggers an inflammatory response responsible for removing necrotic material and inducing tissue repair. TRPM4 is a Ca2+-activated ion channel permeable to monovalent cations. Although its role in cardiomyocyte-driven hypertrophy and arrhythmia post-MI has been established, no study has yet investigated its role in the inflammatory process orchestrated by endothelial cells, immune cells, and fibroblasts. This study aims to assess the role of TRPM4 in 1) survival and cardiac function, 2) inflammation, and 3) healing post-MI. We performed ligation of the left coronary artery or sham intervention on 154 Trpm4 WT or KO mice under isoflurane anesthesia. Survival and echocardiographic functions were monitored up to 5 wk. We collected serum during the acute post-MI phase to analyze proteomes and performed single-cell RNA sequencing on nonmyocytic cells of hearts after 24 and 72 h. Lastly, we assessed chronic fibrosis and angiogenesis. We observed no significant differences in survival or cardiac function, even though our proteomics data showed significantly decreased tissue injury markers (i.e., creatine kinase M and VE-cadherin) in KO serum after 12 h. On the other hand, inflammation, characterized by serum amyloid P component in the serum, higher number of recruited granulocytes, inflammatory monocytes, and macrophages, as well as expression of proinflammatory genes, was significantly higher in KO. This correlated with increased chronic cardiac fibrosis and angiogenesis. Since inflammation and fibrosis are closely linked to adverse remodeling, future therapeutic attempts at inhibiting TRPM4 will need to assess these parameters carefully before proceeding with translational studies.NEW & NOTEWORTHY Deletion of Trpm4 increases markers of cardiac and systemic inflammation within the first 24 h after MI, while inducing an earlier fibrotic transition at 72 h and more overall chronic fibrosis and angiogenesis at 5 wk. The descriptive, robust, and methodologically broad approach of this study sheds light on an important caveat that will need to be taken into account in all future therapeutic attempts to inhibit TRPM4 post-MI.
Collapse
Affiliation(s)
- Mey Boukenna
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
- Department of Cardiology, Bern University Hospital, Inselspital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Jean-Sébastien Rougier
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Parisa Aghagolzadeh
- Experimental Cardiology Unit, Department of Cardiovascular Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | - Sylvain Pradervand
- Centre d'Oncologie de Précision, Département d'Oncologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Sabrina Guichard
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Anne-Flore Hämmerli
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Thierry Pedrazzini
- Experimental Cardiology Unit, Department of Cardiovascular Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | - Hugues Abriel
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| |
Collapse
|
11
|
Ma P, Huang N, Tang J, Zhou Z, Xu J, Chen Y, Zhang M, Huang Q, Cheng Y. The TRPM4 channel inhibitor 9-phenanthrol alleviates cerebral edema after traumatic brain injury in rats. Front Pharmacol 2023; 14:1098228. [PMID: 36865920 PMCID: PMC9971592 DOI: 10.3389/fphar.2023.1098228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Cerebral edema (CE) exerts an important effect on brain injury after traumatic brain injury (TBI). Upregulation of transient receptor potential melastatin 4 (TRPM4) in vascular endothelial cells (ECs) results in damage to capillaries and the blood-brain barrier (BBB), which is critical for the development of CE. Many studies have shown that 9-phenanthrol (9-PH) effectively inhibits TRPM4. The current study aimed to investigate the effect of 9-PH on reducing CE after TBI. In this experiment, we observed that 9-PH markedly reduced brain water content, BBB disruption, proliferation of microglia and astrocytes, neutrophil infiltration, neuronal apoptosis and neurobehavioral deficits. At the molecular level, 9-PH significantly inhibited the protein expression of TRPM4 and MMP-9, alleviated the expression of apoptosis-related molecules and inflammatory cytokines, such as Bax, TNF-α and IL-6, near injured tissue, and diminished serum SUR1 and TRPM4 levels. Mechanistically, treatment with 9-PH inhibited activation of the PI3K/AKT/NF-kB signaling pathway, which was reported to be involved in the expression of MMP-9. Taken together, the results of this study indicate that 9-PH effectively reduces CE and alleviates secondary brain injury partly through the following possible mechanisms: ①9-PH inhibits TRPM4-mediated Na + influx and reduces cytotoxic CE; ②9-PH hinders the expression and activity of MMP-9 by inhibiting the TRPM4 channel and decreases disruption of the BBB, thereby preventing vasogenic cerebral edema. ③9-PH reduces further inflammatory and apoptotic damage to tissues.
Collapse
Affiliation(s)
- Ping Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ning Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Tang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zunjie Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Chen
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Maoxin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Qin Huang, ; Yuan Cheng,
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Qin Huang, ; Yuan Cheng,
| |
Collapse
|
12
|
Zhou Y, He Y, Yan S, Chen L, Zhang R, Xu J, Hu H, Liebeskind DS, Lou M. Reperfusion Injury Is Associated With Poor Outcome in Patients With Recanalization After Thrombectomy. Stroke 2023; 54:96-104. [PMID: 36367100 DOI: 10.1161/strokeaha.122.039337] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The existence of cerebral reperfusion injury in human beings remains controversial. Thus, we aimed to explore the presence of reperfusion injury in acute ischemic stroke patients with recanalization after mechanical thrombectomy and analyzed its impact on neurological outcome. METHODS We reviewed our prospectively collected database CIPPIS (Comparison Influence to Prognosis of CTP and MRP in AIS Patients, NCT03367286), and enrolled anterior circulation large artery occlusion patients with recanalization after mechanical thrombectomy who underwent (1) computed tomography (CT) perfusion on admission and immediately after recanalization to determine reperfusion region, and (2) CT and/or magnetic resonance imaging (MRI) immediately and 24 hours after recanalization to determine lesion areas. The expansion of lesion between recanalization and 24 hours within reperfusion region was potentially caused by reperfusion, thus termed as radiological observed reperfusion injury (RORI). Based on the imaging modality immediately after recanalization, RORI was further divided into RORICT and RORIMRI. We first included a small cohort who had performed both CT and MRI immediately after recanalization to validate the consistency between RORICT and RORIMRI (Study 1). Then the association with RORICT and poor outcome, defined as 3-month modified Rankin Scale score of 3 to 6, was explored in a larger cohort (Study 2). RESULTS Study 1 included 23 patients and good consistency was found between RORICT and RORIMRI (intraclass correlation=0.97, P<0.001). Among 226 patients included in Study 2, a total of 106 (46.9%) were identified with RORI. The ratio of RORI to reperfusion region was 30.1 (16.2, 51.0)% and was independently associated with poor outcome (odds ratio=1.55 per 10% [95% CI' 1.30-1.84]; P<0.001). CONCLUSIONS Our findings suggested that RORI was relatively frequent in stroke patients with recanalization after mechanical thrombectomy and associated with poor outcome despite successful recanalization. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT03367286.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Yaode He
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Shenqiang Yan
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Lin Chen
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Ruoxia Zhang
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Jinjin Xu
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Haitao Hu
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | | | - Min Lou
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| |
Collapse
|
13
|
Wei S, Behn J, Poore CP, Low SW, Nilius B, Fan H, Liao P. Binding epitope for recognition of human TRPM4 channel by monoclonal antibody M4M. Sci Rep 2022; 12:19562. [PMID: 36380063 PMCID: PMC9666640 DOI: 10.1038/s41598-022-22077-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Mouse monoclonal antibody M4M was recently designed to block human TRPM4 channel. The polypeptide for generating M4M is composed of peptide A1 between the transmembrane segment 5 (S5) and the pore, and a second peptide A2 between the pore and the transmembrane segment 6 (S6). Using peptide microarray, a 4-amino acid sequence EPGF within the A2 was identified to be the binding epitope for M4M. Substitution of EPGF with other amino acids greatly reduced binding affinity. Structural analysis of human TRPM4 structure indicates that EPGF is located externally to the channel pore. A1 is close to the EPGF binding epitope in space, albeit separated by a 37-amino acid peptide. Electrophysiological study reveals that M4M could block human TRPM4, but with no effect on rodent TRPM4 which shares a different amino acid sequence ERGS for the binding motif. Our results demonstrate that M4M is a specific inhibitor for human TRPM4.
Collapse
Affiliation(s)
- Shunhui Wei
- grid.276809.20000 0004 0636 696XCalcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Julian Behn
- grid.418325.90000 0000 9351 8132Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, 138671 Singapore ,grid.4280.e0000 0001 2180 6431Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Charlene Priscilla Poore
- grid.276809.20000 0004 0636 696XCalcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - See Wee Low
- grid.276809.20000 0004 0636 696XCalcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Bernd Nilius
- grid.5596.f0000 0001 0668 7884Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Hao Fan
- grid.418325.90000 0000 9351 8132Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, 138671 Singapore
| | - Ping Liao
- grid.276809.20000 0004 0636 696XCalcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore ,grid.486188.b0000 0004 1790 4399Health and Social Sciences, Singapore Institute of Technology, Singapore, Singapore ,grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
14
|
Gu Y, Zhou C, Piao Z, Yuan H, Jiang H, Wei H, Zhou Y, Nan G, Ji X. Cerebral edema after ischemic stroke: Pathophysiology and underlying mechanisms. Front Neurosci 2022; 16:988283. [PMID: 36061592 PMCID: PMC9434007 DOI: 10.3389/fnins.2022.988283] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022] Open
Abstract
Ischemic stroke is associated with increasing morbidity and has become the main cause of death and disability worldwide. Cerebral edema is a serious complication arising from ischemic stroke. It causes an increase in intracranial pressure, rapid deterioration of neurological symptoms, and formation of cerebral hernia, and is an important risk factor for adverse outcomes after stroke. To date, the detailed mechanism of cerebral edema after stroke remains unclear. This limits advances in prevention and treatment strategies as well as drug development. This review discusses the classification and pathological characteristics of cerebral edema, the possible relationship of the development of cerebral edema after ischemic stroke with aquaporin 4, the SUR1-TRPM4 channel, matrix metalloproteinase 9, microRNA, cerebral venous reflux, inflammatory reactions, and cerebral ischemia/reperfusion injury. It also summarizes research on new therapeutic drugs for post-stroke cerebral edema. Thus, this review provides a reference for further studies and for clinical treatment of cerebral edema after ischemic stroke.
Collapse
Affiliation(s)
- Yuhang Gu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, China
| | - Zhe Piao
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Honghua Yuan
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Huimin Jiang
- Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, China
| | - Huimin Wei
- Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, China
| | - Guangxian Nan
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Guangxian Nan,
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Xunming Ji,
| |
Collapse
|
15
|
Shen Z, Xiang M, Chen C, Ding F, Wang Y, Shang C, Xin L, Zhang Y, Cui X. Glutamate excitotoxicity: Potential therapeutic target for ischemic stroke. Biomed Pharmacother 2022; 151:113125. [PMID: 35609367 DOI: 10.1016/j.biopha.2022.113125] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/01/2022] [Accepted: 05/13/2022] [Indexed: 11/29/2022] Open
Abstract
Glutamate-mediated excitotoxicity is an important mechanism leading to post ischemic stroke damage. After acute stroke, the sudden reduction in cerebral blood flow is most initially followed by ion transport protein dysfunction and disruption of ion homeostasis, which in turn leads to impaired glutamate release, reuptake, and excessive N-methyl-D-aspartate receptor (NMDAR) activation, promoting neuronal death. Despite extensive evidence from preclinical studies suggesting that excessive NMDAR stimulation during ischemic stroke is a central step in post-stroke damage, NMDAR blockers have failed to translate into clinical stroke treatment. Current treatment options for stroke are very limited, and there is therefore a great need to develop new targets for neuroprotective therapeutic agents in ischemic stroke to extend the therapeutic time window. In this review, we highlight recent findings on glutamate release, reuptake mechanisms, NMDAR and its downstream cellular signaling pathways in post-ischemic stroke damage, and review the pathological changes in each link to help develop viable new therapeutic targets. We then also summarize potential neuroprotective drugs and therapeutic approaches for these new targets in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zihuan Shen
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Mi Xiang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chen Chen
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Fan Ding
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Yuling Wang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Chang Shang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Laiyun Xin
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yang Zhang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Xiangning Cui
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
16
|
Kovács ZM, Dienes C, Hézső T, Almássy J, Magyar J, Bányász T, Nánási PP, Horváth B, Szentandrássy N. Pharmacological Modulation and (Patho)Physiological Roles of TRPM4 Channel—Part 1: Modulation of TRPM4. Pharmaceuticals (Basel) 2022; 15:ph15010081. [PMID: 35056138 PMCID: PMC8781449 DOI: 10.3390/ph15010081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
Transient receptor potential melastatin 4 is a unique member of the TRPM protein family and, similarly to TRPM5, is Ca2+-sensitive and permeable to monovalent but not divalent cations. It is widely expressed in many organs and is involved in several functions by regulating the membrane potential and Ca2+ homeostasis in both excitable and non-excitable cells. This part of the review discusses the pharmacological modulation of TRPM4 by listing, comparing, and describing both endogenous and exogenous activators and inhibitors of the ion channel. Moreover, other strategies used to study TRPM4 functions are listed and described. These strategies include siRNA-mediated silencing of TRPM4, dominant-negative TRPM4 variants, and anti-TRPM4 antibodies. TRPM4 is receiving more and more attention and is likely to be the topic of research in the future.
Collapse
Affiliation(s)
- Zsigmond Máté Kovács
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Csaba Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Hézső
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
| | - János Magyar
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Division of Sport Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Bányász
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
| | - Péter P. Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Norbert Szentandrássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence:
| |
Collapse
|
17
|
Alquisiras-Burgos I, Franco-Pérez J, Rubio-Osornio M, Aguilera P. The short form of the SUR1 and its functional implications in the damaged brain. Neural Regen Res 2022; 17:488-496. [PMID: 34380876 PMCID: PMC8504400 DOI: 10.4103/1673-5374.320967] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Sulfonylurea receptor (SUR) belongs to the adenosine 5′-triphosphate (ATP)-binding cassette (ABC) transporter family; however, SUR is associated with ion channels and acts as a regulatory subunit determining the opening or closing of the pore. Abcc8 and Abcc9 genes code for the proteins SUR1 and SUR2, respectively. The SUR1 transcript encodes a protein of 1582 amino acids with a mass around 140–177 kDa expressed in the pancreas, brain, heart, and other tissues. It is well known that SUR1 assembles with Kir6.2 and TRPM4 to establish KATP channels and non-selective cation channels, respectively. Abbc8 and 9 are alternatively spliced, and the resulting transcripts encode different isoforms of SUR1 and SUR2, which have been detected by different experimental strategies. Interestingly, the use of binding assays to sulfonylureas and Western blotting has allowed the detection of shorter forms of SUR (~65 kDa). Identity of the SUR1 variants has not been clarified, and some authors have suggested that the shorter forms are unspecific. However, immunoprecipitation assays have shown that SUR2 short forms are part of a functional channel even coexisting with the typical forms of the receptor in the heart. This evidence confirms that the structure of the short forms of the SURs is fully functional and does not lose the ability to interact with the channels. Since structural changes in short forms of SUR modify its affinity to ATP, regulation of its expression might represent an advantage in pathologies where ATP concentrations decrease and a therapeutic target to induce neuroprotection. Remarkably, the expression of SUR1 variants might be induced by conditions associated to the decrease of energetic substrates in the brain (e.g. during stroke and epilepsy). In this review, we want to contribute to the knowledge of SUR1 complexity by analyzing evidence that shows the existence of short SUR1 variants and its possible implications in brain function.
Collapse
Affiliation(s)
- Iván Alquisiras-Burgos
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", CDMX, Mexico
| | - Javier Franco-Pérez
- Laboratorio de Formación Reticular, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", CDMX, Mexico
| | - Moisés Rubio-Osornio
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", CDMX, Mexico
| | - Penélope Aguilera
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", CDMX, Mexico
| |
Collapse
|
18
|
Maggi F, Morelli MB, Nabissi M, Marinelli O, Zeppa L, Aguzzi C, Santoni G, Amantini C. Transient Receptor Potential (TRP) Channels in Haematological Malignancies: An Update. Biomolecules 2021; 11:biom11050765. [PMID: 34065398 PMCID: PMC8160608 DOI: 10.3390/biom11050765] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential (TRP) channels are improving their importance in different cancers, becoming suitable as promising candidates for precision medicine. Their important contribution in calcium trafficking inside and outside cells is coming to light from many papers published so far. Encouraging results on the correlation between TRP and overall survival (OS) and progression-free survival (PFS) in cancer patients are available, and there are as many promising data from in vitro studies. For what concerns haematological malignancy, the role of TRPs is still not elucidated, and data regarding TRP channel expression have demonstrated great variability throughout blood cancer so far. Thus, the aim of this review is to highlight the most recent findings on TRP channels in leukaemia and lymphoma, demonstrating their important contribution in the perspective of personalised therapies.
Collapse
Affiliation(s)
- Federica Maggi
- Department of Molecular Medicine, Sapienza University, 00185 Rome, Italy;
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Maria Beatrice Morelli
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Massimo Nabissi
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Oliviero Marinelli
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Laura Zeppa
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Cristina Aguzzi
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Giorgio Santoni
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Consuelo Amantini
- Immunopathology Laboratory, School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
- Correspondence: ; Tel.: +30-0737403312
| |
Collapse
|
19
|
Low SW, Gao Y, Wei S, Chen B, Nilius B, Liao P. Development and characterization of a monoclonal antibody blocking human TRPM4 channel. Sci Rep 2021; 11:10411. [PMID: 34002002 PMCID: PMC8129085 DOI: 10.1038/s41598-021-89935-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 05/04/2021] [Indexed: 02/06/2023] Open
Abstract
TRPM4 is a calcium-activated non-selective monovalent cation channel implicated in diseases such as stroke. Lack of potent and selective inhibitors remains a major challenge for studying TRPM4. Using a polypeptide from rat TRPM4, we have generated a polyclonal antibody M4P which could alleviate reperfusion injury in a rat model of stroke. Here, we aim to develop a monoclonal antibody that could block human TRPM4 channel. Two mouse monoclonal antibodies M4M and M4M1 were developed to target an extracellular epitope of human TRPM4. Immunohistochemistry and western blot were used to characterize the binding of these antibodies to human TRPM4. Potency of inhibition was compared using electrophysiological methods. We further evaluated the therapeutic potential on a rat model of middle cerebral artery occlusion. Both M4M and M4M1 could bind to human TRPM4 channel on the surface of live cells. Prolonged incubation with TRPM4 blocking antibody internalized surface TRPM4. Comparing to M4M1, M4M is more effective in blocking human TRPM4 channel. In human brain microvascular endothelial cells, M4M successfully inhibited TRPM4 current and ameliorated hypoxia-induced cell swelling. Using wild type rats, neither antibody demonstrated therapeutic potential on stroke. Human TRPM4 channel can be blocked by a monoclonal antibody M4M targeting a key antigenic sequence. For future clinical translation, the antibody needs to be humanized and a transgenic animal carrying human TRPM4 sequence is required for in vivo characterizing its therapeutic potential.
Collapse
Affiliation(s)
- See Wee Low
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - Yahui Gao
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - Shunhui Wei
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - Bo Chen
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - Bernd Nilius
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ping Liao
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore. .,Duke-NUS Medical School, Singapore, Singapore. .,Health and Social Sciences, Singapore Institute of Technology, Singapore, Singapore.
| |
Collapse
|
20
|
Feng J, Zong P, Yan J, Yue Z, Li X, Smith C, Ai X, Yue L. Upregulation of transient receptor potential melastatin 4 (TRPM4) in ventricular fibroblasts from heart failure patients. Pflugers Arch 2021; 473:521-531. [PMID: 33594499 PMCID: PMC8857941 DOI: 10.1007/s00424-021-02525-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/19/2022]
Abstract
The transient receptor potential melastatin 4 (TRPM4) is a Ca2+-activated nonselective monovalent cation channel belonging to the TRP channel superfamily. TRPM4 is widely expressed in various tissues and most abundantly expressed in the heart. TRPM4 plays a critical role in cardiac conduction. Patients carrying a gain-of-function or loss-of-function mutation of TRPM4 display impaired cardiac conduction. Knockout or over-expression of TRPM4 in mice recapitulates conduction defects in patients. Moreover, recent studies have indicated that TRPM4 plays a role in hypertrophy and heart failure. Whereas the role of TRPM4 mediated by cardiac myocytes has been well investigated, little is known about TRPM4 and its role in cardiac fibroblasts. Here we show that in human left ventricular fibroblasts, TRPM4 exhibits typical Ca2+-activation characteristics, linear current-voltage (I-V) relation, and monovalent permeability. TRPM4 currents recorded in fibroblasts from heart failure patients (HF) are more than 2-fold bigger than those from control individuals (CTL). The enhanced functional TRPM4 in HF is not resulted from changed channel properties, as TRPM4 currents from both HF and CTL fibroblasts demonstrate similar sensitivity to intracellular calcium activation and extracellular 9-phenanthrol (9-phen) blockade. Consistent with enhanced TRPM4 activity, the protein level of TRPM4 is about 2-fold higher in HF than that of CTL hearts. Moreover, TRPM4 current in CTL fibroblasts is increased after 24 hours of TGFβ1 treatment, implying that TRPM4 in vivo may be upregulated by fibrogenesis promotor TGFβ1. The upregulated TRPM4 in HF fibroblasts suggests that TRPM4 may play a role in cardiac fibrogenesis under various pathological conditions.
Collapse
Affiliation(s)
- Jianlin Feng
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut School of Medicine (UConn Health), Farmington, CT, 06030, USA
| | - Pengyu Zong
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut School of Medicine (UConn Health), Farmington, CT, 06030, USA
| | - Jiajie Yan
- Department of Physiology and Biophysics, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Zhichao Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut School of Medicine (UConn Health), Farmington, CT, 06030, USA
| | - Xin Li
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut School of Medicine (UConn Health), Farmington, CT, 06030, USA
| | - Chevaughn Smith
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut School of Medicine (UConn Health), Farmington, CT, 06030, USA
| | - Xun Ai
- Department of Physiology and Biophysics, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Lixia Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut School of Medicine (UConn Health), Farmington, CT, 06030, USA.
| |
Collapse
|
21
|
Borgström A, Peinelt C, Stokłosa P. TRPM4 in Cancer-A New Potential Drug Target. Biomolecules 2021; 11:biom11020229. [PMID: 33562811 PMCID: PMC7914809 DOI: 10.3390/biom11020229] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 12/11/2022] Open
Abstract
Transient receptor potential melastatin 4 (TRPM4) is widely expressed in various organs and associated with cardiovascular and immune diseases. Lately, the interest in studies on TRPM4 in cancer has increased. Thus far, TRPM4 has been investigated in diffuse large B-cell lymphoma, prostate, colorectal, liver, breast, urinary bladder, cervical, and endometrial cancer. In several types of cancer TRPM4 is overexpressed and contributes to cancer hallmark functions such as increased proliferation and migration and cell cycle shift. Hence, TRPM4 is a potential prognostic cancer marker and a promising anticancer drug target candidate. Currently, the underlying mechanism by which TRPM4 contributes to cancer hallmark functions is under investigation. TRPM4 is a Ca2+-activated monovalent cation channel, and its ion conductivity can decrease intracellular Ca2+ signaling. Furthermore, TRPM4 can interact with different partner proteins. However, the lack of potent and specific TRPM4 inhibitors has delayed the investigations of TRPM4. In this review, we summarize the potential mechanisms of action and discuss new small molecule TRPM4 inhibitors, as well as the TRPM4 antibody, M4P. Additionally, we provide an overview of TRPM4 in human cancer and discuss TRPM4 as a diagnostic marker and anticancer drug target.
Collapse
|
22
|
Inherited Cardiac Arrhythmia Syndromes: Focus on Molecular Mechanisms Underlying TRPM4 Channelopathies. Cardiovasc Ther 2020; 2020:6615038. [PMID: 33381229 PMCID: PMC7759408 DOI: 10.1155/2020/6615038] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
The Transient Receptor Potential Melastatin 4 (TRPM4) is a transmembrane N-glycosylated ion channel that belongs to the large family of TRP proteins. It has an equal permeability to Na+ and K+ and is activated via an increase of the intracellular calcium concentration and membrane depolarization. Due to its wide distribution, TRPM4 dysfunction has been linked with several pathophysiological processes, including inherited cardiac arrhythmias. Many pathogenic variants of the TRPM4 gene have been identified in patients with different forms of cardiac disorders such as conduction defects, Brugada syndrome, and congenital long QT syndrome. At the cellular level, these variants induce either gain- or loss-of-function of TRPM4 channels for similar clinical phenotypes. However, the molecular mechanisms associating these functional alterations to the clinical phenotypes remain poorly understood. The main objective of this article is to review the major cardiac TRPM4 channelopathies and recent advances regarding their genetic background and the underlying molecular mechanisms.
Collapse
|
23
|
Lavanderos B, Silva I, Cruz P, Orellana-Serradell O, Saldías MP, Cerda O. TRP Channels Regulation of Rho GTPases in Brain Context and Diseases. Front Cell Dev Biol 2020; 8:582975. [PMID: 33240883 PMCID: PMC7683514 DOI: 10.3389/fcell.2020.582975] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Neurological and neuropsychiatric disorders are mediated by several pathophysiological mechanisms, including developmental and degenerative abnormalities caused primarily by disturbances in cell migration, structural plasticity of the synapse, and blood-vessel barrier function. In this context, critical pathways involved in the pathogenesis of these diseases are related to structural, scaffolding, and enzymatic activity-bearing proteins, which participate in Ca2+- and Ras Homologs (Rho) GTPases-mediated signaling. Rho GTPases are GDP/GTP binding proteins that regulate the cytoskeletal structure, cellular protrusion, and migration. These proteins cycle between GTP-bound (active) and GDP-bound (inactive) states due to their intrinsic GTPase activity and their dynamic regulation by GEFs, GAPs, and GDIs. One of the most important upstream inputs that modulate Rho GTPases activity is Ca2+ signaling, positioning ion channels as pivotal molecular entities for Rho GTPases regulation. Multiple non-selective cationic channels belonging to the Transient Receptor Potential (TRP) family participate in cytoskeletal-dependent processes through Ca2+-mediated modulation of Rho GTPases. Moreover, these ion channels have a role in several neuropathological events such as neuronal cell death, brain tumor progression and strokes. Although Rho GTPases-dependent pathways have been extensively studied, how they converge with TRP channels in the development or progression of neuropathologies is poorly understood. Herein, we review recent evidence and insights that link TRP channels activity to downstream Rho GTPase signaling or modulation. Moreover, using the TRIP database, we establish associations between possible mediators of Rho GTPase signaling with TRP ion channels. As such, we propose mechanisms that might explain the TRP-dependent modulation of Rho GTPases as possible pathways participating in the emergence or maintenance of neuropathological conditions.
Collapse
Affiliation(s)
- Boris Lavanderos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Ian Silva
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Pablo Cruz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Octavio Orellana-Serradell
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - María Paz Saldías
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
24
|
Wei S, Low SW, Poore CP, Chen B, Gao Y, Nilius B, Liao P. Comparison of Anti-oncotic Effect of TRPM4 Blocking Antibody in Neuron, Astrocyte and Vascular Endothelial Cell Under Hypoxia. Front Cell Dev Biol 2020; 8:562584. [PMID: 33195194 PMCID: PMC7604339 DOI: 10.3389/fcell.2020.562584] [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] [Received: 05/15/2020] [Accepted: 09/30/2020] [Indexed: 12/31/2022] Open
Abstract
In stroke and other neurological diseases, Transient Receptor Potential Melastatin 4 (TRPM4) has been reported to cause oncotic cell death which is due to an excessive influx of sodium ions. Following stroke, hypoxia condition activates TRPM4 channel, and the sodium influx via TRPM4 is further enhanced by an increased TRPM4 expression. However, the effect of TRPM4 inhibition on oncotic cell death, particularly during the acute stage, remains largely unknown. Recently, we have developed a polyclonal antibody M4P that specifically inhibits TRPM4 channel. M4P blocks the channel via binding to a region close to the channel pore from extracellular space. Using M4P, we evaluated the acute effect of blocking TRPM4 in neurons, astrocytes, and vascular endothelial cells. In a rat stroke model, M4P co-localized with neuronal marker NeuN and endothelial marker vWF, whereas few GFAP positive astrocytes were stained by M4P in the ipsilateral hemisphere. When ATP was acutely depleted in cultured cortical neurons and microvascular endothelial cells, cell swelling was induced. Application of M4P significantly blocked TRPM4 current and attenuated oncosis. TUNEL assay, PI staining and western blot on cleaved Caspase-3 revealed that M4P could ameliorate apoptosis after 24 h hypoxia exposure. In contrast, acute ATP depletion in cultured astrocytes failed to demonstrate an increase of cell volume, and application of M4P or control IgG had no effect on cell volume change. When TRPM4 was overexpressed in astrocytes, acute ATP depletion successfully induced oncosis which could be suppressed by M4P treatment. Our results demonstrate that comparing to astrocytes, neurons, and vascular endothelial cells are more vulnerable to hypoxic injury. During the acute stage of stroke, blocking TRPM4 channel could protect neurons and vascular endothelial cells from oncotic cell death.
Collapse
Affiliation(s)
- Shunhui Wei
- Calcium Signaling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - See Wee Low
- Calcium Signaling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Charlene Priscilla Poore
- Calcium Signaling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Bo Chen
- Calcium Signaling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Yahui Gao
- Calcium Signaling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Bernd Nilius
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ping Liao
- Calcium Signaling Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore.,Health and Social Sciences, Singapore Institute of Technology, Singapore, Singapore
| |
Collapse
|
25
|
Wong KK, Hussain FA. TRPM4 is overexpressed in breast cancer associated with estrogen response and epithelial-mesenchymal transition gene sets. PLoS One 2020; 15:e0233884. [PMID: 32484822 PMCID: PMC7266295 DOI: 10.1371/journal.pone.0233884] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022] Open
Abstract
Ion channels form an important class of drug targets in malignancies. Transient receptor potential cation channel subfamily M member 4 (TRPM4) plays oncological roles in various solid tumors. Herein, we examined TRPM4 protein expression profile by immunohistochemistry (IHC) in breast cancer cases compared with normal breast ducts, its association with clinico-demographical parameters, and its potential function in breast cancers by Gene Set Enrichment Analysis (GSEA). Data-mining demonstrated that TRPM4 transcript levels were significantly higher in The Cancer Genome Atlas series of breast cancer cases (n = 1,085) compared with normal breast tissues (n = 112) (p = 1.03 x 10−11). Our IHC findings in tissue microarrays showed that TRPM4 protein was overexpressed in breast cancers (n = 83/99 TRPM4+; 83.8%) compared with normal breast ducts (n = 5/10 TRPM4+; 50%) (p = 0.022). Higher TRPM4 expression (median frequency cut-off) was significantly associated with higher lymph node status (N1-N2 vs N0; p = 0.024) and higher stage (IIb-IIIb vs I-IIa; p = 0.005). GSEA evaluation in three independent gene expression profiling (GEP) datasets of breast cancer cases (GSE54002, n = 417; GSE20685, n = 327; GSE23720, n = 197) demonstrated significant association of TRPM4 transcript expression with estrogen response and epithelial-mesenchymal transition (EMT) gene sets (p<0.01 and false discovery rate<0.05). These gene sets were not enriched in GEP datasets of normal breast epithelium cases (GSE10797, n = 5; GSE9574, n = 15; GSE20437, n = 18). In conclusion, TRPM4 protein expression is upregulated in breast cancers associated with worse clinico-demographical parameters, and TRPM4 potentially regulates estrogen receptor signaling and EMT progression in breast cancer.
Collapse
Affiliation(s)
- Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- * E-mail:
| | - Faezahtul Arbaeyah Hussain
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
26
|
Alquisiras-Burgos I, Ortiz-Plata A, Franco-Pérez J, Millán A, Aguilera P. Resveratrol reduces cerebral edema through inhibition of de novo SUR1 expression induced after focal ischemia. Exp Neurol 2020; 330:113353. [PMID: 32380020 DOI: 10.1016/j.expneurol.2020.113353] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/26/2020] [Accepted: 05/01/2020] [Indexed: 12/21/2022]
Abstract
Cerebral edema is a clinical problem that frequently follows ischemic infarcts. Sulfonylurea receptor 1 (SUR1) is an inducible protein that can form a heteromultimeric complex with aquaporin 4 (AQP4) that mediate the ion/water transport involved in brain tissue swelling. Transcription of the Abcc8 gene coding for SUR1 depends on the activity of transcriptional factor SP1, which is modulated by the cellular redox environment. Since oxidative stress is implicated in the induced neuronal damage in ischemia and edema formation, the present study aimed to evaluate if the antioxidant resveratrol (RSV) prevents the damage by reducing the de novo expression of SUR1 in the ischemic brain. Male Wistar rats were subjected to 2 h of middle cerebral artery occlusion followed by different times of reperfusion. RSV (1.9 mg/kg; i.v.) was administered at the onset of reperfusion. Brain damage and edema formation were recognized by neurological evaluation, time of survival, TTC (2,3,5-Triphenyltetrazolium chloride) staining, Evans blue extravasation, and water content. RSV mechanism of action was studied by SP1 binding activity measured through the Electrophoretic Mobility Shift Assay, and Abcc8 and Aqp4 gene expression evaluated by qPCR, immunofluorescence, and Western blot. We found that RSV reduced the infarct area and cerebral edema, prevented blood-brain barrier damage, improved neurological performance, and increased survival. Additionally, our findings suggest that the antioxidant activity of RSV targeted SP transcription factors and inhibited SUR1 and AQP4 expression. Thus, RSV by decreasing SUR1 expression could contribute to reducing edema formation, constituting a therapeutic alternative for edema reduction in stroke.
Collapse
Affiliation(s)
- Iván Alquisiras-Burgos
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, CDMX 14269, México
| | - Alma Ortiz-Plata
- Laboratorio de Neuropatología Experimental, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, CDMX 14269, México.
| | - Javier Franco-Pérez
- Laboratorio de Formación Reticular, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, CDMX 14269, México.
| | - Alejandro Millán
- Posgrado en Ciencias Biomédicas, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Lázaro Cárdenas s/n Ciudad Universitaria, Chilpancingo, Guerrero, 39070, México
| | - Penélope Aguilera
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, CDMX 14269, México.
| |
Collapse
|