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
|
Zong P, Legere N, Feng J, Yue L. TRP Channels in Excitotoxicity. Neuroscientist 2025; 31:80-97. [PMID: 38682490 DOI: 10.1177/10738584241246530] [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] [Indexed: 05/01/2024]
Abstract
Glutamate excitotoxicity is a central mechanism contributing to cellular dysfunction and death in various neurological disorders and diseases, such as stroke, traumatic brain injury, epilepsy, schizophrenia, addiction, mood disorders, Huntington's disease, Alzheimer's disease, Parkinson's disease, multiple sclerosis, pathologic pain, and even normal aging-related changes. This detrimental effect emerges from glutamate binding to glutamate receptors, including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, N-methyl-d-aspartate receptors, kainate receptors, and GluD receptors. Thus, excitotoxicity could be prevented by targeting glutamate receptors and their downstream signaling pathways. However, almost all the glutamate receptor antagonists failed to attenuate excitotoxicity in human patients, mainly due to the limited understanding of the underlying mechanisms regulating excitotoxicity. Transient receptor potential (TRP) channels serve as ancient cellular sensors capable of detecting and responding to both external and internal stimuli. The study of human TRP channels has flourished in recent decades since the initial discovery of mammalian TRP in 1995. These channels have been found to play pivotal roles in numerous pathologic conditions, including excitotoxicity. In this review, our focus centers on exploring the intricate interactions between TRP channels and glutamate receptors in excitotoxicity.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA
| | - Nicholas Legere
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| |
Collapse
|
3
|
Chang Y, He Y, Wang D, Zhang K, Zhang Y, Li Z, Zeng S, Xiao S, Pan S, Huang K. ROS-regulated SUR1-TRPM4 drives persistent activation of NLRP3 inflammasome in microglia after whole-brain radiation. Acta Neuropathol Commun 2025; 13:16. [PMID: 39871308 PMCID: PMC11771008 DOI: 10.1186/s40478-025-01932-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/18/2025] [Indexed: 01/29/2025] Open
Abstract
Delayed radiation-induced brain injury (RIBI) characterized by progressive cognitive decline significantly impacts patient outcomes after radiotherapy. The activation of NLRP3 inflammasome within microglia after brain radiation is involved in the progression of RIBI by mediating inflammatory responses. We have previously shown that sulfonylurea receptor 1-transient receptor potential M4 (SUR1-TRPM4) mediates microglial NLRP3-related inflammation following global brain ischemia. However, the role of SUR1-TRPM4 in RIBI remains unclear. Here, we found that whole-brain radiation induced up-regulation and assembly of SUR1-TRPM4, which further activated the NLRP3 inflammasome in microglia and caused persistent neuroinflammation in mice. Blocking SUR1-TRPM4 by glibenclamide or gene deletion of Trpm4 effectively prevented NLRP3-mediated neuroinflammation and alleviated RIBI. Utilizing the mouse model of RIBI and irradiated BV2 cells, we further demonstrated that irradiation caused mitochondrial damage to microglia, leading to violent release of reactive oxygen species (ROS), which enhanced the transcription of SUR1, TRPM4, and NLRP3 inflammasome-related molecules. Moreover, ROS up-regulated ten-eleven translocation 2 (TET2) to enhance TRPM4 expression by mediating the demethylation of the gene promoter, thereby facilitating the assembly of SUR1-TRPM4 in microglia. In summary, this study deciphers that SUR1-TRPM4 crucially mediates the persistent activation of microglial NLRP3 inflammasome under the action of ROS after whole-brain radiation, offering novel therapeutic strategies for delayed RIBI as well as other NLRP3-related neurological disorders involving excessive ROS production.
Collapse
Affiliation(s)
- Yuan Chang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yihua He
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Di Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Kunxue Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuzhen Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhentong Li
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuxin Zeng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sheng Xiao
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Kaibin Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Neurology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China.
| |
Collapse
|
4
|
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
|
5
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 PMCID: PMC11641818 DOI: 10.3390/ijms252312637] [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: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
6
|
Fazio L, Naik VN, Therpurakal RN, Gomez Osorio FM, Rychlik N, Ladewig J, Strüber M, Cerina M, Meuth SG, Budde T. Retigabine, a potassium channel opener, restores thalamocortical neuron functionality in a murine model of autoimmune encephalomyelitis. Brain Behav Immun 2024; 122:202-215. [PMID: 39142423 DOI: 10.1016/j.bbi.2024.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 08/07/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Multiple Sclerosis (MS) is an autoimmune neurodegenerative disease, whose primary hallmark is the occurrence of inflammatory lesions in white and grey matter structures. Increasing evidence in MS patients and respective murine models reported an impaired ionic homeostasis driven by inflammatory-demyelination, thereby profoundly affecting signal propagation. However, the impact of a focal inflammatory lesion on single-cell and network functionality has hitherto not been fully elucidated. OBJECTIVES In this study, we sought to determine the consequences of a localized cortical inflammatory lesion on the excitability and firing pattern of thalamic neurons in the auditory system. Moreover, we tested the neuroprotective effect of Retigabine (RTG), a specific Kv7 channel opener, on disease outcome. METHODS To resemble the human disease, we focally administered pro-inflammatory cytokines, TNF-α and IFN-γ, in the primary auditory cortex (A1) of MOG35-55 immunized mice. Thereafter, we investigated the impact of the induced inflammatory milieu on afferent thalamocortical (TC) neurons, by performing ex vivo recordings. Moreover, we explored the effect of Kv7 channel modulation with RTG on auditory information processing, using in vivo electrophysiological approaches. RESULTS Our results revealed that a cortical inflammatory lesion profoundly affected the excitability and firing pattern of neighboring TC neurons. Noteworthy, RTG restored control-like values and TC tonotopic mapping. CONCLUSION Our results suggest that RTG treatment might robustly mitigate inflammation-induced altered excitability and preserve ascending information processing.
Collapse
Affiliation(s)
- Luca Fazio
- Department of Neurology, University of Düsseldorf, Düsseldorf, Germany.
| | - Venu Narayanan Naik
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.
| | | | | | - Nicole Rychlik
- Institute of Physiology I, University of Münster, Münster, Germany.
| | - Julia Ladewig
- Department of Translational Brain Research, Central Institute of Mental Health (ZI), University of Heidelberg/Medical Faculty Mannheim, Germany; HITBR Hector Institute for Translational Brain Research gGmbH, Mannheim, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Michael Strüber
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe University, Frankfurt, Germany.
| | - Manuela Cerina
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Sven G Meuth
- Department of Neurology, University of Düsseldorf, Düsseldorf, Germany.
| | - Thomas Budde
- Institute of Physiology I, University of Münster, Münster, Germany.
| |
Collapse
|
7
|
Chinigò G, Ruffinatti FA, Munaron L. The potential of TRP channels as new prognostic and therapeutic targets against prostate cancer progression. Biochim Biophys Acta Rev Cancer 2024; 1879:189226. [PMID: 39586480 DOI: 10.1016/j.bbcan.2024.189226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/28/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Prostate cancer (PCa) is the second deadliest cancer among men worldwide. Particularly critical is its development towards metastatic androgen-independent forms for which the current therapies are ineffective. Indeed, the 5-year relative survival for PCa drops dramatically to 34 % in the presence of metastases. The superfamily of Transient Receptor Potential (TRP) channels could answer the urgent request to identify new prognostic and therapeutic tools against metastatic PCa. Indeed, this class of ion channels revealed an appealing de-regulation during PCa development and its progression towards aggressive forms. Altered expression and/or functionality of several TRPs have been associated with the PCa metastatic cascade by significantly impacting tumor growth, invasiveness, and angiogenesis. In this review, we will dissect the contribution of TRP channels in such hallmarks of PCa and then discuss their applicability as new prognostic and therapeutic agents in the fight against metastatic PCa. In particular, the great potential of TRPM8, TRPV6, and TRPA1 in opening the way to new treatment perspectives will be highlighted.
Collapse
Affiliation(s)
- Giorgia Chinigò
- University of Turin, Department of Life Sciences and Systems Biology, via Accademia Albertina 13, 10123 Turin, Italy.
| | | | - Luca Munaron
- University of Turin, Department of Life Sciences and Systems Biology, via Accademia Albertina 13, 10123 Turin, Italy.
| |
Collapse
|
8
|
Li Y, Fu J, Wang H. Advancements in Targeting Ion Channels for the Treatment of Neurodegenerative Diseases. Pharmaceuticals (Basel) 2024; 17:1462. [PMID: 39598374 PMCID: PMC11597607 DOI: 10.3390/ph17111462] [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: 09/30/2024] [Revised: 10/20/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Ion channels are integral membrane proteins embedded in biological membranes, and they comprise specific proteins that control the flow of ion transporters in and out of cells, playing crucial roles in the biological functions of different cells. They maintain the homeostasis of water and ion metabolism by facilitating ion transport and participate in the physiological processes of neurons and glial cells by regulating signaling pathways. Neurodegenerative diseases are a group of disorders characterized by the progressive loss of neurons in the central nervous system (CNS) or peripheral nervous system (PNS). Despite significant progress in understanding the pathophysiological processes of various neurological diseases in recent years, effective treatments for mitigating the damage caused by these diseases remain inadequate. Increasing evidence suggests that ion channels are closely associated with neuroinflammation; oxidative stress; and the characteristic proteins in neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Therefore, studying the pathogenic mechanisms closely related to ion channels in neurodegenerative diseases can help identify more effective therapeutic targets for treating neurodegenerative diseases. Here, we discuss the progress of research on ion channels in different neurodegenerative diseases and emphasize the feasibility and potential of treating such diseases from the perspective of ion channels.
Collapse
Affiliation(s)
- Yuxuan Li
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.L.); (J.F.)
| | - Jingxuan Fu
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.L.); (J.F.)
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300401, China
| | - Hui Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.L.); (J.F.)
| |
Collapse
|
9
|
Yu F, Hubrack S, Raynaud CM, Elmi A, Mackeh R, Agrebi N, Thareja G, Belkadi A, Al Saloos H, Ahmed AA, Purayil SC, Mohamoud YA, Suhre K, Abi Khalil C, Schmidt F, Lo B, Hassan A, Machaca K. Loss of the TRPM4 channel in humans causes immune dysregulation with defective monocyte migration. J Allergy Clin Immunol 2024; 154:792-806. [PMID: 38750824 DOI: 10.1016/j.jaci.2024.02.026] [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: 06/07/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND TRPM4 is a broadly expressed, calcium-activated, monovalent cation channel that regulates immune cell function in mice and cell lines. Clinically, however, partial loss- or gain-of-function mutations in TRPM4 lead to arrhythmia and heart disease, with no documentation of immunologic disorders. OBJECTIVE To characterize functional cellular mechanisms underlying the immune dysregulation phenotype in a proband with a mutated TRPM4 gene. METHODS We employed a combination of biochemical, cell biological, imaging, omics analyses, flow cytometry, and gene editing approaches. RESULTS We report the first human cases to our knowledge with complete loss of the TRPM4 channel, leading to immune dysregulation with frequent bacterial and fungal infections. Single-cell and bulk RNA sequencing point to altered expression of genes affecting cell migration, specifically in monocytes. Inhibition of TRPM4 in T cells and the THP-1 monocyte cell line reduces migration. More importantly, primary T cells and monocytes from TRPM4 patients migrate poorly. Finally, CRISPR knockout of TRPM4 in THP-1 cells greatly reduces their migration potential. CONCLUSION Our results demonstrate that TRPM4 plays a critical role in regulating immune cell migration, leading to increased susceptibility to infections.
Collapse
Affiliation(s)
- Fang Yu
- Calcium Signaling Group, Research Department, Weill Cornell Medicine-Qatar, Doha, Qatar; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY
| | | | | | - Asha Elmi
- Research Department, Sidra Medicine, Doha, Qatar
| | - Rafah Mackeh
- Research Department, Sidra Medicine, Doha, Qatar
| | | | - Gaurav Thareja
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY; Research Department, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Abdelaziz Belkadi
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY; Research Department, Weill Cornell Medicine-Qatar, Doha, Qatar
| | | | | | - Saleema C Purayil
- Allergy & Immunology Division, Department of Medicine, Hamad Medical Corporation, Doha, Qatar
| | | | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY; Research Department, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Charbel Abi Khalil
- Research Department, Weill Cornell Medicine-Qatar, Doha, Qatar; Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Frank Schmidt
- Research Department, Sidra Medicine, Doha, Qatar; Department of Biochemistry, Weill Cornell Medicine, New York, NY
| | - Bernice Lo
- Research Department, Sidra Medicine, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| | - Amel Hassan
- Pediatric Allergy and Immunology Department, Sidra Medicine, Doha, Qatar.
| | - Khaled Machaca
- Calcium Signaling Group, Research Department, Weill Cornell Medicine-Qatar, Doha, Qatar; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY.
| |
Collapse
|
10
|
Woo MS, Mayer C, Binkle-Ladisch L, Sonner JK, Rosenkranz SC, Shaposhnykov A, Rothammer N, Tsvilovskyy V, Lorenz SM, Raich L, Bal LC, Vieira V, Wagner I, Bauer S, Glatzel M, Conrad M, Merkler D, Freichel M, Friese MA. STING orchestrates the neuronal inflammatory stress response in multiple sclerosis. Cell 2024; 187:4043-4060.e30. [PMID: 38878778 DOI: 10.1016/j.cell.2024.05.031] [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: 01/16/2024] [Revised: 04/08/2024] [Accepted: 05/16/2024] [Indexed: 07/28/2024]
Abstract
Inflammation-induced neurodegeneration is a defining feature of multiple sclerosis (MS), yet the underlying mechanisms remain unclear. By dissecting the neuronal inflammatory stress response, we discovered that neurons in MS and its mouse model induce the stimulator of interferon genes (STING). However, activation of neuronal STING requires its detachment from the stromal interaction molecule 1 (STIM1), a process triggered by glutamate excitotoxicity. This detachment initiates non-canonical STING signaling, which leads to autophagic degradation of glutathione peroxidase 4 (GPX4), essential for neuronal redox homeostasis and thereby inducing ferroptosis. Both genetic and pharmacological interventions that target STING in neurons protect against inflammation-induced neurodegeneration. Our findings position STING as a central regulator of the detrimental neuronal inflammatory stress response, integrating inflammation with glutamate signaling to cause neuronal cell death, and present it as a tractable target for treating neurodegeneration in MS.
Collapse
Affiliation(s)
- Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sina C Rosenkranz
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Artem Shaposhnykov
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Svenja M Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lukas Raich
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas C Bal
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Simone Bauer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
11
|
Lee HG, Quintana FJ. STING: Stay near to STIM(1) neuroprotection. Mol Cell 2024; 84:2596-2597. [PMID: 39059368 DOI: 10.1016/j.molcel.2024.06.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024]
Abstract
In a recent publication in Cell, Woo et al.1 report that stimulator of interferon genes (STING) links inflammation with glutamate-driven excitotoxicity to induce ferroptosis, identifying a mechanism of inflammation-induced neurodegeneration and also a novel candidate therapeutic target for multiple sclerosis.
Collapse
Affiliation(s)
- Hong-Gyun Lee
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Gene Lay Institute of Immunology and Inflammation, Boston, MA, USA.
| |
Collapse
|
12
|
Woo MS, Engler JB, Friese MA. The neuropathobiology of multiple sclerosis. Nat Rev Neurosci 2024; 25:493-513. [PMID: 38789516 DOI: 10.1038/s41583-024-00823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
Chronic low-grade inflammation and neuronal deregulation are two components of a smoldering disease activity that drives the progression of disability in people with multiple sclerosis (MS). Although several therapies exist to dampen the acute inflammation that drives MS relapses, therapeutic options to halt chronic disability progression are a major unmet clinical need. The development of such therapies is hindered by our limited understanding of the neuron-intrinsic determinants of resilience or vulnerability to inflammation. In this Review, we provide a neuron-centric overview of recent advances in deciphering neuronal response patterns that drive the pathology of MS. We describe the inflammatory CNS environment that initiates neurotoxicity by imposing ion imbalance, excitotoxicity and oxidative stress, and by direct neuro-immune interactions, which collectively lead to mitochondrial dysfunction and epigenetic dysregulation. The neuronal demise is further amplified by breakdown of neuronal transport, accumulation of cytosolic proteins and activation of cell death pathways. Continuous neuronal damage perpetuates CNS inflammation by activating surrounding glia cells and by directly exerting toxicity on neighbouring neurons. Further, we explore strategies to overcome neuronal deregulation in MS and compile a selection of neuronal actuators shown to impact neurodegeneration in preclinical studies. We conclude by discussing the therapeutic potential of targeting such neuronal actuators in MS, including some that have already been tested in interventional clinical trials.
Collapse
Affiliation(s)
- Marcel S Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
13
|
Yang B, Ma D, Zhu X, Wu Z, An Q, Zhao J, Gao X, Zhang L. Roles of TRP and PIEZO receptors in autoimmune diseases. Expert Rev Mol Med 2024; 26:e10. [PMID: 38659380 PMCID: PMC11140548 DOI: 10.1017/erm.2023.23] [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: 01/31/2023] [Revised: 04/15/2023] [Accepted: 08/21/2023] [Indexed: 04/26/2024]
Abstract
Autoimmune diseases are pathological autoimmune reactions in the body caused by various factors, which can lead to tissue damage and organ dysfunction. They can be divided into organ-specific and systemic autoimmune diseases. These diseases usually involve various body systems, including the blood, muscles, bones, joints and soft tissues. The transient receptor potential (TRP) and PIEZO receptors, which resulted in David Julius and Ardem Patapoutian winning the Nobel Prize in Physiology or Medicine in 2021, attracted people's attention. Most current studies on TRP and PIEZO receptors in autoimmune diseases have been carried out on animal model, only few clinical studies have been conducted. Therefore, this study aimed to review existing studies on TRP and PIEZO to understand the roles of these receptors in autoimmune diseases, which may help elucidate novel treatment strategies.
Collapse
Affiliation(s)
- Baoqi Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xueqing Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Zewen Wu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Qi An
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Jingwen Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xinnan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| |
Collapse
|
14
|
Pironet A, Vandewiele F, Vennekens R. Exploring the role of TRPM4 in calcium-dependent triggered activity and cardiac arrhythmias. J Physiol 2024; 602:1605-1621. [PMID: 37128952 DOI: 10.1113/jp283831] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023] Open
Abstract
Cardiac arrhythmias pose a major threat to a patient's health, yet prove to be often difficult to predict, prevent and treat. A key mechanism in the occurrence of arrhythmias is disturbed Ca2+ homeostasis in cardiac muscle cells. As a Ca2+-activated non-selective cation channel, TRPM4 has been linked to Ca2+-induced arrhythmias, potentially contributing to translating an increase in intracellular Ca2+ concentration into membrane depolarisation and an increase in cellular excitability. Indeed, evidence from genetically modified mice, analysis of mutations in human patients and the identification of a TRPM4 blocking compound that can be applied in vivo further underscore this hypothesis. Here, we provide an overview of these data in the context of our current understanding of Ca2+-dependent arrhythmias.
Collapse
Affiliation(s)
- Andy Pironet
- Laboratory of Ion Channel Research, VIB Centre for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Frone Vandewiele
- Laboratory of Ion Channel Research, VIB Centre for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, VIB Centre for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Rezzani R, Favero G, Gianò M, Pinto D, Labanca M, van Noorden CJ, Rinaldi F. Transient Receptor Potential Channels in the Healthy and Diseased Blood-Brain Barrier. J Histochem Cytochem 2024; 72:199-231. [PMID: 38590114 PMCID: PMC11020746 DOI: 10.1369/00221554241246032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
The large family of transient receptor potential (TRP) channels are integral membrane proteins that function as environmental sensors and act as ion channels after activation by mechanical (touch), physical (heat, pain), and chemical stimuli (pungent compounds such as capsaicin). Most TRP channels are localized in the plasma membrane of cells but some of them are localized in membranes of organelles and function as intracellular Ca2+-ion channels. TRP channels are involved in neurological disorders but their precise role(s) and relevance in these disorders are not clear. Endothelial cells of the blood-brain barrier (BBB) express TRP channels such as TRP vanilloid 1-4 and are involved in thermal detection by regulating BBB permeability. In neurological disorders, TRP channels in the BBB are responsible for edema formation in the brain. Therefore, drug design to modulate locally activity of TRP channels in the BBB is a hot topic. Today, the application of TRP channel antagonists against neurological disorders is still limited.
Collapse
Affiliation(s)
- Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Gaia Favero
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
| | - Marzia Gianò
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Pinto
- Human Microbiome Advanced Project Institute, Milan, Italy
| | - Mauro Labanca
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Fabio Rinaldi
- Human Microbiome Advanced Project Institute, Milan, Italy
| |
Collapse
|
16
|
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
|
17
|
Chubanov V, Köttgen M, Touyz RM, Gudermann T. TRPM channels in health and disease. Nat Rev Nephrol 2024; 20:175-187. [PMID: 37853091 DOI: 10.1038/s41581-023-00777-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
Different cell channels and transporters tightly regulate cytoplasmic levels and the intraorganelle distribution of cations. Perturbations in these processes lead to human diseases that are frequently associated with kidney impairment. The family of melastatin-related transient receptor potential (TRPM) channels, which has eight members in mammals (TRPM1-TRPM8), includes ion channels that are highly permeable to divalent cations, such as Ca2+, Mg2+ and Zn2+ (TRPM1, TRPM3, TRPM6 and TRPM7), non-selective cation channels (TRPM2 and TRPM8) and monovalent cation-selective channels (TRPM4 and TRPM5). Three family members contain an enzymatic protein moiety: TRPM6 and TRPM7 are fused to α-kinase domains, whereas TRPM2 is linked to an ADP-ribose-binding NUDT9 homology domain. TRPM channels also function as crucial cellular sensors involved in many physiological processes, including mineral homeostasis, blood pressure, cardiac rhythm and immunity, as well as photoreception, taste reception and thermoreception. TRPM channels are abundantly expressed in the kidney. Mutations in TRPM genes cause several inherited human diseases, and preclinical studies in animal models of human disease have highlighted TRPM channels as promising new therapeutic targets. Here, we provide an overview of this rapidly evolving research area and delineate the emerging role of TRPM channels in kidney pathophysiology.
Collapse
Affiliation(s)
- Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.
| | - Michael Köttgen
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, Freiburg, Germany
| | - Rhian M Touyz
- Research Institute of McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.
| |
Collapse
|
18
|
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
|
19
|
Niu L, Liu H, Li X, Wang L, Hua H, Cao Q, Xiang Q, Cai T, Zhu D. Design, synthesis, and biological evaluation of 2-(naphthalen-1-yloxy)-N-phenylacetamide derivatives as TRPM4 inhibitors for the treatment of prostate cancer. Bioorg Med Chem 2024; 98:117584. [PMID: 38168629 DOI: 10.1016/j.bmc.2023.117584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024]
Abstract
Transient receptor potential melastatin 4 (TRPM4) is considered to be a potential target for cancer and other human diseases. Herein, a series of 2-(naphthalen-1-yloxy)-N-phenylacetamide derivatives were designed and synthesized as new TRPM4 inhibitors, aiming to improve cellular potency. One of the most promising compounds, 7d (ZX08903), displayed promising antiproliferative activity against prostate cancer cell lines. 7d also suppressed colony formation and the expression of androgen receptor (AR) protein in prostate cancer cells. Furthermore, 7d can concentration-dependently induce cell apoptosis in prostate cancer cells. Collectively, these findings indicated that compound 7d may serve as a promising lead compound for further anticancer drug development.
Collapse
Affiliation(s)
- Le Niu
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo No.2 Hospital, Ningbo, 315010, China; Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, and Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Huina Liu
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Xiaomei Li
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Lin Wang
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, and Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Hui Hua
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Qiaofeng Cao
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, and Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Qiuping Xiang
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Ting Cai
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo No.2 Hospital, Ningbo, 315010, China.
| | - Dongsheng Zhu
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, and Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
20
|
Fan H, Yang Y, Bai Q, Wang D, Shi X, Zhang L, Yang Y. Neuroprotective Effects of Sinomenine on Experimental Autoimmune Encephalomyelitis via Anti-Inflammatory and Nrf2-Dependent Anti-Oxidative Stress Activity. Neuromolecular Med 2023; 25:545-562. [PMID: 37735290 DOI: 10.1007/s12017-023-08756-z] [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: 04/11/2023] [Accepted: 08/24/2023] [Indexed: 09/23/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system (CNS). Sinomenine (SIN), a bioactive alkaloid extracted from the Chinese medicinal plant Sinomenium acutum, has powerful anti-inflammatory and immunosuppressive therapeutic benefits. In our previous research, we found that SIN increased resistance to oxidative stress via the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway in PC12 neuronal cells. However, whether SIN can improve the symptoms and pathological features of experimental autoimmune encephalomyelitis (EAE), a murine model of MS, via the Nrf2 signaling pathway remains unclear. EAE was immunized followed by SIN treatment. Then we evaluated the effects of SIN in EAE. Subsequently, primary microglia were cultured to explore the effect of SIN on microglia activation. Further, the levels of Nrf2 and its downstream molecules were detected to assess the molecular mechanisms of SIN. We demonstrated that SIN effectively ameliorated the severity of EAE, accompanied by a reduction in the demyelination, axonal damage and inhibition of inflammatory cell infiltration. Mechanistically, SIN decreased the inflammatory cytokines expression, and suppressed microglia and astrocytes activation in EAE mice. Furthermore, SIN suppressed lipopolysaccharide (LPS)-induced microglial activation and the production of pro-inflammatory factors in vitro. Moreover, SIN inhibited oxidative stress via the activation of the Nrf2 signaling pathway. Our work proves that SIN exerts its neuroprotective effects by the Nrf2-dependent anti-oxidative stress and diminishing neuroinflammation, suggesting that the "antioxiflammation" effect of SIN is expected to be an ideal treatment strategy for MS/EAE.
Collapse
Affiliation(s)
- Hua Fan
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China.
| | - Yang Yang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Qianqian Bai
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Dongmei Wang
- School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, 471003, China
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Lele Zhang
- Department of traditional Chinese medicine, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Yanhui Yang
- Department of Trauma center, The First Affiliated Hospital, College of Clinical Medicine , Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
21
|
Ren K, Pei J, Guo Y, Jiao Y, Xing H, Xie Y, Yang Y, Feng Q, Yang J. Regulated necrosis pathways: a potential target for ischemic stroke. BURNS & TRAUMA 2023; 11:tkad016. [PMID: 38026442 PMCID: PMC10656754 DOI: 10.1093/burnst/tkad016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/24/2022] [Indexed: 12/01/2023]
Abstract
Globally, ischemic stroke causes millions of deaths per year. The outcomes of ischemic stroke are largely determined by the amount of ischemia-related and reperfusion-related neuronal death in the infarct region. In the infarct region, cell injuries follow either the regulated pathway involving precise signaling cascades, such as apoptosis and autophagy, or the nonregulated pathway, which is uncontrolled by any molecularly defined effector mechanisms such as necrosis. However, numerous studies have recently found that a certain type of necrosis can be regulated and potentially modified by drugs and is nonapoptotic; this type of necrosis is referred to as regulated necrosis. Depending on the signaling pathway, various elements of regulated necrosis contribute to the development of ischemic stroke, such as necroptosis, pyroptosis, ferroptosis, pathanatos, mitochondrial permeability transition pore-mediated necrosis and oncosis. In this review, we aim to summarize the underlying molecular mechanisms of regulated necrosis in ischemic stroke and explore the crosstalk and interplay among the diverse types of regulated necrosis. We believe that targeting these regulated necrosis pathways both pharmacologically and genetically in ischemia-induced neuronal death and protection could be an efficient strategy to increase neuronal survival and regeneration in ischemic stroke.
Collapse
Affiliation(s)
- Kaidi Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Jinyan Pei
- Quality Management Department, Henan No. 3 Provincial People’s Hospital, Henan No. 3 Provincial People’s Hospital, Zhengzhou 450052, China
| | - Yuanyuan Guo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yuxue Jiao
- Quality Management Department, Henan No. 3 Provincial People’s Hospital, Henan No. 3 Provincial People’s Hospital, Zhengzhou 450052, China
| | - Han Xing
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yang Yang
- Research Center for Clinical System Biology, Translational Medicine Center, No. 1 Jianshe Dong Road, ErQi District, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qi Feng
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
| | - Jing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
22
|
Vydra Bousova K, Zouharova M, Jiraskova K, Vetyskova V. Interaction of Calmodulin with TRPM: An Initiator of Channel Modulation. Int J Mol Sci 2023; 24:15162. [PMID: 37894842 PMCID: PMC10607381 DOI: 10.3390/ijms242015162] [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: 08/19/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Transient receptor potential melastatin (TRPM) channels, a subfamily of the TRP superfamily, constitute a diverse group of ion channels involved in mediating crucial cellular processes like calcium homeostasis. These channels exhibit complex regulation, and one of the key regulatory mechanisms involves their interaction with calmodulin (CaM), a cytosol ubiquitous calcium-binding protein. The association between TRPM channels and CaM relies on the presence of specific CaM-binding domains in the channel structure. Upon CaM binding, the channel undergoes direct and/or allosteric structural changes and triggers down- or up-stream signaling pathways. According to current knowledge, ion channel members TRPM2, TRPM3, TRPM4, and TRPM6 are directly modulated by CaM, resulting in their activation or inhibition. This review specifically focuses on the interplay between TRPM channels and CaM and summarizes the current known effects of CaM interactions and modulations on TRPM channels in cellular physiology.
Collapse
|
23
|
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
|
24
|
Yan J, Bading H. The Disruption of NMDAR/TRPM4 Death Signaling with TwinF Interface Inhibitors: A New Pharmacological Principle for Neuroprotection. Pharmaceuticals (Basel) 2023; 16:1085. [PMID: 37631001 PMCID: PMC10458786 DOI: 10.3390/ph16081085] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
With the discovery that the acquisition of toxic features by extrasynaptic NMDA receptors (NMDARs) involves their physical interaction with the non-selective cation channel, TRPM4, it has become possible to develop a new pharmacological principle for neuroprotection, namely the disruption of the NMDAR/TRPM4 death signaling complex. This can be accomplished through the expression of the TwinF domain, a 57-amino-acid-long stretch of TRPM4 that mediates its interaction with NMDARs, but also using small molecule TwinF interface (TI) inhibitors, also known as NMDAR/TRPM4 interaction interface inhibitors. Both TwinF and small molecule TI inhibitors detoxify extrasynaptic NMDARs without interfering with synaptic NMDARs, which serve important physiological functions in the brain. As the toxic signaling of extrasynaptic NMDARs contributes to a wide range of neurodegenerative conditions, TI inhibitors may offer therapeutic options for currently untreatable human neurodegenerative diseases including Amyotrophic Lateral Sclerosis, Alzheimer's disease, and Huntington's disease.
Collapse
Affiliation(s)
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
25
|
Zhao Q, Li H, Li H, Zhang J. Research progress on pleiotropic neuroprotective drugs for traumatic brain injury. Front Pharmacol 2023; 14:1185533. [PMID: 37475717 PMCID: PMC10354289 DOI: 10.3389/fphar.2023.1185533] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/26/2023] [Indexed: 07/22/2023] Open
Abstract
Traumatic brain injury (TBI) has become one of the most important causes of death and disability worldwide. A series of neuroinflammatory responses induced after TBI are key factors for persistent neuronal damage, but at the same time, such inflammatory responses can also promote debris removal and tissue repair after TBI. The concept of pleiotropic neuroprotection delves beyond the single-target treatment approach, considering the multifaceted impacts following TBI. This notion embarks deeper into the research-oriented treatment paradigm, focusing on multi-target interventions that inhibit post-TBI neuroinflammation with enhanced therapeutic efficacy. With an enriched comprehension of TBI's physiological mechanisms, this review dissects the advancements in developing pleiotropic neuroprotective pharmaceuticals to mitigate TBI. The aim is to provide insights that may contribute to the early clinical management of the condition.
Collapse
Affiliation(s)
- Qinghui Zhao
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Huige Li
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Hongru Li
- Zhumadian Central Hospital, Zhumadian, China
| | - Jianhua Zhang
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| |
Collapse
|
26
|
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
|
27
|
Mundrucz L, Kecskés A, Henn-Mike N, Kóbor P, Buzás P, Vennekens R, Kecskés M. TRPM4 regulates hilar mossy cell loss in temporal lobe epilepsy. BMC Biol 2023; 21:96. [PMID: 37101159 PMCID: PMC10134545 DOI: 10.1186/s12915-023-01604-3] [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: 11/22/2022] [Accepted: 04/18/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Mossy cells comprise a large fraction of excitatory neurons in the hippocampal dentate gyrus, and their loss is one of the major hallmarks of temporal lobe epilepsy (TLE). The vulnerability of mossy cells in TLE is well known in animal models as well as in patients; however, the mechanisms leading to cellular death is unclear. RESULTS Transient receptor potential melastatin 4 (TRPM4) is a Ca2+-activated non-selective cation channel regulating diverse physiological functions of excitable cells. Here, we identified that TRPM4 is present in hilar mossy cells and regulates their intrinsic electrophysiological properties including spontaneous activity and action potential dynamics. Furthermore, we showed that TRPM4 contributes to mossy cells death following status epilepticus and therefore modulates seizure susceptibility and epilepsy-related memory deficits. CONCLUSIONS Our results provide evidence for the role of TRPM4 in MC excitability both in physiological and pathological conditions.
Collapse
Affiliation(s)
- Laura Mundrucz
- Institute of Physiology, Medical School, University of Pécs, Pécs, 7624, Hungary
| | - Angéla Kecskés
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, 7624, Hungary
- Szentagothai Research Centre, Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary
| | - Nóra Henn-Mike
- Institute of Physiology, Medical School, University of Pécs, Pécs, 7624, Hungary
- Szentagothai Research Centre, Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary
| | - Péter Kóbor
- Institute of Physiology, Medical School, University of Pécs, Pécs, 7624, Hungary
| | - Péter Buzás
- Institute of Physiology, Medical School, University of Pécs, Pécs, 7624, Hungary
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, Biomedical Sciences Group, Department of Cellular and Molecular Medicine, VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, Louvain, 3000, Belgium
| | - Miklós Kecskés
- Institute of Physiology, Medical School, University of Pécs, Pécs, 7624, Hungary.
| |
Collapse
|
28
|
Li J, Lu L, Binder K, Xiong J, Ye L, Cheng YH, Majri-Morrison S, Lu W, Lee JW, Zhang Z, Wu YZ, Zheng L, Lenardo MJ. Mechanisms of antigen-induced reversal of CNS inflammation in experimental demyelinating disease. SCIENCE ADVANCES 2023; 9:eabo2810. [PMID: 36857453 PMCID: PMC9977187 DOI: 10.1126/sciadv.abo2810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 12/27/2022] [Indexed: 06/18/2023]
Abstract
Autoimmune central nervous system (CNS) demyelinating diseases are a major public health burden and poorly controlled by current immunosuppressants. More precise immunotherapies with higher efficacy and fewer side effects are sought. We investigated the effectiveness and mechanism of an injectable myelin-based antigenic polyprotein MMPt (myelin oligodendrocyte glycoprotein, myelin basic protein and proteolipid protein, truncated). We find that it suppresses mouse experimental autoimmune encephalomyelitis without major side effects. MMPt induces rapid apoptosis of the encephalitogenic T cells and suppresses inflammation in the affected CNS. Intravital microscopy shows that MMPt is taken up by perivascular F4/80+ cells but not conventional antigen-presenting dendritic cells, B cells, or microglia. MMPt-stimulated F4/80+ cells induce reactive T cell immobilization and apoptosis in situ, resulting in reduced infiltration of inflammatory cells and chemokine production. Our study reveals alternative mechanisms that explain how cognate antigen suppresses CNS inflammation and may be applicable for effectively and safely treating demyelinating diseases.
Collapse
Affiliation(s)
- Jian Li
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lisen Lu
- MoE Key Laboratory for Biomedical Photonics, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Kyle Binder
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jian Xiong
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan H. Cheng
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sonia Majri-Morrison
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wei Lu
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jae W. Lee
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zhihong Zhang
- MoE Key Laboratory for Biomedical Photonics, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yu-zhang Wu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lixin Zheng
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael J. Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
29
|
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
|
30
|
Yamada D, Vu S, Wu X, Shi Z, Morris D, Bloomstein JD, Huynh M, Zheng J, Hwang ST. Gain-of-function of TRPM4 predisposes mice to psoriasiform dermatitis. Front Immunol 2022; 13:1025499. [PMID: 36341417 PMCID: PMC9632438 DOI: 10.3389/fimmu.2022.1025499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022] Open
Abstract
Transient receptor potential melastatin 4 (TRPM4) is a Ca2+-activated, monovalent cation channel that is expressed in a wide range of cells. We previously reported two gain-of-function (GoF) mutations of TRPM4 as the cause of progressive symmetric erythrokeratodermia (PSEK), which shares similar clinical and histopathological features with psoriasis. Using CRISPR/Cas9 technology, we generated TRPM4I1029M mice that have the equivalent mutation to one of the two genetic mutations found in human PSEK (equivalent to human TRPM4I1033M). Using this mutant mice, we examined the effects of TRPM4 GoF at the cellular and phenotypic levels to elucidate the pathological mechanisms underlying PSEK. In the absence of experimental stimulation, TRPM4I1029M mice did not show a phenotype. When treated with imiquimod (IMQ), however, TRPM4I1029M mice were predisposed to more severe psoriasiform dermatitis (PsD) than wild-type (WT), which was characterized by greater accumulation of CCR6-expressing γδ T cells and higher mRNA levels of Il17a. In TRPM4I1029M mice, dendritic cells showed enhanced migration and keratinocytes exhibited increased proliferation. Moreover, a TRPM4 inhibitor, glibenclamide, ameliorated PsD in WT and TRPM4I1029M mice. Our results indicate elevated TRPM4 activities boosted susceptibility to cutaneous stimuli, likely through elevation of membrane potential and alteration of downstream cellular signaling, resulting in enhanced inflammation. Our results further suggest a possible therapeutic application of TRPM4 inhibitors in psoriasis.
Collapse
Affiliation(s)
- Daisuke Yamada
- Department of Dermatology, University of California, Davis, Sacramento, CA, United States
| | - Simon Vu
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Xuesong Wu
- Department of Dermatology, University of California, Davis, Sacramento, CA, United States
| | - Zhenrui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Desiree Morris
- Kirk Kerkorian School of Medicine at University of Nevada, Las Vegas, Las, Vegas, NV, United States
| | - Joshua D Bloomstein
- Department of Dermatology, University of California, Davis, Sacramento, CA, United States
| | - Mindy Huynh
- Department of Dermatology, University of California, Davis, Sacramento, CA, United States
| | - Jie Zheng
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Samuel T Hwang
- Department of Dermatology, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|
31
|
Chen J, Chang Y, Zhu J, Peng Y, Li Z, Zhang K, Zhang Y, Lin C, Lin Z, Pan S, Huang K. Flufenamic acid improves survival and neurologic outcome after successful cardiopulmonary resuscitation in mice. J Neuroinflammation 2022; 19:214. [PMID: 36050694 PMCID: PMC9438280 DOI: 10.1186/s12974-022-02571-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/19/2022] [Indexed: 11/10/2022] Open
Abstract
Background Brain injury is the main cause of high mortality and disability after successful cardiopulmonary resuscitation (CPR) from sudden cardiac arrest (CA). The transient receptor potential M4 (TRPM4) channel is a novel target for ameliorating blood–brain barrier (BBB) disruption and neuroinflammation. Herein, we tested whether flufenamic acid (FFA), which is reported to block TRPM4 with high potency, could confer neuroprotection against brain injury secondary to CA/CPR and whether its action was exerted by blocking the TRPM4 channel. Methods Wild-type (WT) and Trpm4 knockout (Trpm4−/−) mice subjected to 10-min CA/CPR were randomized to receive FFA or vehicle once daily. Post-CA/CPR brain injuries including neurologic deficits, survival rate, histological damage, edema formation, BBB destabilization and neuroinflammation were assessed. Results In WT mice subjected to CA/CPR, FFA was effective in improving survival and neurologic outcome, reducing neuropathological injuries, attenuating brain edema, lessening the leakage of IgG and Evans blue dye, restoring tight junction protein expression and promoting microglia/macrophages from the pro-inflammatory subtype toward the anti-inflammatory subtype. In comparison to WT mice, Trpm4−/− mice exhibited less neurologic deficiency, milder histological impairment, more BBB integrity and more anti-inflammatory microglia/macrophage polarization. As expected, FFA did not provide a benefit of superposition compared with vehicle in the Trpm4−/− mice after CA/CPR. Conclusions FFA mitigates BBB breach and modifies the functional status of microglia/macrophages, thereby improving survival and neurologic deficits following CA/CPR. The neuroprotective effects occur at least partially by interfering with the TRPM4 channel in the neurovascular unit. These results indicate the significant clinical potential of FFA to improve the prognosis for CA victims who are successfully resuscitated. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02571-2.
Collapse
Affiliation(s)
- Jiancong Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Yuan Chang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Juan Zhu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Yuqin Peng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Zheqi Li
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Kunxue Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Yuzhen Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Chuman Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Zhenzhou Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China.
| | - Kaibin Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China.
| |
Collapse
|
32
|
Rothammer N, Woo MS, Bauer S, Binkle-Ladisch L, Di Liberto G, Egervari K, Wagner I, Haferkamp U, Pless O, Merkler D, Engler JB, Friese MA. G9a dictates neuronal vulnerability to inflammatory stress via transcriptional control of ferroptosis. SCIENCE ADVANCES 2022; 8:eabm5500. [PMID: 35930635 PMCID: PMC9355351 DOI: 10.1126/sciadv.abm5500] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 06/23/2022] [Indexed: 06/15/2023]
Abstract
Neuroinflammation leads to neuronal stress responses that contribute to neuronal dysfunction and loss. However, treatments that stabilize neurons and prevent their destruction are still lacking. Here, we identify the histone methyltransferase G9a as a druggable epigenetic regulator of neuronal vulnerability to inflammation. In murine experimental autoimmune encephalomyelitis (EAE) and human multiple sclerosis (MS), we found that the G9a-catalyzed repressive epigenetic mark H3K9me2 was robustly induced by neuroinflammation. G9a activity repressed anti-ferroptotic genes, diminished intracellular glutathione levels, and triggered the iron-dependent programmed cell death pathway ferroptosis. Conversely, pharmacological treatment of EAE mice with a G9a inhibitor restored anti-ferroptotic gene expression, reduced inflammation-induced neuronal loss, and improved clinical outcome. Similarly, neuronal anti-ferroptotic gene expression was reduced in MS brain tissue and was boosted by G9a inhibition in human neuronal cultures. This study identifies G9a as a critical transcriptional enhancer of neuronal ferroptosis and potential therapeutic target to counteract inflammation-induced neurodegeneration.
Collapse
Affiliation(s)
- Nicola Rothammer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marcel S. Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Simone Bauer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Giovanni Di Liberto
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Kristof Egervari
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Manuel A. Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
33
|
Herwerth M, Kenet S, Schifferer M, Winkler A, Weber M, Snaidero N, Wang M, Lohrberg M, Bennett JL, Stadelmann C, Hemmer B, Misgeld T. A new form of axonal pathology in a spinal model of neuromyelitis optica. Brain 2022; 145:1726-1742. [PMID: 35202467 PMCID: PMC9166560 DOI: 10.1093/brain/awac079] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 01/31/2022] [Accepted: 02/12/2022] [Indexed: 11/14/2022] Open
Abstract
Neuromyelitis optica is a chronic neuroinflammatory disease, which primarily targets astrocytes and often results in severe axon injury of unknown mechanism. Neuromyelitis optica patients harbour autoantibodies against the astrocytic water channel protein, aquaporin-4 (AQP4-IgG), which induce complement-mediated astrocyte lysis and subsequent axon damage. Using spinal in vivo imaging in a mouse model of such astrocytopathic lesions, we explored the mechanism underlying neuromyelitis optica-related axon injury. Many axons showed a swift and morphologically distinct 'pearls-on-string' transformation also readily detectable in human neuromyelitis optica lesions, which especially affected small calibre axons independently of myelination. Functional imaging revealed that calcium homeostasis was initially preserved in this 'acute axonal beading' state, ruling out disruption of the axonal membrane, which sets this form of axon injury apart from previously described forms of traumatic and inflammatory axon damage. Morphological, pharmacological and genetic analyses showed that AQP4-IgG-induced axon injury involved osmotic stress and ionic overload, but does not appear to use canonical pathways of Wallerian-like degeneration. Subcellular analysis demonstrated remodelling of the axonal cytoskeleton in beaded axons, especially local loss of microtubules. Treatment with the microtubule stabilizer epothilone, a putative therapy approach for traumatic and degenerative axonopathies, prevented axonal beading, while destabilizing microtubules sensitized axons for beading. Our results reveal a distinct form of immune-mediated axon pathology in neuromyelitis optica that mechanistically differs from known cascades of post-traumatic and inflammatory axon loss, and suggest a new strategy for neuroprotection in neuromyelitis optica and related diseases.
Collapse
Affiliation(s)
- Marina Herwerth
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Selin Kenet
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians University, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Anne Winkler
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Melanie Weber
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Nicolas Snaidero
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Mengzhe Wang
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Melanie Lohrberg
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Jeffrey L. Bennett
- Departments of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado School of Medicine, Aurora, USA
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
34
|
A Novel Role of the TRPM4 Ion Channel in Exocytosis. Cells 2022; 11:cells11111793. [PMID: 35681487 PMCID: PMC9180413 DOI: 10.3390/cells11111793] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 11/23/2022] Open
Abstract
Under physiological conditions, the widely expressed calcium-activated TRPM4 channel conducts sodium into cells. This sodium influx depolarizes the plasma membrane and reduces the driving force for calcium entry. The aberrant expression or function of TRPM4 has been reported in various diseases, including different types of cancer. TRPM4 is mainly localized in the plasma membrane, but it is also found in intracellular vesicles, which can undergo exocytosis. In this study, we show that calcium-induced exocytosis in the colorectal cancer cell line HCT116 is dependent on TRPM4. In addition, the findings from some studies of prostate cancer cell lines suggest a more general role of TRPM4 in calcium-induced exocytosis in cancer cells. Furthermore, calcium-induced exocytosis depends on TRPM4 ion conductivity. Additionally, an increase in intracellular calcium results in the delivery of TRPM4 to the plasma membrane. This process also depends on TRPM4 ion conductivity. TRPM4-dependent exocytosis and the delivery of TRPM4 to the plasma membrane are mediated by SNARE proteins. Finally, we provide evidence that calcium-induced exocytosis depends on TRPM4 ion conductivity, not within the plasma membrane, but rather in TRPM4-containing vesicles.
Collapse
|
35
|
A glibenclamide-sensitive TRPM4-mediated component of CA1 excitatory postsynaptic potentials appears in experimental autoimmune encephalomyelitis. Sci Rep 2022; 12:6000. [PMID: 35397639 PMCID: PMC8994783 DOI: 10.1038/s41598-022-09875-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 03/16/2022] [Indexed: 12/29/2022] Open
Abstract
The transient receptor potential melastatin 4 (TRPM4) channel contributes to disease severity in the murine experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis and to neuronal cell death in models of excitotoxicity and traumatic brain injury. As TRPM4 is activated by intracellular calcium and conducts monovalent cations, we hypothesized that TRPM4 may contribute to and boost excitatory synaptic transmission in CA1 pyramidal neurons of the hippocampus. Using single-spine calcium imaging and electrophysiology, we found no effect of the TRPM4 antagonists 9-phenanthrol and glibenclamide on synaptic transmission in hippocampal slices from healthy mice. In contrast, glibenclamide but not 9-phenanthrol reduced excitatory synaptic potentials in slices from EAE mice, an effect that was absent in slices from EAE mice lacking TRPM4. We conclude that TRPM4 plays little role in basal hippocampal synaptic transmission, but a glibenclamide-sensitive TRPM4-mediated contribution to excitatory postsynaptic responses is upregulated at the acute phase of EAE.
Collapse
|
36
|
Rosmus DD, Lange C, Ludwig F, Ajami B, Wieghofer P. The Role of Osteopontin in Microglia Biology: Current Concepts and Future Perspectives. Biomedicines 2022; 10:biomedicines10040840. [PMID: 35453590 PMCID: PMC9027630 DOI: 10.3390/biomedicines10040840] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/26/2022] [Accepted: 03/27/2022] [Indexed: 12/14/2022] Open
Abstract
The innate immune landscape of the central nervous system (CNS), including the brain and the retina, consists of different myeloid cell populations with distinct tasks to fulfill. Whereas the CNS borders harbor extraparenchymal CNS-associated macrophages whose main duty is to build up a defense against invading pathogens and other damaging factors from the periphery, the resident immune cells of the CNS parenchyma and the retina, microglia, are highly dynamic cells with a plethora of functions during homeostasis and disease. Therefore, microglia are constantly sensing their environment and closely interacting with surrounding cells, which is in part mediated by soluble factors. One of these factors is Osteopontin (OPN), a multifunctional protein that is produced by different cell types in the CNS, including microglia, and is upregulated in neurodegenerative and neuroinflammatory conditions. In this review, we discuss the current literature about the interaction between microglia and OPN in homeostasis and several disease entities, including multiple sclerosis (MS), Alzheimer’s and cerebrovascular diseases (AD, CVD), amyotrophic lateral sclerosis (ALS), age-related macular degeneration (AMD) and diabetic retinopathy (DR), in the context of the molecular pathways involved in OPN signaling shaping the function of microglia. As nearly all CNS diseases are characterized by pathological alterations in microglial cells, accompanied by the disturbance of the homeostatic microglia phenotype, the emergence of disease-associated microglia (DAM) states and their interplay with factors shaping the DAM-signature, such as OPN, is of great interest for therapeutical interventions in the future.
Collapse
Affiliation(s)
| | - Clemens Lange
- Eye Center, Freiburg Medical Center, University of Freiburg, 79106 Freiburg, Germany; (C.L.); (F.L.)
- Ophtha-Lab, Department of Ophthalmology, St. Franziskus Hospital, 48145 Muenster, Germany
| | - Franziska Ludwig
- Eye Center, Freiburg Medical Center, University of Freiburg, 79106 Freiburg, Germany; (C.L.); (F.L.)
| | - Bahareh Ajami
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA;
| | - Peter Wieghofer
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Medical Faculty, Augsburg University, 86159 Augsburg, Germany
- Correspondence:
| |
Collapse
|
37
|
Yu X, Lv J, Wu J, Chen Y, Chen F, Wang L. The autoimmune encephalitis-related cytokine TSLP in the brain primes neuroinflammation by activating the JAK2-NLRP3 axis. Clin Exp Immunol 2022; 207:113-122. [PMID: 35020848 PMCID: PMC8802176 DOI: 10.1093/cei/uxab023] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/08/2021] [Accepted: 11/20/2021] [Indexed: 12/28/2022] Open
Abstract
NLRP3 inflammasome hyperactivation contributes to neuroinflammation in autoimmune disorders, but the underlying regulatory mechanism remains to be elucidated. We demonstrate that compared with wild-type (WT) mice, mice lacking thymic stromal lymphopoietin (TSLP) receptor (TSLPR) (Tslpr−/− mice) exhibit a significantly decreased experimental autoimmune encephalomyelitis (EAE) score, reduced CD4+ T cell infiltration, and restored myelin basic protein (MBP) expression in the brain after EAE induction by myelin oligodendrocyte glycoprotein35–55 (MOG35–55). TSLPR signals through Janus kinase (JAK)2, but not JAK1 or JAK3, to induce NLRP3 expression, and Tslpr−/− mice with EAE show decreased JAK2 phosphorylation and NLRP3 expression in the brain. JAK2 inhibition by ruxolitinib mimicked loss of TSLPR function in vivo and further decreased TSLP expression in the EAE mouse brain. The NLRP3 inhibitor MCC950 decreased CD4+ T cell infiltration, restored MBP expression, and decreased IL-1β and TSLP levels, verifying the pro-inflammatory role of NLRP3. In vitro experiments using BV-2 murine microglia revealed that TSLP directly induced NLRP3 expression, phosphorylation of JAK2 but not JAK1orJAK3, and IL-1β release, which were markedly inhibited by ruxolitinib. Furthermore, EAE induction led to an increase in the Th17 cell number, a decrease in the regulatory T (Treg) cell number in the blood, and an increase in the expression of the cytokine IL-17A in the WT mouse brain, which was drastically reversed in Tslpr−/− mice. In addition, ruxolitinib suppressed the increase in IL-17A expression in the EAE mouse brain. These findings identify TSLP as a prospective target for treating JAK2-NLRP3 axis-associated autoimmune inflammatory disorders.
Collapse
Affiliation(s)
- Xueyuan Yu
- Department of Clinical Laboratory, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jiajia Lv
- Department of Pediatrics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Wu
- Department of Clinical Laboratory, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yong Chen
- Department of Clinical Laboratory, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Fei Chen
- Department of Clinical Laboratory, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Wang
- Department of Clinical Laboratory, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
38
|
Maestrelli F, Cirri M, De Luca E, Biagi D, Mura P. Role of Cyclodextrins and Drug Solid State Properties on Flufenamic Acid Dissolution Performance from Tablets. Pharmaceutics 2022; 14:pharmaceutics14020284. [PMID: 35214017 PMCID: PMC8880332 DOI: 10.3390/pharmaceutics14020284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022] Open
Abstract
Flufenamic acid (FFA) is a non-steroidal anti-inflammatory drug characterised by a low solubility and problems of variable dissolution rate and bio-inequivalence. Different FFA batches, obtained by different suppliers, showed different powder characteristics (particle size, shape and surface properties) that may affect its dissolution behaviour from solid dosage forms. Aim of this work was the improvement of FFA solubility and dissolution rate by the use of cyclodextrins (CDs) and the obtainment of an effective tablet formulation by direct compression. Several CDs have been tested, both in solution and in solid state and several binary systems drug-CDs have been obtained with different techniques, with the scope to select the most effective system. Grinding technique with randomly methylated-β-cyclodextrin (RAMEB) was the only one that allowed the complete drug amorphization, together with the highest improvement in drug dissolution rate, and was then selected for tablets formulation. Conventional and immediate release tablets were obtained and fully characterised for technological properties. In both cases an improved and well reproducible drug dissolution performance was obtained, independently from the FFA supplier and thus no more affected by the differences observed between the original FFA crystalline samples.
Collapse
Affiliation(s)
- Francesca Maestrelli
- Department of Chemistry “U. Schiff”, University of Florence, Via Schiff 6, Sesto Fiorentino, 50019 Florence, Italy; (F.M.); (E.D.L.); (P.M.)
| | - Marzia Cirri
- Department of Chemistry “U. Schiff”, University of Florence, Via Schiff 6, Sesto Fiorentino, 50019 Florence, Italy; (F.M.); (E.D.L.); (P.M.)
- Correspondence:
| | - Enrico De Luca
- Department of Chemistry “U. Schiff”, University of Florence, Via Schiff 6, Sesto Fiorentino, 50019 Florence, Italy; (F.M.); (E.D.L.); (P.M.)
| | - Diletta Biagi
- Menarini Manufacturing Logistic and Services s.r.l. (AMMLS), Via dei Sette Santi 1/3, 50131 Florence, Italy;
| | - Paola Mura
- Department of Chemistry “U. Schiff”, University of Florence, Via Schiff 6, Sesto Fiorentino, 50019 Florence, Italy; (F.M.); (E.D.L.); (P.M.)
| |
Collapse
|
39
|
Pharmacological Modulation and (Patho)Physiological Roles of TRPM4 Channel-Part 2: TRPM4 in Health and Disease. Pharmaceuticals (Basel) 2021; 15:ph15010040. [PMID: 35056097 PMCID: PMC8779181 DOI: 10.3390/ph15010040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 02/06/2023] Open
Abstract
Transient receptor potential melastatin 4 (TRPM4) is a unique member of the TRPM protein family and, similarly to TRPM5, is Ca2+ sensitive and permeable for monovalent but not divalent cations. It is widely expressed in many organs and is involved in several functions; it regulates membrane potential and Ca2+ homeostasis in both excitable and non-excitable cells. This part of the review discusses the currently available knowledge about the physiological and pathophysiological roles of TRPM4 in various tissues. These include the physiological functions of TRPM4 in the cells of the Langerhans islets of the pancreas, in various immune functions, in the regulation of vascular tone, in respiratory and other neuronal activities, in chemosensation, and in renal and cardiac physiology. TRPM4 contributes to pathological conditions such as overactive bladder, endothelial dysfunction, various types of malignant diseases and central nervous system conditions including stroke and injuries as well as in cardiac conditions such as arrhythmias, hypertrophy, and ischemia-reperfusion injuries. TRPM4 claims more and more attention and is likely to be the topic of research in the future.
Collapse
|
40
|
Cui J, Zhao S, Li Y, Zhang D, Wang B, Xie J, Wang J. Regulated cell death: discovery, features and implications for neurodegenerative diseases. Cell Commun Signal 2021; 19:120. [PMID: 34922574 PMCID: PMC8684172 DOI: 10.1186/s12964-021-00799-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/30/2021] [Indexed: 12/18/2022] Open
Abstract
Regulated cell death (RCD) is a ubiquitous process in living organisms that is essential for tissue homeostasis or to restore biological balance under stress. Over the decades, various forms of RCD have been reported and are increasingly being found to involve in human pathologies and clinical outcomes. We focus on five high-profile forms of RCD, including apoptosis, pyroptosis, autophagy-dependent cell death, necroptosis and ferroptosis. Cumulative evidence supports that not only they have different features and various pathways, but also there are extensive cross-talks between modes of cell death. As the understanding of RCD pathway in evolution, development, physiology and disease continues to improve. Here we review an updated classification of RCD on the discovery and features of processes. The prominent focus will be placed on key mechanisms of RCD and its critical role in neurodegenerative disease. Video abstract.
Collapse
Affiliation(s)
- Juntao Cui
- School of Basic Medicine, Qingdao University, Qingdao, 266071 China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071 China
| | - Suhan Zhao
- School of Basic Medicine, Qingdao University, Qingdao, 266071 China
- School of Clinical Medicine, Qingdao University, Qingdao, 266071 China
| | - Yinghui Li
- School of Basic Medicine, Qingdao University, Qingdao, 266071 China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071 China
| | - Danyang Zhang
- School of Basic Medicine, Qingdao University, Qingdao, 266071 China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071 China
| | - Bingjing Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071 China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071 China
| | - Junxia Xie
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071 China
| | - Jun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071 China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071 China
| |
Collapse
|
41
|
Trpm5 channels encode bistability of spinal motoneurons and ensure motor control of hindlimbs in mice. Nat Commun 2021; 12:6815. [PMID: 34819493 PMCID: PMC8613399 DOI: 10.1038/s41467-021-27113-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 11/02/2021] [Indexed: 11/09/2022] Open
Abstract
Bistable motoneurons of the spinal cord exhibit warmth-activated plateau potential driven by Na+ and triggered by a brief excitation. The thermoregulating molecular mechanisms of bistability and their role in motor functions remain unknown. Here, we identify thermosensitive Na+-permeable Trpm5 channels as the main molecular players for bistability in mouse motoneurons. Pharmacological, genetic or computational inhibition of Trpm5 occlude bistable-related properties (slow afterdepolarization, windup, plateau potentials) and reduce spinal locomotor outputs while central pattern generators for locomotion operate normally. At cellular level, Trpm5 is activated by a ryanodine-mediated Ca2+ release and turned off by Ca2+ reuptake through the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump. Mice in which Trpm5 is genetically silenced in most lumbar motoneurons develop hindlimb paresis and show difficulties in executing high-demanding locomotor tasks. Overall, by encoding bistability in motoneurons, Trpm5 appears indispensable for producing a postural tone in hindlimbs and amplifying the locomotor output.
Collapse
|
42
|
Jha RM, Rani A, Desai SM, Raikwar S, Mihaljevic S, Munoz-Casabella A, Kochanek PM, Catapano J, Winkler E, Citerio G, Hemphill JC, Kimberly WT, Narayan R, Sahuquillo J, Sheth KN, Simard JM. Sulfonylurea Receptor 1 in Central Nervous System Injury: An Updated Review. Int J Mol Sci 2021; 22:11899. [PMID: 34769328 PMCID: PMC8584331 DOI: 10.3390/ijms222111899] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Sulfonylurea receptor 1 (SUR1) is a member of the adenosine triphosphate (ATP)-binding cassette (ABC) protein superfamily, encoded by Abcc8, and is recognized as a key mediator of central nervous system (CNS) cellular swelling via the transient receptor potential melastatin 4 (TRPM4) channel. Discovered approximately 20 years ago, this channel is normally absent in the CNS but is transcriptionally upregulated after CNS injury. A comprehensive review on the pathophysiology and role of SUR1 in the CNS was published in 2012. Since then, the breadth and depth of understanding of the involvement of this channel in secondary injury has undergone exponential growth: SUR1-TRPM4 inhibition has been shown to decrease cerebral edema and hemorrhage progression in multiple preclinical models as well as in early clinical studies across a range of CNS diseases including ischemic stroke, traumatic brain injury, cardiac arrest, subarachnoid hemorrhage, spinal cord injury, intracerebral hemorrhage, multiple sclerosis, encephalitis, neuromalignancies, pain, liver failure, status epilepticus, retinopathies and HIV-associated neurocognitive disorder. Given these substantial developments, combined with the timeliness of ongoing clinical trials of SUR1 inhibition, now, another decade later, we review advances pertaining to SUR1-TRPM4 pathobiology in this spectrum of CNS disease-providing an overview of the journey from patch-clamp experiments to phase III trials.
Collapse
Affiliation(s)
- Ruchira M. Jha
- Department of Neurology, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (R.M.J.); (S.M.D.)
- Department of Translational Neuroscience, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (A.R.); (S.R.); (S.M.); (A.M.-C.)
- Department of Neurosurgery, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (J.C.); (E.W.)
| | - Anupama Rani
- Department of Translational Neuroscience, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (A.R.); (S.R.); (S.M.); (A.M.-C.)
| | - Shashvat M. Desai
- Department of Neurology, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (R.M.J.); (S.M.D.)
| | - Sudhanshu Raikwar
- Department of Translational Neuroscience, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (A.R.); (S.R.); (S.M.); (A.M.-C.)
| | - Sandra Mihaljevic
- Department of Translational Neuroscience, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (A.R.); (S.R.); (S.M.); (A.M.-C.)
| | - Amanda Munoz-Casabella
- Department of Translational Neuroscience, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (A.R.); (S.R.); (S.M.); (A.M.-C.)
| | - Patrick M. Kochanek
- Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Joshua Catapano
- Department of Neurosurgery, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (J.C.); (E.W.)
| | - Ethan Winkler
- Department of Neurosurgery, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (J.C.); (E.W.)
| | - Giuseppe Citerio
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy;
- Neurointensive Care Unit, Department of Neuroscience, San Gerardo Hospital, ASST—Monza, 20900 Monza, Italy
| | - J. Claude Hemphill
- Department of Neurology, University of California, San Francisco, CA 94143, USA;
| | - W. Taylor Kimberly
- Division of Neurocritical Care and Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Raj Narayan
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, North Shore University Hospital, Manhasset, NY 11549, USA;
| | - Juan Sahuquillo
- Neurotrauma and Neurosurgery Research Unit (UNINN), Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain;
- Neurotraumatology and Neurosurgery Research Unit, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
- Department of Neurosurgery, Vall d’Hebron University Hospital, 08035 Barcelona, Spain
| | - Kevin N. Sheth
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, School of Medicine, Yale University, New Haven, CT 06510, USA;
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
43
|
Investigation of Novel Small Molecular TRPM4 Inhibitors in Colorectal Cancer Cells. Cancers (Basel) 2021; 13:cancers13215400. [PMID: 34771564 PMCID: PMC8582472 DOI: 10.3390/cancers13215400] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Transient receptor potential melastatin 4 (TRPM4) ion channel malfunction or aberrant expression is implicated in many diseases, including different cancers and cardiovascular diseases. Currently, there is a need for specific and potent TRPM4 inhibitors. They would allow to study the role of TRPM4 in disease models and to validate it as a potential target in therapies, including anti-cancer therapy. In colorectal cancer (CRC), TRPM4 is upregulated, and its conductivity plays a role in the regulation of viability and cell cycle of CRC cells. In this study, we tested three novel TRPM4 inhibitors, CBA, NBA, and LBA, in CRC cells. In HCT116 cells, we show that NBA inhibits TRPM4 currents in the micromolar range and alters proliferation and cell cycle. Furthermore, NBA decreases the viability of Colo205 cells. This makes NBA a promising candidate for further evaluation as a specific TRPM4 inhibitor in other cellular systems and disease models. Abstract (1) Background: Transient receptor potential melastatin (TRPM4) ion channel aberrant expression or malfunction contributes to different types of cancer, including colorectal cancer (CRC). However, TRPM4 still needs to be validated as a potential target in anti-cancer therapy. Currently, the lack of potent and selective TRPM4 inhibitors limits further studies on TRPM4 in cancer disease models. In this study, we validated novel TRPM4 inhibitors, CBA, NBA, and LBA, in CRC cells. (2) Methods: The potency to inhibit TRPM4 conductivity in CRC cells was assessed with the whole-cell patch clamp technique. Furthermore, the impact of TRPM4 inhibitors on cellular functions, such as viability, proliferation, and cell cycle, were assessed in cellular assays. (3) Results: We show that in CRC cells, novel TRPM4 inhibitors irreversibly block TRPM4 currents in a low micromolar range. NBA decreases proliferation and alters the cell cycle in HCT116 cells. Furthermore, NBA reduces the viability of the Colo205 cell line, which highly expresses TRPM4. (4) Conclusions: NBA is a promising new TRPM4 inhibitor candidate, which could be used to study the role of TRPM4 in cancer disease models and other diseases.
Collapse
|
44
|
Çoban G, Yildiz P, Doğan B, Şahin N, Gücin Z. Expression of transient receptor potential melastatin 4 in differential diagnosis of eosinophilic renal tumors. Mol Clin Oncol 2021; 15:230. [PMID: 34631055 DOI: 10.3892/mco.2021.2393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 07/02/2021] [Indexed: 11/05/2022] Open
Abstract
Immunohistochemical and molecular studies to differentiate eosinophilic kidney tumors are gradually increasing. The present study investigated the role of transient receptor potential cation channel subfamily M member 4 (TRPM4), a non-selective cation channel associated with migration, proliferation and invasion in cancer cells, in this differentiation. The aim was to investigate the effectiveness of TRPM4 in differentiation of eosinophilic kidney tumors. The study included a total of 112 patients, including 97 eosinophilic kidney tumors with the diagnoses of 33 eosinophilic clear cell renal cell carcinoma (CCRCC), 35 eosinophilic chromophobe renal cell carcinoma (ChRCC), 8 papillary renal cell carcinoma type 2 (P2RCC), 21 renal oncocytoma (RO), as well as 15 papillary renal cell carcinoma type 1 to differentiate from P2RCC. For TRPM4, diffuse staining (>10%) was observed in 2 CCRCC, 15 ChRCC, 20 RO and 4 P2RCC cases. There was a significant difference between eosinophilic CCRCC and other eosinophilic tumors (P<0.05). While basolateral staining was observed in papillary tumors, membrane staining was observed in other stained cases. It was hypothesized that the use of TRPM4 along with morphological findings, cytokeratin 7 and other markers may be useful for the differentiation of eosinophilic kidney tumors.
Collapse
Affiliation(s)
- Ganime Çoban
- Department of Pathology, Faculty of Medicine, Bezmialem Vakif University, Istanbul 34093, Turkey
| | - Pelin Yildiz
- Department of Pathology, Faculty of Medicine, Bezmialem Vakif University, Istanbul 34093, Turkey
| | - Bayram Doğan
- Department of Urology, Faculty of Medicine, Bezmialem Vakif University, Istanbul 34093, Turkey
| | - Nurhan Şahin
- Department of Pathology, Faculty of Medicine, Bezmialem Vakif University, Istanbul 34093, Turkey
| | - Zühal Gücin
- Department of Pathology, Faculty of Medicine, Bezmialem Vakif University, Istanbul 34093, Turkey
| |
Collapse
|
45
|
Advances in TRP channel drug discovery: from target validation to clinical studies. Nat Rev Drug Discov 2021; 21:41-59. [PMID: 34526696 PMCID: PMC8442523 DOI: 10.1038/s41573-021-00268-4] [Citation(s) in RCA: 291] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 12/20/2022]
Abstract
Transient receptor potential (TRP) channels are multifunctional signalling molecules with many roles in sensory perception and cellular physiology. Therefore, it is not surprising that TRP channels have been implicated in numerous diseases, including hereditary disorders caused by defects in genes encoding TRP channels (TRP channelopathies). Most TRP channels are located at the cell surface, which makes them generally accessible drug targets. Early drug discovery efforts to target TRP channels focused on pain, but as our knowledge of TRP channels and their role in health and disease has grown, these efforts have expanded into new clinical indications, ranging from respiratory disorders through neurological and psychiatric diseases to diabetes and cancer. In this Review, we discuss recent findings in TRP channel structural biology that can affect both drug development and clinical indications. We also discuss the clinical promise of novel TRP channel modulators, aimed at both established and emerging targets. Last, we address the challenges that these compounds may face in clinical practice, including the need for carefully targeted approaches to minimize potential side-effects due to the multifunctional roles of TRP channels.
Collapse
|
46
|
Lowes DJ, Miao J, Al-Waqfi RA, Avad KA, Hevener KE, Peters BM. Identification of Dual-Target Compounds with Antifungal and Anti-NLRP3 Inflammasome Activity. ACS Infect Dis 2021; 7:2522-2535. [PMID: 34260210 PMCID: PMC11344480 DOI: 10.1021/acsinfecdis.1c00270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Invasive and superficial infections caused by the Candida species result in significant global morbidity and mortality. As the pathogenicity of these organisms is intimately intertwined with host immune response, therapies to target both the fungus and host inflammation may be warranted. Structural similarities exist between established inhibitors of the NLRP3 inflammasome and those of fungal acetohydroxyacid synthase (AHAS). Therefore, we leveraged this information to conduct an in silico molecular docking screen to find novel polypharmacologic inhibitors of these targets that resulted in the identification of 12 candidate molecules. Of these, compound 10 significantly attenuated activation of the NLPR3 inflammasome by LPS + ATP, while also demonstrating growth inhibitory activity against C. albicans that was alleviated in the presence of exogenous branched chain amino acids, consistent with targeting of fungal AHAS. SAR studies delineated an essential molecular scaffold required for dual activity. Ultimately, 10 and its analog 10a resulted in IC50 (IL-1β release) and MIC50 (fungal growth) values with low μM potency against several Candida species. Collectively, this work demonstrates promising potential of dual-target approaches for improved management of fungal infections.
Collapse
Affiliation(s)
- David J Lowes
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Jian Miao
- Graduate Program in Pharmaceutical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Rand A Al-Waqfi
- Graduate Program in Pharmaceutical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Kristiana A Avad
- Graduate Program in Pharmaceutical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Doctor of Pharmacy Program, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Kirk E Hevener
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
47
|
Shi R, Fu Y, Zhao D, Boczek T, Wang W, Guo F. Cell death modulation by transient receptor potential melastatin channels TRPM2 and TRPM7 and their underlying molecular mechanisms. Biochem Pharmacol 2021; 190:114664. [PMID: 34175300 DOI: 10.1016/j.bcp.2021.114664] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
Transient receptor potential melastatin (TRPM) channels are members of the transient receptor potential (TRP) channels, a family of evolutionarily conserved integral membrane proteins. TRPM channels are nonselective cation channels, mediating the influx of various ions including Ca2+, Na+ and Zn2+. The function of TRPM channels is vital for cell proliferation, cell development and cell death. Cell death is a key procedure during embryonic development, organism homeostasis, aging and disease. The category of cell death modalities, beyond the traditionally defined concepts of necrosis, autophagy, and apoptosis, were extended with the discovery of pyroptosis, necroptosis and ferroptosis. As upstream signaling regulators of cell death, TRPM channels have been involved inrelevant pathologies. In this review, we introduced several cell death modalities, then summarized the contribution of TRPM channels (especially TRPM2 and TRPM7) to different cell death modalities and discussed the underlying regulatory mechanisms. Our work highlighted the possibility of TRPM channels as potential therapeutic targets in cell death-related diseases.
Collapse
Affiliation(s)
- Ruixue Shi
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yu Fu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Dongyi Zhao
- The University of Tokyo, Department of Pharmaceutical Science, 1130033, Japan
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, 92215, Poland.
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
48
|
Wang C, Chen J, Wang M, Naruse K, Takahashi K. Role of the TRPM4 channel in mitochondrial function, calcium release, and ROS generation in oxidative stress. Biochem Biophys Res Commun 2021; 566:190-196. [PMID: 34144257 DOI: 10.1016/j.bbrc.2021.03.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 01/09/2023]
Abstract
Ischemic heart disease is one of the most common causes of death worldwide. Mitochondrial dysfunction, excessive reactive oxygen species (ROS) generation, and calcium (Ca2+) overload are three key factors leading to myocardial death during ischemia-reperfusion (I/R) injury. Inhibition of TRPM4, a Ca2+-activated nonselective cation channel, protects the rat heart from I/R injury, but the specific mechanism underlying this effect is unclear. In this study, we investigated the mechanism of cardioprotection against I/R injury via TRPM4 using hydrogen peroxide (H2O2), a major contributor to oxidative stress, as an I/R injury model. We knocked out the TRPM4 gene in the rat cardiomyocyte cell line H9c2 using CRISPR/Cas9. Upon H2O2 treatment, intracellular Ca2+ level and ROS production increased in wild type (WT) cells but not in TRPM4 knockout (TRPM4KO) cells. With this treatment, two indicators of mitochondrial function, mitochondrial membrane potential (ΔΨm) and intracellular ATP levels, decreased in WT but not in TRPM4KO cells. Taken together, these findings suggest that blockade of the TRPM4 channel might protect the myocardium from oxidative stress by maintaining the mitochondrial membrane potential and intracellular ATP levels, possibly through preventing aberrant increases in intracellular Ca2+ and ROS.
Collapse
Affiliation(s)
- Chen Wang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Jian Chen
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan; Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Mengxue Wang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Keiji Naruse
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| |
Collapse
|
49
|
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
|
50
|
Woo MS, Ufer F, Rothammer N, Di Liberto G, Binkle L, Haferkamp U, Sonner JK, Engler JB, Hornig S, Bauer S, Wagner I, Egervari K, Raber J, Duvoisin RM, Pless O, Merkler D, Friese MA. Neuronal metabotropic glutamate receptor 8 protects against neurodegeneration in CNS inflammation. J Exp Med 2021; 218:e20201290. [PMID: 33661276 PMCID: PMC7938362 DOI: 10.1084/jem.20201290] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 12/17/2020] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system with continuous neuronal loss. Treatment of clinical progression remains challenging due to lack of insights into inflammation-induced neurodegenerative pathways. Here, we show that an imbalance in the neuronal receptor interactome is driving glutamate excitotoxicity in neurons of MS patients and identify the MS risk-associated metabotropic glutamate receptor 8 (GRM8) as a decisive modulator. Mechanistically, GRM8 activation counteracted neuronal cAMP accumulation, thereby directly desensitizing the inositol 1,4,5-trisphosphate receptor (IP3R). This profoundly limited glutamate-induced calcium release from the endoplasmic reticulum and subsequent cell death. Notably, we found Grm8-deficient neurons to be more prone to glutamate excitotoxicity, whereas pharmacological activation of GRM8 augmented neuroprotection in mouse and human neurons as well as in a preclinical mouse model of MS. Thus, we demonstrate that GRM8 conveys neuronal resilience to CNS inflammation and is a promising neuroprotective target with broad therapeutic implications.
Collapse
Affiliation(s)
- Marcel S. Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Ufer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Giovanni Di Liberto
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Lars Binkle
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology, Hamburg, Germany
| | - Jana K. Sonner
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Sönke Hornig
- Experimentelle Neuropädiatrie, Klinik für Kinder und Jugendmedizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Bauer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Kristof Egervari
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR
| | - Robert M. Duvoisin
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology, Hamburg, Germany
| | - Doron Merkler
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Manuel A. Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|