1
|
Martins-Marques T, Vasconcelos-Cardoso M, Geli MI, Aasen T, Kwak BR, Girao H. The ins and outs of connexins and pannexins beyond the cell surface. Trends Biochem Sci 2025:S0968-0004(25)00102-1. [PMID: 40382259 DOI: 10.1016/j.tibs.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/11/2025] [Accepted: 04/23/2025] [Indexed: 05/20/2025]
Abstract
Classically implicated in the transport of small molecules between neighboring cells or into the extracellular milieu, emerging evidence implicates connexins and pannexins in other noncanonical biological functions. Propelled by recent technological advances and genetic approaches, this review aims to provide a comprehensive and thought-provoking perspective on unconventional functions of connexins and pannexins that could be shared with other cell surface proteins. Although multiple studies have linked dysfunctional channel activity with human disorders, the contribution of non-junctional roles of connexins and pannexins during disease pathophysiology is now beginning to blossom. We highlight the potential regulatory signals and players involved in unfamiliar connexin and pannexin activities that can pave the way to design novel therapeutic tools.
Collapse
Affiliation(s)
- Tania Martins-Marques
- Università di Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Università di Coimbra, Faculty of Medicine, Coimbra, Portugal; Università di Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Maria Vasconcelos-Cardoso
- Università di Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Università di Coimbra, Faculty of Medicine, Coimbra, Portugal; Università di Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Maria Isabel Geli
- Institute for Molecular Biology of Barcelona, CSIC, Barcelona, Spain
| | - Trond Aasen
- Vall d´Hebron Institute of Research, CIBERONC, Barcelona, Spain
| | - Brenda R Kwak
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Henrique Girao
- Università di Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Università di Coimbra, Faculty of Medicine, Coimbra, Portugal; Università di Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical and Academic Centre of Coimbra, Coimbra, Portugal.
| |
Collapse
|
2
|
Henze E, Burkhardt RN, Fox BW, Schwertfeger TJ, Gelsleichter E, Michalski K, Kramer L, Lenfest M, Boesch JM, Lin H, Schroeder FC, Kawate T. ATP-release pannexin channels are gated by lysophospholipids. eLife 2025; 14:RP107067. [PMID: 40309905 PMCID: PMC12045621 DOI: 10.7554/elife.107067] [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: 05/02/2025] Open
Abstract
In addition to its role as cellular energy currency, adenosine triphosphate (ATP) serves as an extracellular messenger that mediates diverse cell-to-cell communication. Compelling evidence supports that ATP is released from cells through pannexins, a family of membrane proteins that form heptameric large-pore channels. However, the activation mechanisms that trigger ATP release by pannexins remain poorly understood. Here, we discover lysophospholipids as endogenous pannexin activators, using activity-guided fractionation of mouse tissue extracts combined with untargeted metabolomics and electrophysiology. We show that lysophospholipids directly and reversibly activate pannexins in the absence of other proteins. Secretomics experiments reveal that lysophospholipid-activated pannexin 1 leads to the release of not only ATP but also other signaling metabolites, such as 5'-methylthioadenosine, which is important for immunomodulation. We also demonstrate that lysophospholipids activate endogenous pannexin 1 in human monocytes, leading to the release of IL-1β through inflammasome activation. Our results provide a connection between lipid metabolism and purinergic signaling, both of which play major roles in immune responses.
Collapse
Affiliation(s)
- Erik Henze
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| | - Russell N Burkhardt
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Bennett William Fox
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Tyler J Schwertfeger
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Eric Gelsleichter
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Kevin Michalski
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| | - Lydia Kramer
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| | - Margret Lenfest
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Jordyn M Boesch
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
- Department of Molecular Biology and Genetics, Cornell UniversityIthacaUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Frank C Schroeder
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| |
Collapse
|
3
|
Illanes-González J, Flores-Muñoz C, Vitureira N, Ardiles ÁO. Pannexin 1 channels: A bridge between synaptic plasticity and learning and memory processes. Neurosci Biobehav Rev 2025; 174:106173. [PMID: 40274202 DOI: 10.1016/j.neubiorev.2025.106173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/13/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
The Pannexin 1 channel is a membrane protein widely expressed in various vertebrate cell types, including microglia, astrocytes, and neurons within the central nervous system. Growing research has demonstrated the significant involvement of Panx1 in synaptic physiology, such as its contribution to long-term synaptic plasticity, with a particular focus on the hippocampus, an essential structure for learning and memory. Investigations studying the role of Panx1 in synaptic plasticity have utilized knockout animal models and channel inhibition techniques, revealing that the absence or blockade of Panx1 channels in this region promotes synaptic potentiation, dendritic arborization, and spine formation. Despite substantial progress, the precise mechanism by which Panx1 regulates synaptic plasticity remains to be determined. Nevertheless, evidence suggests that Panx1 may exert its influence by releasing signaling molecules, such as adenosine triphosphate (ATP), or through the clearance of endocannabinoids (eCBs). This review aims to comprehensively explore the current literature on the role of Panx1 in synapses. By examining relevant articles, we seek to enhance our understanding of Panx1's contribution to synaptic fundamental processes and the potential implications for cognitive function.
Collapse
Affiliation(s)
- Javiera Illanes-González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; Centro para la Investigación Traslacional en Neurofarmacología, CItNe, Universidad de Valparaíso, Valparaíso, Chile
| | - Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; Centro para la Investigación Traslacional en Neurofarmacología, CItNe, Universidad de Valparaíso, Valparaíso, Chile
| | - Nathalia Vitureira
- Unidad Académica de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Álvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; Centro para la Investigación Traslacional en Neurofarmacología, CItNe, Universidad de Valparaíso, Valparaíso, Chile; Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
4
|
Wang J, Kuang S, Wei Z, Liang S. Research progress of connexins in epileptogensis. ACTA EPILEPTOLOGICA 2025; 7:14. [PMID: 40217413 PMCID: PMC11960343 DOI: 10.1186/s42494-025-00203-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/12/2025] [Indexed: 04/15/2025] Open
Abstract
Epilepsy, a chronic neurological disorder, is characterized by dysfunction in neural networks. Gap junctions and hemichannels, which are integral to the astrocyte connection network, play a critical role in epilepsy. Connexins, the components of astrocyte gap junctions and hemichannels, can be activated to transfer glutamate, adenosine triphosphate, and other chemicals, potentially leading to seizures. Connexins therefore hold significant potential for epilepsy treatment. This review focuses on connexin 43 and provides a brief overview of other connexins and pannexin 1. Understanding the relationship between connexins and epilepsy offers theoretical support for developing new antiseizure medications.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of Functional Neurosurgery, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, No. 56, South Lishi Road, Xicheng District, Beijing, 100045, China
| | - Suhui Kuang
- Department of Functional Neurosurgery, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, No. 56, South Lishi Road, Xicheng District, Beijing, 100045, China
| | - Zhirong Wei
- Department of Functional Neurosurgery, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, No. 56, South Lishi Road, Xicheng District, Beijing, 100045, China
| | - Shuli Liang
- Department of Functional Neurosurgery, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, No. 56, South Lishi Road, Xicheng District, Beijing, 100045, China.
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, 100045, China.
| |
Collapse
|
5
|
Martins-Silva C, Anderson CL, Boyce AKJ, Andrade TES, Tizziani T, Lopes KHS, Micke GA, Cregan SP, Dos Santos ARS, Thompson RJ. The Ethanolic Extract of Polygala paniculata L. Blocks Panx1 Channels and Reduces Ischemic Brain Infarct in a Dose- and Sex-Dependent Way. Mol Neurobiol 2025; 62:3258-3275. [PMID: 39271622 DOI: 10.1007/s12035-024-04453-5] [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: 05/27/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Polygala paniculata L. is a native plant from tropical America. The therapeutic potential of the hydroalcoholic extract of P. paniculata (HEPp) has been scientifically explored due to folk medicine reports on its action against several afflictions. HEPp contains several bioactive molecules with neuroprotective activities, making it a promising candidate for stroke treatment. This study used electrophysiological, biochemical, and in vivo experiments to evaluate the molecular mechanisms underlying HEPp as a neuroprotective therapy for stroke targeting Pannexin-1 (Panx1). Panx1 is a non-selective channel that opens during ischemia and contributes to neuronal death. HEPp was not toxic to cortical neurons and pre-treatment with the extract reduced neuronal death promoted by oxygen and glucose deprivation in a dose-dependent manner. Additionally, HEPp blocked Panx1 currents in a dose-dependent manner and the effect, which was shown to be partially due to rutin. Animals submitted to photothrombosis and post-treated with HEPp had reduced infarct volume, and the effective dose was lower in males (1 mg/kg) than in females (10 mg/kg). On the other hand, in Panx1 KD mice (50% Panx1 levels), the acute treatment reduced the infarct volume only in males. Upon chronic treatment with HEPp, a reduction in Panx1 protein levels was observed. The current study provides reliable evidence of the neuroprotective properties of HEPp in both in vitro and in vivo models of stroke. The underlying mechanism involves, at least in part, the inhibition of Panx1 channel function and possibly downregulation of protein levels, suppressing the secondary events that lead to apoptosis and inflammation.
Collapse
Affiliation(s)
- Cristina Martins-Silva
- Department of Physiological Sciences, Health Sciences Center, Laboratory of Neurochemistry and Behaviour (LabNeC), Graduate Program in Biochemistry, Federal University of Espirito Santo, Vitoria, ES, 29043910, Brazil.
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada.
| | - Connor L Anderson
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Andrew K J Boyce
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Tassiane E S Andrade
- Laboratory of Neurobiology of Pain and Inflammation, Department of Physiological Sciences, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
- Neuroscience Program, Department of Physiology and Pharmacology, University of Western Ontario, Robarts Research Institute, University of Western Ontario, 100 Perth Dr, London, ON, N6A 5K8, Canada
| | - Tiago Tizziani
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Kheytiany H S Lopes
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Gustavo A Micke
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Sean P Cregan
- Neuroscience Program, Department of Physiology and Pharmacology, University of Western Ontario, Robarts Research Institute, University of Western Ontario, 100 Perth Dr, London, ON, N6A 5K8, Canada
| | - Adair Roberto Soares Dos Santos
- Laboratory of Neurobiology of Pain and Inflammation, Department of Physiological Sciences, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Roger J Thompson
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada
| |
Collapse
|
6
|
Henze E, Burkhardt RN, Fox BW, Schwertfeger TJ, Gelsleichter E, Michalski K, Kramer L, Lenfest M, Boesch JM, Lin H, Schroeder FC, Kawate T. ATP-release pannexin channels are gated by lysophospholipids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.23.563601. [PMID: 37961151 PMCID: PMC10634739 DOI: 10.1101/2023.10.23.563601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In addition to its role as cellular energy currency, adenosine triphosphate (ATP) serves as an extracellular messenger that mediates diverse cell-to-cell communication. Compelling evidence supports that ATP is released from cells through pannexins, a family of membrane proteins that form heptameric large-pore channels. However, the activation mechanisms that trigger ATP release by pannexins remain poorly understood. Here, we discover lysophospholipids as endogenous pannexin activators, using activity-guided fractionation of mouse tissue extracts combined with untargeted metabolomics and electrophysiology. We show that lysophospholipids directly and reversibly activate pannexins in the absence of other proteins. Secretomics experiments reveal that lysophospholipid-activated pannexin 1 leads to the release of not only ATP but also other signaling metabolites, such as 5'-methylthioadenosine, which is important for immunomodulation. We also demonstrate that lysophospholipids activate endogenous pannexin 1 in human monocytes, leading to the release of IL-1β through inflammasome activation. Our results provide a connection between lipid metabolism and purinergic signaling, both of which play major roles in immune responses.
Collapse
|
7
|
Li J, Lou L, Chen W, Qiang X, Zhu C, Wang H. Connexin 43 and Pannexin 1 hemichannels as endogenous regulators of innate immunity in sepsis. Front Immunol 2024; 15:1523306. [PMID: 39763679 PMCID: PMC11701031 DOI: 10.3389/fimmu.2024.1523306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/05/2024] [Indexed: 02/02/2025] Open
Abstract
Sepsis is a life-threatening organ dysfunction resulting from a dysregulated host response to infections that is initiated by the body's innate immune system. Nearly a decade ago, we discovered that bacterial lipopolysaccharide (LPS) and serum amyloid A (SAA) upregulated Connexin 43 (Cx43) and Pannexin 1 (Panx1) hemichannels in macrophages. When overexpressed, these hemichannels contribute to sepsis pathogenesis by promoting ATP efflux, which intensifies the double-stranded RNA-activated protein kinase R (PKR)-dependent inflammasome activation, pyroptosis, and the release of pathogenic damage-associated molecular pattern (DAMP) molecules, such as HMGB1. Mimetic peptides targeting specific regions of Cx43 and Panx1 can distinctly modulate hemichannel activity in vitro, and diversely impact sepsis-induced lethality in vivo. Along with extensive supporting evidence from others, we now propose that hemichannel molecules play critical roles as endogenous regulators of innate immunity in sepsis.
Collapse
Affiliation(s)
- Jianhua Li
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Li Lou
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Weiqiang Chen
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Xiaoling Qiang
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Cassie Zhu
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Haichao Wang
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
8
|
Nielsen AC, Anderson CL, Ens C, Boyce AKJ, Thompson RJ. Non-ionotropic NMDAR signalling activates Panx1 to induce P2X4R-dependent long-term depression in the hippocampus. J Physiol 2024. [PMID: 39709529 DOI: 10.1113/jp285193] [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: 04/01/2024] [Accepted: 11/28/2024] [Indexed: 12/23/2024] Open
Abstract
In recent years, evidence supporting non-ionotropic signalling by the NMDA receptor (niNMDAR) has emerged, including roles in long-term depression (LTD). Here, we investigated whether niNMDAR-pannexin-1 (Panx1) contributes to LTD at the CA3-CA1 hippocampal synapse. Using whole-cell, patch clamp electrophysiology in rat hippocampal slices, we show that a low-frequency stimulation (3 Hz) of the Schaffer collaterals produces LTD that is blocked by continuous but not transient application of the NMDAR competitive antagonist, MK-801. After transient MK-801, LTD involved pannexin-1 and sarcoma (Src) kinase. We show that pannexin-1 is not permeable to Ca2+, but probably releases ATP to induce LTD via P2X4 purinergic receptors because LTD after transient MK-801 application was prevented by 5-BDBD. Thus, we conclude that niNMDAR activation of Panx1 can link glutamatergic and purinergic pathways to produce LTD following low frequency synaptic stimulation when NMDARs are transiently inhibited. KEY POINTS: Differential effect of short-term D-APV and MK-801 application on long-term depression (LTD) suggests that the NMDA receptor (niNMDAR) contributes to later phases of synaptic depression. niNMDAR LTD involved sarcoma (Src) kinase and pannexin-1 (Panx1), which is a pathway previously identified to be active during excitotoxicity. Panx1 was not calcium permeable but may contribute to late phase LTD via ATP release. Panx1 blockers prevent LTD, and this was rescued with exogenous ATP application. Inhibition of LTD with 5-BDBD suggests the downstream involvement of postsynaptic P2X4 receptors.
Collapse
Affiliation(s)
- Allison C Nielsen
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Connor L Anderson
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carina Ens
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Andrew K J Boyce
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Roger J Thompson
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
9
|
Henze E, Ehrlich JJ, Robertson JL, Gelsleichter E, Kawate T. The C-terminal activating domain promotes pannexin 1 channel opening. Proc Natl Acad Sci U S A 2024; 121:e2411898121. [PMID: 39671183 DOI: 10.1073/pnas.2411898121] [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: 06/13/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
Pannexin 1 (Panx1) constitutes a large pore channel responsible for the release of adenosine triphosphate (ATP) from apoptotic cells. Strong evidence indicates that caspase-mediated cleavage of the C-terminus promotes the opening of the Panx1 channel by unplugging the pore. However, this simple pore-plugging mechanism alone cannot account for the observation that a Panx1 construct ending before the caspase cleavage site remains closed. Here, we show that a helical region located immediately before the caspase cleavage site, referred to as the "C-terminal activating domain (CAD)", plays a pivotal role in facilitating Panx1 activation. Electrophysiology and mutagenesis studies uncovered that two conserved leucine residues within the CAD play a pivotal role. Cryoelectron microscopy (Cryo-EM) analysis of the construct ending before reaching the CAD demonstrated that the N terminus extends into an intracellular pocket. In contrast, the construct including the CAD revealed that this domain occupies the intracellular pocket, causing the N terminus to flip upward within the pore. Analysis of electrostatic free energy landscape in the closed conformation indicated that the intracellular side of the ion permeation pore may be occupied by anions like ATP, creating an electrostatic barrier for anions attempting to permeate the pore. When the N terminus flips up, it diminishes the positively charged surface, thereby reducing the drive to accumulate anions inside the pore. This dynamic change in the electrostatic landscape likely contributes to the selection of permeant ions. Collectively, these experiments put forth a mechanism in which C-terminal cleavage liberates the CAD, causing the repositioning of the N terminus to promote Panx1 channel opening.
Collapse
Affiliation(s)
- Erik Henze
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | | | - Janice L Robertson
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Eric Gelsleichter
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| |
Collapse
|
10
|
Majumder D. Ischemic Stroke: Pathophysiology and Evolving Treatment Approaches. Neurosci Insights 2024; 19:26331055241292600. [PMID: 39444789 PMCID: PMC11497522 DOI: 10.1177/26331055241292600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
Stroke remains a leading cause of mortality and disability, with ischemic stroke being the most common type. It occurs due to reduced cerebral blood flow, leading to a cascade of events initiated by oxygen and nutrient deprivation, triggering excitotoxicity, oxidative stress, and inflammation and finally culminating in neuronal injury and death. Key molecular players in ischemic stroke include glutamate receptors, acid-sensing ion channels, and purinergic receptors, exacerbating cellular damage through calcium influx, oxidative stress, and mitochondrial dysfunction. Understanding these mechanisms has shaped therapeutic strategies, such as neuroprotective agents and stem cell therapies. Current treatments such as tissue plasminogen activator (tPA) emphasize timely intervention, yet challenges persist in patient-specific variability and accessibility. This review provides an overview of ischemic stroke pathophysiology, emphasizing cellular responses to ischemia and current and future therapeutic approaches including stem cell therapies aimed at mitigating stroke-induced disabilities and improving long-term outcomes.
Collapse
|
11
|
Villéga F, Fernandes A, Jézéquel J, Uyttersprot F, Benac N, Zenagui S, Bastardo L, Gréa H, Bouchet D, Villetelle L, Nicole O, Rogemond V, Honnorat J, Dupuis JP, Groc L. Ketamine alleviates NMDA receptor hypofunction through synaptic trapping. Neuron 2024; 112:3311-3328.e9. [PMID: 39047728 DOI: 10.1016/j.neuron.2024.06.028] [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: 10/26/2023] [Revised: 04/16/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024]
Abstract
Activity-dependent modulations of N-methyl-D-aspartate glutamate receptor (NMDAR) trapping at synapses regulate excitatory neurotransmission and shape cognitive functions. Although NMDAR synaptic destabilization has been associated with severe neurological and psychiatric conditions, tuning NMDAR synaptic trapping to assess its clinical relevance for the treatment of brain conditions remains a challenge. Here, we report that ketamine (KET) and other clinically relevant NMDAR open channel blockers (OCBs) promote interactions between NMDAR and PDZ-domain-containing scaffolding proteins and enhance NMDAR trapping at synapses. We further show that KET-elicited trapping enhancement compensates for depletion in synaptic receptors triggered by autoantibodies from patients with anti-NMDAR encephalitis. Preventing synaptic depletion mitigates impairments in NMDAR-mediated CaMKII signaling and alleviates anxiety- and sensorimotor-gating-related behavioral deficits provoked by autoantibodies. Altogether, these findings reveal an unexpected dimension of OCB action and stress the potential of targeting receptor anchoring in NMDAR-related synaptopathies.
Collapse
Affiliation(s)
- Frédéric Villéga
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France; Department of Pediatric Neurology, CIC-1401, University Children's Hospital of Bordeaux, Bordeaux, France
| | - Alexandra Fernandes
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Julie Jézéquel
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Floriane Uyttersprot
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Nathan Benac
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Sarra Zenagui
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Laurine Bastardo
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Hélène Gréa
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Delphine Bouchet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Léa Villetelle
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Olivier Nicole
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Véronique Rogemond
- Synaptopathies and Autoantibodies Team, Institut NeuroMyoGene-MeLis, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon1, 69373 Lyon, France; French Reference Centre on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, 69677 Bron, France
| | - Jérôme Honnorat
- Synaptopathies and Autoantibodies Team, Institut NeuroMyoGene-MeLis, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon1, 69373 Lyon, France; French Reference Centre on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, 69677 Bron, France
| | - Julien P Dupuis
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France.
| | - Laurent Groc
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France.
| |
Collapse
|
12
|
Li X, Shi W, Zhao Z, Matsuura T, Lu J, Che J, Chen Q, Zhou Z, Xue M, Hao S, Xu F, Bi G, Kaang B, Collingridge GL, Zhuo M. Increased GluK1 Subunit Receptors in Corticostriatal Projection from the Anterior Cingulate Cortex Contributed to Seizure-Like Activities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308444. [PMID: 39225597 PMCID: PMC11497107 DOI: 10.1002/advs.202308444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 07/26/2024] [Indexed: 09/04/2024]
Abstract
The corticostriatal connection plays a crucial role in cognitive, emotional, and motor control. However, the specific roles and synaptic transmissions of corticostriatal connection are less studied, especially the corticostriatal transmission from the anterior cingulate cortex (ACC). Here, a direct glutamatergic excitatory synaptic transmission in the corticostriatal projection from the ACC is found. Kainate receptors (KAR)-mediated synaptic transmission is increased in this corticostriatal connection both in vitro and in vivo seizure-like activities. GluK1 containing KARs and downstream calcium-stimulated adenylyl cyclase subtype 1 (AC1) are involved in the upregulation of KARs following seizure-like activities. Inhibiting the activities of ACC or its corticostriatal connection significantly attenuated pentylenetetrazole (PTZ)-induced seizure. Additionally, injection of GluK1 receptor antagonist UBP310 or the AC1 inhibitor NB001 both show antiepileptic effects. The studies provide direct evidence that KARs are involved in seizure activity in the corticostriatal connection and the KAR-AC1 signaling pathway is a potential novel antiepileptic strategy.
Collapse
Affiliation(s)
- Xu‐Hui Li
- Center for Neuron and DiseaseFrontier Institute of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
- Department of PhysiologyFaculty of MedicineUniversity of TorontoMedical Science Building, 1 King's College CircleTorontoOntarioM5S 1A8Canada
| | - Wantong Shi
- Center for Neuron and DiseaseFrontier Institute of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Zhi‐Xia Zhao
- Center for Neuron and DiseaseFrontier Institute of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Takanori Matsuura
- Department of PhysiologyFaculty of MedicineUniversity of TorontoMedical Science Building, 1 King's College CircleTorontoOntarioM5S 1A8Canada
- Department of OrthopaedicsSchool of MedicineUniversity of Occupational and Environmental HealthYahatanishi‐kuKitakyushu807–8555Japan
| | - Jing‐Shan Lu
- Center for Neuron and DiseaseFrontier Institute of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Jingmin Che
- Shaanxi Provincial Key Laboratory of Infection and Immune DiseasesShaanxi Provincial People's HospitalXi'anShaanxi710068China
| | - Qi‐Yu Chen
- CAS Key Laboratory of Brain Connectome and ManipulationInterdisciplinary Center for Brain InformationThe Brain Cognition and Brain Disease InstituteShenzhen Institute of Advanced TechnologyShenzhenGuangdong518055China
| | - Zhaoxiang Zhou
- Department of PhysiologyFaculty of MedicineUniversity of TorontoMedical Science Building, 1 King's College CircleTorontoOntarioM5S 1A8Canada
- Department of NeurologyFirst Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510130China
| | - Man Xue
- Center for Neuron and DiseaseFrontier Institute of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Shun Hao
- Department of PharmacologyQingdao University School of PharmacyQingdaoShandong266071China
| | - Fang Xu
- CAS Key Laboratory of Brain Connectome and ManipulationInterdisciplinary Center for Brain InformationThe Brain Cognition and Brain Disease InstituteShenzhen Institute of Advanced TechnologyShenzhenGuangdong518055China
| | - Guo‐Qiang Bi
- CAS Key Laboratory of Brain Connectome and ManipulationInterdisciplinary Center for Brain InformationThe Brain Cognition and Brain Disease InstituteShenzhen Institute of Advanced TechnologyShenzhenGuangdong518055China
| | - Bong‐Kiun Kaang
- Department of Biological SciencesCollege of Natural SciencesSeoul National UniversitySeoul151–746South Korea
| | - Graham L. Collingridge
- Department of PhysiologyFaculty of MedicineUniversity of TorontoMedical Science Building, 1 King's College CircleTorontoOntarioM5S 1A8Canada
| | - Min Zhuo
- Center for Neuron and DiseaseFrontier Institute of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
- Department of PhysiologyFaculty of MedicineUniversity of TorontoMedical Science Building, 1 King's College CircleTorontoOntarioM5S 1A8Canada
- Department of NeurologyFirst Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510130China
- Department of PharmacologyQingdao University School of PharmacyQingdaoShandong266071China
| |
Collapse
|
13
|
Casillas Martinez A, Wicki-Stordeur LE, Ariano AV, Swayne LA. Dual role for pannexin 1 at synapses: regulating functional and morphological plasticity. J Physiol 2024. [PMID: 39264228 DOI: 10.1113/jp285228] [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: 04/03/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
Pannexin 1 (PANX1) is an ion and metabolite membrane channel and scaffold protein enriched in synaptic compartments of neurons in the central nervous system. In addition to a well-established link between PANX1 and synaptic plasticity, we recently identified a role for PANX1 in the regulation of dendritic spine stability. Notably, PANX1 and its interacting proteins are linked to neurological conditions involving dendritic spine loss. Understanding the dual role of PANX1 in synaptic function and morphology may help to shed light on these links. We explore potential mechanisms, including PANX1's interactions with postsynaptic receptors and cytoskeleton regulating proteins. Finally, we contextualize PANX1's dual role within neurological diseases involving dendritic spine and synapse dysfunction.
Collapse
Affiliation(s)
| | - Leigh E Wicki-Stordeur
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Annika V Ariano
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
14
|
Dalkara T, Kaya Z, Erdener ŞE. Unraveling the interplay of neuroinflammatory signaling between parenchymal and meningeal cells in migraine headache. J Headache Pain 2024; 25:124. [PMID: 39080518 PMCID: PMC11290240 DOI: 10.1186/s10194-024-01827-x] [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: 04/01/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The initiation of migraine headaches and the involvement of neuroinflammatory signaling between parenchymal and meningeal cells remain unclear. Experimental evidence suggests that a cascade of inflammatory signaling originating from neurons may extend to the meninges, thereby inducing neurogenic inflammation and headache. This review explores the role of parenchymal inflammatory signaling in migraine headaches, drawing upon recent advancements. BODY: Studies in rodents have demonstrated that sterile meningeal inflammation can stimulate and sensitize meningeal nociceptors, culminating in headaches. The efficacy of relatively blood-brain barrier-impermeable anti-calcitonin gene-related peptide antibodies and triptans in treating migraine attacks, both with and without aura, supports the concept of migraine pain originating in meninges. Additionally, PET studies utilizing inflammation markers have revealed meningeal inflammatory activity in patients experiencing migraine with aura, particularly over the occipital cortex generating visual auras. The parenchymal neuroinflammatory signaling involving neurons, astrocytes, and microglia, which eventually extends to the meninges, can link non-homeostatic perturbations in the insensate brain to pain-sensitive meninges. Recent experimental research has brought deeper insight into parenchymal signaling mechanisms: Neuronal pannexin-1 channels act as stress sensors, initiating the inflammatory signaling by inflammasome formation and high-mobility group box-1 release in response to transient perturbations such as cortical spreading depolarization (CSD) or synaptic metabolic insufficiency caused by transcriptional changes induced by migraine triggers like sleep deprivation and stress. After a single CSD, astrocytes respond by upregulating the transcription of proinflammatory enzymes and mediators, while microglia are involved in restoring neuronal structural integrity; however, repeated CSDs may prompt microglia to adopt a pro-inflammatory state. Transcriptional changes from pro- to anti-inflammatory within 24 h may serve to dampen the inflammatory signaling. The extensive coverage of brain surface and perivascular areas by astrocyte endfeet suggests their role as an interface for transporting inflammatory mediators to the cerebrospinal fluid to contribute to meningeal nociception. CONCLUSION We propose that neuronal stress induced by CSD or synaptic activity-energy mismatch may initiate a parenchymal inflammatory signaling cascade, transmitted to the meninges, thereby triggering lasting headaches characteristic of migraine, with or without aura. This neuroinflammatory interplay between parenchymal and meningeal cells points to the potential for novel targets for migraine treatment and prophylaxis.
Collapse
Affiliation(s)
- Turgay Dalkara
- Departments of Neuroscience and, Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey.
| | - Zeynep Kaya
- Department of Neurology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Şefik Evren Erdener
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
15
|
Obot P, Cibelli A, Pan J, Velíšek L, Velíšková J, Scemes E. Pannexin1 Mediates Early-Life Seizure-Induced Social Behavior Deficits. ASN Neuro 2024; 16:2371164. [PMID: 39024558 PMCID: PMC11262470 DOI: 10.1080/17590914.2024.2371164] [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: 01/11/2024] [Accepted: 04/11/2024] [Indexed: 07/20/2024] Open
Abstract
There is a high co-morbidity between childhood epilepsy and autism spectrum disorder (ASD), with age of seizure onset being a critical determinant of behavioral outcomes. The interplay between these comorbidities has been investigated in animal models with results showing that the induction of seizures at early post-natal ages leads to learning and memory deficits and to autistic-like behavior in adulthood. Modifications of the excitation/inhibition (glutamate/GABA, ATP/adenosine) balance that follows early-life seizures (ELS) are thought to be the physiological events that underlie neuropsychiatric and neurodevelopmental disorders. Although alterations in purinergic/adenosinergic signaling have been implicated in seizures and ASD, it is unknown whether the ATP release channels, Pannexin1 (Panx1), contribute to ELS-induced behavior changes. To tackle this question, we used the ELS-kainic acid model in transgenic mice with global and cell type specific deletion of Panx1 to evaluate whether these channels were involved in behavioral deficits that occur later in life. Our studies show that ELS results in Panx1 dependent social behavior deficits and also in poor performance in a spatial memory test that does not involve Panx1. These findings provide support for a link between ELS and adult behavioral deficits. Moreover, we identify neuronal and not astrocyte Panx1 as a potential target to specifically limit astrogliosis and social behavioral deficits resultant from early-life seizures.
Collapse
Affiliation(s)
- Price Obot
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| | - Antonio Cibelli
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jian Pan
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| | - Libor Velíšek
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
- Department of Neurology, New York Medical College, Valhalla, New York, USA
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Jana Velíšková
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
- Department of Neurology, New York Medical College, Valhalla, New York, USA
- Department of Obstetrics and Gynecology, New York Medical College, Valhalla, New York, USA
| | - Eliana Scemes
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
16
|
Henze E, Ehrlich JJ, Robertson JL, Kawate T. The C-terminal activating domain promotes Panx1 channel opening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598903. [PMID: 38915727 PMCID: PMC11195165 DOI: 10.1101/2024.06.13.598903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Pannexin 1 (Panx1) constitutes a large pore channel responsible for the release of ATP from apoptotic cells. Strong evidence indicates that caspase-mediated cleavage of the C-terminus promotes the opening of the Panx1 channel by unplugging the pore. However, this simple pore-plugging mechanism alone cannot account for the observation that a Panx1 construct ending before the caspase cleavage site remains closed. Here, we show that a helical region located immediately before the caspase cleavage site, referred to as the "C-terminal activating domain (CAD)," plays a pivotal role in facilitating Panx1 activation. Electrophysiology and mutagenesis studies uncovered that two conserved leucine residues within the CAD plays a pivotal role. Cryo-EM analysis of the construct ending before reaching the CAD demonstrated that the N-terminus extends into an intracellular pocket. In contrast, the construct including the CAD revealed that this domain occupies the intracellular pocket, causing the N-terminus to flip upward within the pore. Analysis of electrostatic free energy landscape in the closed conformation indicated that the intracellular side of the ion permeation pore may be occupied by anions like ATP, creating an electrostatic barrier for anions attempting to permeate the pore. When the N-terminus flips up, it diminishes the positively charged surface, thereby reducing the drive to accumulate anions inside the pore. This dynamic change in the electrostatic landscape likely contributes to the selection of permeant ions. Collectively, these experiments put forth a novel mechanism in which C-terminal cleavage liberates the CAD, causing the repositioning of the N-terminus to promote Panx1 channel opening.
Collapse
Affiliation(s)
- Erik Henze
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | - Janice L. Robertson
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
17
|
Tichauer JE, Lira M, Cerpa W, Orellana JA, Sáez JC, Rovegno M. Inhibition of astroglial hemichannels prevents synaptic transmission decline during spreading depression. Biol Res 2024; 57:39. [PMID: 38867288 PMCID: PMC11167948 DOI: 10.1186/s40659-024-00519-9] [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: 04/01/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Spreading depression (SD) is an intriguing phenomenon characterized by massive slow brain depolarizations that affect neurons and glial cells. This phenomenon is repetitive and produces a metabolic overload that increases secondary damage. However, the mechanisms associated with the initiation and propagation of SD are unknown. Multiple lines of evidence indicate that persistent and uncontrolled opening of hemichannels could participate in the pathogenesis and progression of several neurological disorders including acute brain injuries. Here, we explored the contribution of astroglial hemichannels composed of connexin-43 (Cx43) or pannexin-1 (Panx1) to SD evoked by high-K+ stimulation in brain slices. RESULTS Focal high-K+ stimulation rapidly evoked a wave of SD linked to increased activity of the Cx43 and Panx1 hemichannels in the brain cortex, as measured by light transmittance and dye uptake analysis, respectively. The activation of these channels occurs mainly in astrocytes but also in neurons. More importantly, the inhibition of both the Cx43 and Panx1 hemichannels completely prevented high K+-induced SD in the brain cortex. Electrophysiological recordings also revealed that Cx43 and Panx1 hemichannels critically contribute to the SD-induced decrease in synaptic transmission in the brain cortex and hippocampus. CONCLUSIONS Targeting Cx43 and Panx1 hemichannels could serve as a new therapeutic strategy to prevent the initiation and propagation of SD in several acute brain injuries.
Collapse
Affiliation(s)
- Juan E Tichauer
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Matías Lira
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Waldo Cerpa
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Juan C Sáez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.
| | - Maximiliano Rovegno
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
18
|
Vitureira N, Rafael A, Abudara V. P2X7 receptors and pannexin1 hemichannels shape presynaptic transmission. Purinergic Signal 2024; 20:223-236. [PMID: 37713157 PMCID: PMC11189373 DOI: 10.1007/s11302-023-09965-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023] Open
Abstract
Over the last decades, since the discovery of ATP as a transmitter, accumulating evidence has been reported about the role of this nucleotide and purinergic receptors, in particular P2X7 receptors, in the modulation of synaptic strength and plasticity. Purinergic signaling has emerged as a crucial player in orchestrating the molecular interaction between the components of the tripartite synapse, and much progress has been made in how this neuron-glia interaction impacts neuronal physiology under basal and pathological conditions. On the other hand, pannexin1 hemichannels, which are functionally linked to P2X7 receptors, have appeared more recently as important modulators of excitatory synaptic function and plasticity under diverse contexts. In this review, we will discuss the contribution of ATP, P2X7 receptors, and pannexin hemichannels to the modulation of presynaptic strength and its impact on motor function, sensory processing, synaptic plasticity, and neuroglial communication, with special focus on the P2X7 receptor/pannexin hemichannel interplay. We also address major hypotheses about the role of this interaction in physiological and pathological circumstances.
Collapse
Affiliation(s)
- Nathalia Vitureira
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - Alberto Rafael
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Verónica Abudara
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
19
|
Rangel-Sandoval C, Soula M, Li WP, Castillo PE, Hunt DL. NMDAR-mediated activation of pannexin1 channels contributes to the detonator properties of hippocampal mossy fiber synapses. iScience 2024; 27:109681. [PMID: 38680664 PMCID: PMC11046245 DOI: 10.1016/j.isci.2024.109681] [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: 09/28/2023] [Revised: 02/23/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024] Open
Abstract
Pannexins are large-pore ion channels expressed throughout the mammalian brain that participate in various neuropathologies; however, their physiological roles remain obscure. Here, we report that pannexin1 channels (Panx1) can be synaptically activated under physiological recording conditions in rodent acute hippocampal slices. Specifically, NMDA receptor (NMDAR)-mediated responses at the mossy fiber to CA3 pyramidal cell synapse were followed by a slow postsynaptic inward current that could activate CA3 pyramidal cells but was absent in Panx1 knockout mice. Immunoelectron microscopy revealed that Panx1 was localized near the postsynaptic density. Further, Panx1-mediated currents were potentiated by metabotropic receptors and bidirectionally modulated by burst-timing-dependent plasticity of NMDAR-mediated transmission. Lastly, Panx1 channels were preferentially recruited when NMDAR activation enters a supralinear regime, resulting in temporally delayed burst-firing. Thus, Panx1 can contribute to synaptic amplification and broadening the temporal associativity window for co-activated pyramidal cells, thereby supporting the auto-associative functions of the CA3 region.
Collapse
Affiliation(s)
- Cinthia Rangel-Sandoval
- Department of Neurosurgery, Department of Neurology, Department of Biomedical Sciences, Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marisol Soula
- Dominick P. Purpura Department of Neuroscience, Department of Psychiatry and Behavioral Sciences. Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wei-Ping Li
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Pablo E. Castillo
- Dominick P. Purpura Department of Neuroscience, Department of Psychiatry and Behavioral Sciences. Albert Einstein College of Medicine, Bronx, NY, USA
| | - David L. Hunt
- Department of Neurosurgery, Department of Neurology, Department of Biomedical Sciences, Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
20
|
Xing Q, Cibelli A, Yang GL, Dohare P, Li QH, Scemes E, Guan FX, Spray DC. Neuronal Panx1 drives peripheral sensitization in experimental plantar inflammatory pain. Mil Med Res 2024; 11:27. [PMID: 38685116 PMCID: PMC11057180 DOI: 10.1186/s40779-024-00525-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The channel-forming protein Pannexin1 (Panx1) has been implicated in both human studies and animal models of chronic pain, but the underlying mechanisms remain incompletely understood. METHODS Wild-type (WT, n = 24), global Panx1 KO (n = 24), neuron-specific Panx1 KO (n = 20), and glia-specific Panx1 KO (n = 20) mice were used in this study at Albert Einstein College of Medicine. The von Frey test was used to quantify pain sensitivity in these mice following complete Freund's adjuvant (CFA) injection (7, 14, and 21 d). The qRT-PCR was employed to measure mRNA levels of Panx1, Panx2, Panx3, Cx43, Calhm1, and β-catenin. Laser scanning confocal microscopy imaging, Sholl analysis, and electrophysiology were utilized to evaluate the impact of Panx1 on neuronal excitability and morphology in Neuro2a and dorsal root ganglion neurons (DRGNs) in which Panx1 expression or function was manipulated. Ethidium bromide (EtBr) dye uptake assay and calcium imaging were employed to investigate the role of Panx1 in adenosine triphosphate (ATP) sensitivity. β-galactosidase (β-gal) staining was applied to determine the relative cellular expression levels of Panx1 in trigeminal ganglia (TG) and DRG of transgenic mice. RESULTS Global or neuron-specific Panx1 deletion markedly decreased pain thresholds after CFA stimuli (7, 14, and 21 d; P < 0.01 vs. WT group), indicating that Panx1 was positively correlated with pain sensitivity. In Neuro2a, global Panx1 deletion dramatically reduced neurite extension and inward currents compared to the WT group (P < 0.05), revealing that Panx1 enhanced neurogenesis and excitability. Similarly, global Panx1 deletion significantly suppressed Wnt/β-catenin dependent DRG neurogenesis following 5 d of nerve growth factor (NGF) treatment (P < 0.01 vs. WT group). Moreover, Panx1 channels enhanced DRG neuron response to ATP after CFA injection (P < 0.01 vs. Panx1 KO group). Furthermore, ATP release increased Ca2+ responses in DRGNs and satellite glial cells surrounding them following 7 d of CFA treatment (P < 0.01 vs. Panx1 KO group), suggesting that Panx1 in glia also impacts exaggerated neuronal excitability. Interestingly, neuron-specific Panx1 deletion was found to markedly reduce differentiation in cultured DRGNs, as evidenced by stunted neurite outgrowth (P < 0.05 vs. Panx1 KO group; P < 0.01 vs. WT group or GFAP-Cre group), blunted activation of Wnt/β-catenin signaling (P < 0.01 vs. WT, Panx1 KO and GFAP-Cre groups), and diminished cell excitability (P < 0.01 vs. GFAP-Cre group) and response to ATP stimulation (P < 0.01 vs. WT group). Analysis of β-gal staining showed that cellular expression levels of Panx1 in neurons are significantly higher (2.5-fold increase) in the DRG than in the TG. CONCLUSIONS The present study revealed that neuronal Panx1 is a prominent driver of peripheral sensitivity in the setting of inflammatory pain through cell-autonomous effects on neuronal excitability. This hyperexcitability dependence on neuronal Panx1 contrasts with inflammatory orofacial pain, where similar studies revealed a prominent role for glial Panx1. The apparent differences in Panx1 expression in neuronal and non-neuronal TG and DRG cells are likely responsible for the distinct impact of these cell types in the two pain models.
Collapse
Affiliation(s)
- Qu Xing
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Antonio Cibelli
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, 70125, Italy
| | - Greta Luyuan Yang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, 06459, USA
| | - Preeti Dohare
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, 12208, USA
| | - Qing-Hua Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Eliana Scemes
- Department of Anatomy and Cell Biology, New York Medical College, Valhalla, NY, 10595, USA
| | - Fang-Xia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450001, China.
| | - David C Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
21
|
Seeholzer LF, Julius D. Neuroendocrine cells initiate protective upper airway reflexes. Science 2024; 384:295-301. [PMID: 38669574 PMCID: PMC11407116 DOI: 10.1126/science.adh5483] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 02/21/2024] [Indexed: 04/28/2024]
Abstract
Airway neuroendocrine (NE) cells have been proposed to serve as specialized sensory epithelial cells that modulate respiratory behavior by communicating with nearby nerve endings. However, their functional properties and physiological roles in the healthy lung, trachea, and larynx remain largely unknown. In this work, we show that murine NE cells in these compartments have distinct biophysical properties but share sensitivity to two commonly aspirated noxious stimuli, water and acid. Moreover, we found that tracheal and laryngeal NE cells protect the airways by releasing adenosine 5'-triphosphate (ATP) to activate purinoreceptive sensory neurons that initiate swallowing and expiratory reflexes. Our work uncovers the broad molecular and biophysical diversity of NE cells across the airways and reveals mechanisms by which these specialized excitable cells serve as sentinels for activating protective responses.
Collapse
Affiliation(s)
- Laura F. Seeholzer
- Department of Physiology, University of California, San Francisco; San Francisco, 94143, USA
| | - David Julius
- Department of Physiology, University of California, San Francisco; San Francisco, 94143, USA
| |
Collapse
|
22
|
McAllister BB, Stokes-Heck S, Harding EK, van den Hoogen NJ, Trang T. Targeting Pannexin-1 Channels: Addressing the 'Gap' in Chronic Pain. CNS Drugs 2024; 38:77-91. [PMID: 38353876 DOI: 10.1007/s40263-024-01061-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/07/2024] [Indexed: 02/22/2024]
Abstract
Chronic pain complicates many diseases and is notoriously difficult to treat. In search of new therapeutic targets, pannexin-1 (Panx1) channels have sparked intense interest as a key mechanism involved in a variety of chronic pain conditions. Panx1 channels are transmembrane proteins that release ions and small molecules, such as adenosine triphosphate (ATP). They are expressed along important nodes of the pain pathway, modulating activity of diverse cell types implicated in the development and progression of chronic pain caused by injury or pathology. This review highlights advances that have unlocked the core structure and machinery controlling Panx1 function with a focus on understanding and treating chronic pain.
Collapse
Affiliation(s)
- Brendan B McAllister
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Sierra Stokes-Heck
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Erika K Harding
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Nynke J van den Hoogen
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Tuan Trang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
23
|
Cibelli A, Dohare P, Spray DC, Scemes E. Differential activation of mouse and human Panx1 channel variants. PLoS One 2023; 18:e0295710. [PMID: 38100403 PMCID: PMC10723736 DOI: 10.1371/journal.pone.0295710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
Pannexins are ubiquitously expressed in human and mouse tissues. Pannexin 1 (Panx1), the most thoroughly characterized member of this family, forms plasmalemmal membrane channels permeable to relatively large molecules, such as ATP. Although human and mouse Panx1 amino acid sequences are conserved in the presently known regulatory sites involved in trafficking and modulation of the channel, differences are reported in the N- and C-termini of the protein, and the mechanisms of channel activation by different stimuli remain controversial. Here we used a neuroblastoma cell line to study the activation properties of endogenous mPanx1 and exogenously expressed hPanx1. Dye uptake and electrophysiological recordings revealed that in contrast to mouse Panx1, the human ortholog is insensitive to stimulation with high extracellular [K+] but responds similarly to activation of the purinergic P2X7 receptor. The two most frequent Panx1 polymorphisms found in the human population, Q5H (rs1138800) and E390D (rs74549886), exogenously expressed in Panx1-null N2a cells revealed that regarding P2X7 receptor mediated Panx1 activation, the Q5H mutant is a gain of function whereas the E390D mutant is a loss of function variant. Collectively, we demonstrate differences in the activation between human and mouse Panx1 orthologs and suggest that these differences may have translational implications for studies where Panx1 has been shown to have significant impact.
Collapse
Affiliation(s)
- Antonio Cibelli
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Preeti Dohare
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - David C. Spray
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Eliana Scemes
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States of America
| |
Collapse
|
24
|
O'Donnell BL, Penuela S. Skin in the game: pannexin channels in healthy and cancerous skin. Biochem J 2023; 480:1929-1949. [PMID: 38038973 DOI: 10.1042/bcj20230176] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
The skin is a highly organized tissue composed of multiple layers and cell types that require coordinated cell to cell communication to maintain tissue homeostasis. In skin cancer, this organized structure and communication is disrupted, prompting the malignant transformation of healthy cells into melanoma, basal cell carcinoma or squamous cell carcinoma tumours. One such family of channel proteins critical for cellular communication is pannexins (PANX1, PANX2, PANX3), all of which are present in the skin. These heptameric single-membrane channels act as conduits for small molecules and ions like ATP and Ca2+ but have also been shown to have channel-independent functions through their interacting partners or action in signalling pathways. Pannexins have diverse roles in the skin such as in skin development, aging, barrier function, keratinocyte differentiation, inflammation, and wound healing, which were discovered through work with pannexin knockout mice, organotypic epidermis models, primary cells, and immortalized cell lines. In the context of cutaneous cancer, PANX1 is present at high levels in melanoma tumours and functions in melanoma carcinogenesis, and both PANX1 and PANX3 expression is altered in non-melanoma skin cancer. PANX2 has thus far not been implicated in any skin cancer. This review will discuss pannexin isoforms, structure, trafficking, post-translational modifications, interactome, and channel activity. We will also outline the expression, localization, and function of pannexin channels within the diverse cell types of the epidermis, dermis, hypodermis, and adnexal structures of the skin, and how these properties are exploited or abrogated in instances of skin cancer.
Collapse
Affiliation(s)
- Brooke L O'Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
- Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| |
Collapse
|
25
|
Weilinger NL, Yang K, Choi HB, Groten CJ, Wendt S, Murugan M, Wicki-Stordeur LE, Bernier LP, Velayudhan PS, Zheng J, LeDue JM, Rungta RL, Tyson JR, Snutch TP, Wu LJ, MacVicar BA. Pannexin-1 opening in neuronal edema causes cell death but also leads to protection via increased microglia contacts. Cell Rep 2023; 42:113128. [PMID: 37742194 PMCID: PMC10824275 DOI: 10.1016/j.celrep.2023.113128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/26/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
Neuronal swelling during cytotoxic edema is triggered by Na+ and Cl- entry and is Ca2+ independent. However, the causes of neuronal death during swelling are unknown. Here, we investigate the role of large-conductance Pannexin-1 (Panx1) channels in neuronal death during cytotoxic edema. Panx1 channel inhibitors reduce and delay neuronal death in swelling triggered by voltage-gated Na+ entry with veratridine. Neuronal swelling causes downstream production of reactive oxygen species (ROS) that opens Panx1 channels. We confirm that ROS activates Panx1 currents with whole-cell electrophysiology and find scavenging ROS is neuroprotective. Panx1 opening and subsequent ATP release attract microglial processes to contact swelling neurons. Depleting microglia using the CSF1 receptor antagonist PLX3397 or blocking P2Y12 receptors exacerbates neuronal death, suggesting that the Panx1-ATP-dependent microglia contacts are neuroprotective. We conclude that cytotoxic edema triggers oxidative stress in neurons that opens Panx1 to trigger death but also initiates neuroprotective feedback mediated by microglia contacts.
Collapse
Affiliation(s)
- Nicholas L Weilinger
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Kai Yang
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hyun B Choi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Christopher J Groten
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Stefan Wendt
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | | - Leigh E Wicki-Stordeur
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Louis-Philippe Bernier
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Prashanth S Velayudhan
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jeffrey M LeDue
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ravi L Rungta
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Stomatology and Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - John R Tyson
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Terrance P Snutch
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Brian A MacVicar
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
26
|
Akula SK, Chen AY, Neil JE, Shao DD, Mo A, Hylton NK, DiTroia S, Ganesh VS, Smith RS, O’Kane K, Yeh RC, Marciano JH, Kirkham S, Kenny CJ, Song JHT, Al Saffar M, Millan F, Harris DJ, Murphy AV, Klemp KC, Braddock SR, Brand H, Wong I, Talkowski ME, O’Donnell-Luria A, Lai A, Hill RS, Mochida GH, Doan RN, Barkovich AJ, Yang E, Amrom D, Andermann E, Poduri A, Walsh CA. Exome Sequencing and the Identification of New Genes and Shared Mechanisms in Polymicrogyria. JAMA Neurol 2023; 80:980-988. [PMID: 37486637 PMCID: PMC10366952 DOI: 10.1001/jamaneurol.2023.2363] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/23/2023] [Indexed: 07/25/2023]
Abstract
Importance Polymicrogyria is the most commonly diagnosed cortical malformation and is associated with neurodevelopmental sequelae including epilepsy, motor abnormalities, and cognitive deficits. Polymicrogyria frequently co-occurs with other brain malformations or as part of syndromic diseases. Past studies of polymicrogyria have defined heterogeneous genetic and nongenetic causes but have explained only a small fraction of cases. Objective To survey germline genetic causes of polymicrogyria in a large cohort and to consider novel polymicrogyria gene associations. Design, Setting, and Participants This genetic association study analyzed panel sequencing and exome sequencing of accrued DNA samples from a retrospective cohort of families with members with polymicrogyria. Samples were accrued over more than 20 years (1994 to 2020), and sequencing occurred in 2 stages: panel sequencing (June 2015 to January 2016) and whole-exome sequencing (September 2019 to March 2020). Individuals seen at multiple clinical sites for neurological complaints found to have polymicrogyria on neuroimaging, then referred to the research team by evaluating clinicians, were included in the study. Targeted next-generation sequencing and/or exome sequencing were performed on probands (and available parents and siblings) from 284 families with individuals who had isolated polymicrogyria or polymicrogyria as part of a clinical syndrome and no genetic diagnosis at time of referral from clinic, with sequencing from 275 families passing quality control. Main Outcomes and Measures The number of families in whom genetic sequencing yielded a molecular diagnosis that explained the polymicrogyria in the family. Secondarily, the relative frequency of different genetic causes of polymicrogyria and whether specific genetic causes were associated with co-occurring head size changes were also analyzed. Results In 32.7% (90 of 275) of polymicrogyria-affected families, genetic variants were identified that provided satisfactory molecular explanations. Known genes most frequently implicated by polymicrogyria-associated variants in this cohort were PIK3R2, TUBB2B, COL4A1, and SCN3A. Six candidate novel polymicrogyria genes were identified or confirmed: de novo missense variants in PANX1, QRICH1, and SCN2A and compound heterozygous variants in TMEM161B, KIF26A, and MAN2C1, each with consistent genotype-phenotype relationships in multiple families. Conclusions and Relevance This study's findings reveal a higher than previously recognized rate of identifiable genetic causes, specifically of channelopathies, in individuals with polymicrogyria and support the utility of exome sequencing for families affected with polymicrogyria.
Collapse
Affiliation(s)
- Shyam K. Akula
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
- Harvard-MIT MD/PhD Program, Harvard Medical School, Boston, Massachusetts
| | - Allen Y. Chen
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Division of Rheumatology, Hospital for Special Surgery, New York, New York
| | - Jennifer E. Neil
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Diane D. Shao
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Alisa Mo
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Norma K. Hylton
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
- Harvard-MIT MD/PhD Program, Harvard Medical School, Boston, Massachusetts
| | - Stephanie DiTroia
- Program in Medical and Population Genetics, Center for Genomic Medicine, Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Vijay S. Ganesh
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Richard S. Smith
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Katherine O’Kane
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Rebecca C. Yeh
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Jack H. Marciano
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Samantha Kirkham
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Connor J. Kenny
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Janet H. T. Song
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Muna Al Saffar
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
- Department of Genetics and Genomics, United Arab Emirates University, United Arab Emirates
| | | | - David J. Harris
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Andrea V. Murphy
- Division of Medical Genetics, Our Lady of the Lake Health System, Baton Rouge, Louisiana
| | - Kara C. Klemp
- Division of Medical Genetics, Department of Pediatrics Saint Louis University School of Medicine, St Louis, Missouri
| | - Stephen R. Braddock
- Division of Medical Genetics, Department of Pediatrics Saint Louis University School of Medicine, St Louis, Missouri
| | - Harrison Brand
- Program in Medical and Population Genetics, Center for Genomic Medicine, Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
| | - Isaac Wong
- Program in Medical and Population Genetics, Center for Genomic Medicine, Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
| | - Michael E. Talkowski
- Program in Medical and Population Genetics, Center for Genomic Medicine, Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
| | - Anne O’Donnell-Luria
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Program in Medical and Population Genetics, Center for Genomic Medicine, Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Abbe Lai
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts
| | - Robert Sean Hill
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Ganeshwaran H. Mochida
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
| | - Ryan N. Doan
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
| | - A. James Barkovich
- Benioff Children’s Hospital, Departments of Radiology, Pediatrics, Neurology, and Neurological Surgery, University of California, San Francisco, San Francisco
| | - Edward Yang
- Department of Radiology, Boston Children’s Hospital, Boston, Massachusetts
| | - Dina Amrom
- Neurogenetics Unit, Montreal Neurological Hospital and Institute, Montreal, Quebec, Canada
- Department of Neurology & Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Neurology, Queen Fabiola Children’s University Hospital, Brussels, Belgium
- Pediatric Neurology Unit, Centre Hospitalier de Luxembourg, Grand-Duchy of Luxembourg
| | - Eva Andermann
- Department of Neurology & Neurosurgery, McGill University, Montreal, Quebec, Canada
- Pediatric Neurology Unit, Centre Hospitalier de Luxembourg, Grand-Duchy of Luxembourg
- Epilepsy Research Group, Montreal Neurological Hospital and Institute, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Annapurna Poduri
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts
| | - Christopher A. Walsh
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, and Allen Discovery Center for Human Brain Evolution, Boston, Massachusetts
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts
- Harvard-MIT MD/PhD Program, Harvard Medical School, Boston, Massachusetts
- Program in Medical and Population Genetics, Center for Genomic Medicine, Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
27
|
Wu YL, Yang AH, Chiu YH. Recent advances in the structure and activation mechanisms of metabolite-releasing Pannexin 1 channels. Biochem Soc Trans 2023; 51:1687-1699. [PMID: 37622532 DOI: 10.1042/bst20230038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023]
Abstract
Pannexin 1 (PANX1) is a widely expressed large-pore ion channel located in the plasma membrane of almost all vertebrate cells. It possesses a unique ability to act as a conduit for both inorganic ions (e.g. potassium or chloride) and bioactive metabolites (e.g. ATP or glutamate), thereby activating varying signaling pathways in an autocrine or paracrine manner. Given its crucial role in cell-cell interactions, the activity of PANX1 has been implicated in maintaining homeostasis of cardiovascular, immune, and nervous systems. Dysregulation of PANX1 has also been linked to numerous diseases, such as ischemic stroke, seizure, and inflammatory disorders. Therefore, the mechanisms underlying different modes of PANX1 activation and its context-specific channel properties have gathered significant attention. In this review, we summarize the roles of PANX1 in various physiological processes and diseases, and analyze the accumulated lines of evidence supporting diverse molecular mechanisms associated with different PANX1 activation modalities. We focus on examining recent discoveries regarding PANX1 regulations by reversible post-translational modifications, elevated intracellular calcium concentration, and protein-protein interactions, as well as by irreversible cleavage of its C-terminal tail. Additionally, we delve into the caveats in the proposed PANX1 gating mechanisms and channel open-closed configurations by critically analyzing the structural insights derived from cryo-EM studies and the unitary properties of PANX1 channels. By doing so, we aim to identify potential research directions for a better understanding of the functions and regulations of PANX1 channels.
Collapse
Affiliation(s)
- Yi-Ling Wu
- Department of Life Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
| | - Ai-Hsing Yang
- Department of Life Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
| | - Yu-Hsin Chiu
- Department of Life Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
| |
Collapse
|
28
|
Lissoni A, Tao S, Allewaert R, Witschas K, Leybaert L. Cx43 Hemichannel and Panx1 Channel Modulation by Gap19 and 10Panx1 Peptides. Int J Mol Sci 2023; 24:11612. [PMID: 37511370 PMCID: PMC10380488 DOI: 10.3390/ijms241411612] [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: 06/30/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
Cx43 hemichannels (HCs) and Panx1 channels are two genetically distant protein families. Despite the lack of sequence homology, Cx43 and Panx1 channels have been the subject of debate due to their overlapping expression and the fact that both channels present similarities in terms of their membrane topology and electrical properties. Using the mimetic peptides Gap19 and 10Panx1, this study aimed to investigate the cross-effects of these peptides on Cx43 HCs and Panx1 channels. The single-channel current activity from stably expressing HeLa-Cx43 and C6-Panx1 cells was recorded using patch-clamp experiments in whole-cell voltage-clamp mode, demonstrating 214 pS and 68 pS average unitary conductances for the respective channels. Gap19 was applied intracellularly while 10Panx1 was applied extracellularly at different concentrations (100, 200 and 500 μM) and the average nominal open probability (NPo) was determined for each testing condition. A concentration of 100 µM Gap19 more than halved the NPo of Cx43 HCs, while 200 µM 10Panx1 was necessary to obtain a half-maximal NPo reduction in the Panx1 channels. Gap19 started to significantly inhibit the Panx1 channels at 500 µM, reducing the NPo by 26% while reducing the NPo of the Cx43 HCs by 84%. In contrast 10Panx1 significantly reduced the NPo of the Cx43 HCs by 37% at 100 µM and by 83% at 200 µM, a concentration that caused the half-maximal inhibition of the Panx1 channels. These results demonstrate that 10Panx1 inhibits Cx43 HCs over the 100-500 µM concentration range while 500 µM intracellular Gap19 is necessary to observe some inhibition of Panx1 channels.
Collapse
|
29
|
García-Rojas F, Flores-Muñoz C, Santander O, Solis P, Martínez AD, Ardiles ÁO, Fuenzalida M. Pannexin-1 Modulates Inhibitory Transmission and Hippocampal Synaptic Plasticity. Biomolecules 2023; 13:887. [PMID: 37371467 DOI: 10.3390/biom13060887] [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: 05/04/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Pannexin-1 (Panx1) hemichannel is a non-selective transmembrane channel that may play important roles in intercellular signaling by allowing the permeation of ions and metabolites, such as ATP. Although recent evidence shows that the Panx1 hemichannel is involved in controlling excitatory synaptic transmission, the role of Panx1 in inhibitory transmission remains unknown. Here, we studied the contribution of Panx1 to the GABAergic synaptic efficacy onto CA1 pyramidal neurons (PyNs) by using patch-clamp recordings and pharmacological approaches in wild-type and Panx1 knock-out (Panx1-KO) mice. We reported that blockage of the Panx1 hemichannel with the mimetic peptide 10Panx1 increases the synaptic level of endocannabinoids (eCB) and the activation of cannabinoid receptors type 1 (CB1Rs), which results in a decrease in hippocampal GABAergic efficacy, shifting excitation/inhibition (E/I) balance toward excitation and facilitating the induction of long-term potentiation. Our finding provides important insight unveiling that Panx1 can strongly influence the overall neuronal excitability and play a key role in shaping synaptic changes affecting the amplitude and direction of plasticity, as well as learning and memory processes.
Collapse
Affiliation(s)
- Francisca García-Rojas
- Centro de Neurobiología y Fisiopatología Integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Carolina Flores-Muñoz
- Centro de Neurobiología y Fisiopatología Integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Odra Santander
- Centro de Neurobiología y Fisiopatología Integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Pamela Solis
- Centro de Neurobiología y Fisiopatología Integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Programa de Magister en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Agustín D Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Álvaro O Ardiles
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro Interdisciplinario de Estudios en Salud, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2572007, Chile
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| |
Collapse
|
30
|
García-Rodríguez C, Mujica P, Illanes-González J, López A, Vargas C, Sáez JC, González-Jamett A, Ardiles ÁO. Probenecid, an Old Drug with Potential New Uses for Central Nervous System Disorders and Neuroinflammation. Biomedicines 2023; 11:1516. [PMID: 37371611 DOI: 10.3390/biomedicines11061516] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
Probenecid is an old uricosuric agent used in clinics to treat gout and reduce the renal excretion of antibiotics. In recent years, probenecid has gained attention due to its ability to interact with membrane proteins such as TRPV2 channels, organic anion transporters, and pannexin 1 hemichannels, which suggests new potential therapeutic utilities in medicine. Some current functions of probenecid include their use as an adjuvant to increase the bioavailability of several drugs in the Central Nervous System (CNS). Numerous studies also suggest that this drug has important neuroprotective, antiepileptic, and anti-inflammatory properties, as evidenced by their effect against neurological and neurodegenerative diseases. In these studies, the use of probenecid as a Panx1 hemichannel blocker to reduce neuroinflammation is highlighted since neuroinflammation is a major trigger for diverse CNS alterations. Although the clinical use of probenecid has declined over the years, advances in its use in preclinical research indicate that it may be useful to improve conventional therapies in the psychiatric field where the drugs used have a low bioavailability, either because of a deficient passage through the blood-brain barrier or a high efflux from the CNS or also a high urinary clearance. This review summarizes the history, pharmacological properties, and recent research uses of probenecid and discusses its future projections as a potential pharmacological strategy to intervene in neurodegeneration as an outcome of neuroinflammation.
Collapse
Affiliation(s)
- Claudia García-Rodríguez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Paula Mujica
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Javiera Illanes-González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Araceli López
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Camilo Vargas
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Juan C Sáez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Arlek González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Álvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| |
Collapse
|
31
|
Koval M, Schug WJ, Isakson BE. Pharmacology of pannexin channels. Curr Opin Pharmacol 2023; 69:102359. [PMID: 36858833 PMCID: PMC10023479 DOI: 10.1016/j.coph.2023.102359] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/18/2023] [Accepted: 01/29/2023] [Indexed: 03/02/2023]
Abstract
Pannexin channels play fundamental roles in regulating inflammation and have been implicated in many diseases including hypertension, stroke, and neuropathic pain. Thus, the ability to pharmacologically block these channels is a vital component of several therapeutic approaches. Pharmacologic interrogation of model systems also provides a means to discover new roles for pannexins in cell physiology. Here, we review the state of the art for agents that can be used to block pannexin channels, with a focus on chemical pharmaceuticals and peptide mimetics that act on pannexin 1. Guidance on interpreting results obtained with pannexin pharmacologics in experimental systems is discussed, as well as strengths and caveats of different agents, including specificity and feasibility of clinical application.
Collapse
Affiliation(s)
- Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Wyatt J Schug
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
32
|
Parrish RR, MacKenzie-Gray Scott C, Jackson-Taylor T, Grundmann A, McLeod F, Codadu NK, Călin A, Alfonsa H, Wykes RC, Voipio J, Trevelyan AJ. Indirect Effects of Halorhodopsin Activation: Potassium Redistribution, Nonspecific Inhibition, and Spreading Depolarization. J Neurosci 2023; 43:685-692. [PMID: 36639898 PMCID: PMC9899079 DOI: 10.1523/jneurosci.1141-22.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
The movement of ions in and out of neurons can exert significant effects on neighboring cells. Here we report several experimentally important consequences of activation of the optogenetic chloride pump, halorhodopsin. We recorded extracellular K+ concentration ([K+]extra) in neocortical brain slices prepared from young adult mice (both sexes) which express halorhodopsin in pyramidal cells. Strong halorhodopsin activation induced a pronounced drop in [K+]extra that persisted for the duration of illumination. Pharmacological blockade of K+ channels reduced the amplitude of this drop, indicating that it represents K+ redistribution into cells during the period of hyperpolarization. Halorhodopsin thus drives the inward movement of both Cl- directly, and K+ secondarily. When the illumination period ended, a rebound surge in extracellular [K+] developed over tens of seconds, partly reflecting the previous inward redistribution of K+, but additionally driven by clearance of Cl- coupled to K+ by the potassium-chloride cotransporter, KCC2. The drop in [K+]extra during light activation leads to a small (2-3 mV) hyperpolarization also of other cells that do not express halorhodopsin. Its activation therefore has both direct and indirect inhibitory effects. Finally, we show that persistent strong activation of halorhodopsin causes cortical spreading depolarizations (CSDs), both in vitro and in vivo This novel means of triggering CSDs is unusual, in that the events can arise during the actual period of illumination, when neurons are being hyperpolarized and [K+]extra is low. We suggest that this fundamentally different experimental model of CSDs will open up new avenues of research to explain how they occur naturally.SIGNIFICANCE STATEMENT Halorhodopsin is a light-activated electrogenic chloride pump, which has been widely used to inhibit neurons optogenetically. Here, we demonstrate three previously unrecognized consequences of its use: (1) intense activation leads to secondary movement of K+ ions into the cells; (2) the resultant drop in extracellular [K+] reduces excitability also in other, nonexpressing cells; and (3) intense persistent halorhodopsin activation can trigger cortical spreading depolarization (CSD). Halorhodopsin-induced CSDs can occur when neurons are hyperpolarized and extracellular [K+] is low. This contrasts with the most widely used experimental models that trigger CSDs with high [K+]. Both models, however, are consistent with the hypothesis that CSDs arise following net inward ionic movement into the principal neuron population.
Collapse
Affiliation(s)
- R Ryley Parrish
- Newcastle University Biosciences Institute, Medical School, Newcastle upon Tyne, NE2 4HH, United Kingdom
- Department of Cell Biology and Physiology, Brigham Young University, Provo 84602, Utah
| | | | - Tom Jackson-Taylor
- Newcastle University Biosciences Institute, Medical School, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Alex Grundmann
- Newcastle University Biosciences Institute, Medical School, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Faye McLeod
- Newcastle University Biosciences Institute, Medical School, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Neela K Codadu
- Queen Square Institute of Neurology, University College London, WC1N 3BG, United Kingdom
| | - Alexandru Călin
- Newcastle University Biosciences Institute, Medical School, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Hannah Alfonsa
- Newcastle University Biosciences Institute, Medical School, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Rob C Wykes
- Nanomedicine Lab, University of Manchester, Manchester, M13 9PL, United Kingdom
- Queen Square Institute of Neurology, University College London, WC1N 3BG, United Kingdom
| | - Juha Voipio
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences, University of Helsinki, Helsinki, 00014, Finland
| | - Andrew J Trevelyan
- Newcastle University Biosciences Institute, Medical School, Newcastle upon Tyne, NE2 4HH, United Kingdom
| |
Collapse
|
33
|
Vasile F, Dossi E, Moulard J, Ezan P, Lecoin L, Cohen-Salmon M, Mailly P, Le Bert M, Couillin I, Bemelmans A, Rouach N. Pannexin 1 activity in astroglia sets hippocampal neuronal network patterns. PLoS Biol 2022; 20:e3001891. [PMID: 36477165 PMCID: PMC9728857 DOI: 10.1371/journal.pbio.3001891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/24/2022] [Indexed: 12/12/2022] Open
Abstract
Astroglial release of molecules is thought to actively modulate neuronal activity, but the nature, release pathway, and cellular targets of these neuroactive molecules are still unclear. Pannexin 1, expressed by neurons and astrocytes, form nonselective large pore channels that mediate extracellular exchange of molecules. The functional relevance of these channels has been mostly studied in brain tissues, without considering their specific role in different cell types, or in neurons. Thus, our knowledge of astroglial pannexin 1 regulation and its control of neuronal activity remains very limited, largely due to the lack of tools targeting these channels in a cell-specific way. We here show that astroglial pannexin 1 expression in mice is developmentally regulated and that its activation is activity-dependent. Using astrocyte-specific molecular tools, we found that astroglial-specific pannexin 1 channel activation, in contrast to pannexin 1 activation in all cell types, selectively and negatively regulates hippocampal networks, with their disruption inducing a drastic switch from bursts to paroxysmal activity. This decrease in neuronal excitability occurs via an unconventional astroglial mechanism whereby pannexin 1 channel activity drives purinergic signaling-mediated regulation of hyperpolarisation-activated cyclic nucleotide (HCN)-gated channels. Our findings suggest that astroglial pannexin 1 channel activation serves as a negative feedback mechanism crucial for the inhibition of hippocampal neuronal networks.
Collapse
Affiliation(s)
- Flora Vasile
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, PSL Research University, Paris, France
| | - Elena Dossi
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, PSL Research University, Paris, France
| | - Julien Moulard
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, PSL Research University, Paris, France
| | - Pascal Ezan
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, PSL Research University, Paris, France
| | - Laure Lecoin
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, PSL Research University, Paris, France
| | - Martine Cohen-Salmon
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, PSL Research University, Paris, France
| | - Philippe Mailly
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, PSL Research University, Paris, France
| | - Marc Le Bert
- CNRS UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Isabelle Couillin
- CNRS UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Alexis Bemelmans
- Université Paris-Saclay, Commissariat à l’Energie Atomique et aux Energies Alternatives, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, PSL Research University, Paris, France
- * E-mail:
| |
Collapse
|
34
|
Garré JM, Bukauskas FF, Bennett MVL. Single channel properties of pannexin-1 and connexin-43 hemichannels and P2X7 receptors in astrocytes cultured from rodent spinal cords. Glia 2022; 70:2260-2275. [PMID: 35915989 PMCID: PMC9560969 DOI: 10.1002/glia.24250] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/11/2022]
Abstract
Astrocytes express surface channels involved in purinergic signaling. Among these channels, pannexin-1 (Px1) and connexin-43 (Cx43) hemichannels (HCs) release ATP that acts directly, or through its derivatives, on neurons and glia via purinergic receptors. Although HCs are functional, that is, open and close under physiological and pathological conditions, single channel properties of Px1 HCs in astrocytes have not been defined. Here, we developed a dual voltage clamp technique in HeLa cells expressing human Px1-YFP, and then applied this system to rodent spinal astrocytes to compare their single channel properties with other surface channels, that is, Cx43 HCs and P2X7 receptors (P2X7Rs). Channels were recorded in cell attached patches and evoked with ramp cycles applied through another pipette in whole cell voltage clamp. The mean unitary conductances of Px1 HCs were comparable in HeLa Px1-YFP cells and spinal astrocytes, ~42 and ~48 pS, respectively. Based on their unitary conductance, voltage-dependence, and unitary activity after pharmacological and gene silencing, Px1 HCs in astrocytes could be distinguished from Cx43 HCs and P2X7Rs. Channel activity of Px1 HCs and P2X7Rs was greater than that of Cx43 HCs in control astrocytes during ramps. Unitary activity of Px1 HCs was decreased and that of Cx43 HCs and P2X7Rs increased in astrocytes treated with fibroblast growth factor 1 (FGF-1). In summary, we resolved single channel properties of three different surface channels involved in purinergic signaling in spinal astrocytes, which were differentially modulated by FGF-1, a growth factor involved in neurodevelopment, inflammation and repair.
Collapse
Affiliation(s)
- Juan Mauricio Garré
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Feliksas F Bukauskas
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Michael V L Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
35
|
Flores-Muñoz C, García-Rojas F, Pérez MA, Santander O, Mery E, Ordenes S, Illanes-González J, López-Espíndola D, González-Jamett AM, Fuenzalida M, Martínez AD, Ardiles ÁO. The Long-Term Pannexin 1 Ablation Produces Structural and Functional Modifications in Hippocampal Neurons. Cells 2022; 11:cells11223646. [PMID: 36429074 PMCID: PMC9688914 DOI: 10.3390/cells11223646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/29/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022] Open
Abstract
Enhanced activity and overexpression of Pannexin 1 (Panx1) channels contribute to neuronal pathologies such as epilepsy and Alzheimer's disease (AD). The Panx1 channel ablation alters the hippocampus's glutamatergic neurotransmission, synaptic plasticity, and memory flexibility. Nevertheless, Panx1-knockout (Panx1-KO) mice still retain the ability to learn, suggesting that compensatory mechanisms stabilize their neuronal activity. Here, we show that the absence of Panx1 in the adult brain promotes a series of structural and functional modifications in the Panx1-KO hippocampal synapses, preserving spontaneous activity. Compared to the wild-type (WT) condition, the adult hippocampal neurons of Panx1-KO mice exhibit enhanced excitability, a more complex dendritic branching, enhanced spine maturation, and an increased proportion of multiple synaptic contacts. These modifications seem to rely on the actin-cytoskeleton dynamics as an increase in the actin polymerization and an imbalance between the Rac1 and the RhoA GTPase activities were observed in Panx1-KO brain tissues. Our findings highlight a novel interaction between Panx1 channels, actin, and Rho GTPases, which appear to be relevant for synapse stability.
Collapse
Affiliation(s)
- Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Francisca García-Rojas
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Miguel A. Pérez
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Escuela de Ciencias de la Salud, Universidad de Viña del Mar, Viña del Mar 2572007, Chile
| | - Odra Santander
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Elena Mery
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Stefany Ordenes
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Javiera Illanes-González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Daniela López-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2529002, Chile
- Centro de Investigaciones Biomédicas, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2529002, Chile
| | - Arlek M. González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Correspondence: (M.F.); (A.D.M.); (Á.O.A.)
| | - Agustín D. Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Correspondence: (M.F.); (A.D.M.); (Á.O.A.)
| | - Álvaro O. Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro Interdisciplinario de estudios en salud, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2572007, Chile
- Correspondence: (M.F.); (A.D.M.); (Á.O.A.)
| |
Collapse
|
36
|
KCC2 drives chloride microdomain formation in dendritic blebbing. Cell Rep 2022; 41:111556. [DOI: 10.1016/j.celrep.2022.111556] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/23/2022] [Accepted: 09/30/2022] [Indexed: 11/22/2022] Open
|
37
|
Abstract
Pannexin-1 (Panx1) channels contribute to neurological disorders, including stroke and epilepsy, where their function has been linked to N-methyl D-aspartate (NMDA) receptors (NMDARs). We discovered that Ca2+ entry via NMDARs recruits endoplasmic reticulum–resident STIM proteins to activate Panx1 by binding to a hydrophobic region localized to the Panx1 N terminus. Using loss-of-function approaches, combined with molecular replacement and use of a STIM/Panx1 function–blocking antibody, we demonstrate that disrupting the STIM/Panx1 interaction prevents Panx1 activation by NMDARs, but not by hypotonic stimuli. Thus, our findings serve as a basis for the design of modality-specific inhibitors against STIM-dependent Panx1 activation that will aid in understanding the multimodal functions of Panx1 and their contribution to physiology and pathology. Pannexin-1 (Panx1) is a large-pore ion and solute permeable channel highly expressed in the nervous system, where it subserves diverse processes, including neurite outgrowth, dendritic spine formation, and N-methyl D-aspartate (NMDA) receptor (NMDAR)-dependent plasticity. Moreover, Panx1 dysregulation contributes to neurological disorders, including neuropathic pain, epilepsy, and excitotoxicity. Despite progress in understanding physiological and pathological functions of Panx1, the mechanisms that regulate its activity, including its ion and solute permeability, remain poorly understood. In this study, we identify endoplasmic reticulum (ER)-resident stromal interaction molecules (STIM1/2), which are Ca2+ sensors that communicate events within the ER to plasma membrane channels, as binding and signaling partners of Panx1. We demonstrate that Panx1 is activated to its large-pore configuration in response to stimuli that recruit STIM1/2 and map the interaction interface to a hydrophobic region within the N terminus of Panx1. We further characterize a Panx1 N terminus–recognizing antibody as a function-blocking tool able to prevent large-pore Panx1 activation by STIM1/2. Using either the function-blocking antibody or re-expression of Panx1 deletion mutants in Panx1 knockout (KO) neurons, we show that STIM recruitment couples Ca2+ entry via NMDARs to Panx1 activation, thereby identifying a model of NMDAR-STIM-Panx1 signaling in neurons. Our study highlights a previously unrecognized and important role of the Panx1 N terminus in regulating channel activation and membrane localization. Considering past work demonstrating an intimate functional relation between NMDARs and Panx1, our study opens avenues for understanding activation modality and context-specific functions of Panx1, including functions linked to diverse STIM-regulated cellular responses.
Collapse
|
38
|
McDouall A, Zhou KQ, Bennet L, Green CR, Gunn AJ, Davidson JO. Connexins, Pannexins and Gap Junctions in Perinatal Brain Injury. Biomedicines 2022; 10:1445. [PMID: 35740466 PMCID: PMC9220888 DOI: 10.3390/biomedicines10061445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022] Open
Abstract
Perinatal brain injury secondary to hypoxia-ischemia and/or infection/inflammation remains a major cause of disability. Therapeutic hypothermia significantly improves outcomes, but in randomized controlled trials nearly half of infants still died or survived with disability, showing that additional interventions are needed. There is growing evidence that brain injury spreads over time from injured to previously uninjured regions of the brain. At least in part, this spread is related to opening of connexin hemichannels and pannexin channels, both of which are large conductance membrane channels found in many brain cells. Opening of these membrane channels releases adenosine triphosphate (ATP), and other neuroactive molecules, into the extracellular space. ATP has an important role in normal signaling, but pathologically can trigger the assembly of the multi-protein inflammasome complex. The inflammasome complex promotes activation of inflammatory caspases, and release of inflammatory cytokines. Overall, the connexin hemichannel appears to play a primary role in propagation of injury and chronic disease, and connexin hemichannel blockade has been shown to be neuroprotective in multiple animal models. Thus, there is potential for some blockers of connexin or pannexin channels to be developed into targeted interventions that could be used in conjunction with or separate to therapeutic hypothermia.
Collapse
Affiliation(s)
- Alice McDouall
- U1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand; (A.M.); (K.Q.Z.); (L.B.); (A.J.G.)
| | - Kelly Q. Zhou
- U1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand; (A.M.); (K.Q.Z.); (L.B.); (A.J.G.)
| | - Laura Bennet
- U1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand; (A.M.); (K.Q.Z.); (L.B.); (A.J.G.)
| | - Colin R. Green
- Department of Ophthalmology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand;
| | - Alistair J. Gunn
- U1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand; (A.M.); (K.Q.Z.); (L.B.); (A.J.G.)
| | - Joanne O. Davidson
- U1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand; (A.M.); (K.Q.Z.); (L.B.); (A.J.G.)
| |
Collapse
|
39
|
NMDA and P2X7 Receptors Require Pannexin 1 Activation to Initiate and Maintain Nociceptive Signaling in the Spinal Cord of Neuropathic Rats. Int J Mol Sci 2022; 23:ijms23126705. [PMID: 35743148 PMCID: PMC9223805 DOI: 10.3390/ijms23126705] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 02/01/2023] Open
Abstract
Pannexin 1 (Panx1) is involved in the spinal central sensitization process in rats with neuropathic pain, but its interaction with well-known, pain-related, ligand-dependent receptors, such as NMDA receptors (NMDAR) and P2X7 purinoceptors (P2X7R), remains largely unexplored. Here, we studied whether NMDAR- and P2X7R-dependent nociceptive signaling in neuropathic rats require the activation of Panx1 channels to generate spinal central sensitization, as assessed by behavioral (mechanical hyperalgesia) and electrophysiological (C-reflex wind-up potentiation) indexes. Administration of either a selective NMDAR agonist i.t. (NMDA, 2 mM) or a P2X7R agonist (BzATP, 150 μM) significantly increased both the mechanical hyperalgesia and the C-reflex wind-up potentiation, effects that were rapidly reversed (minutes) by i.t. administration of a selective pannexin 1 antagonist (10panx peptide, 300 μM), with the scores even reaching values of rats without neuropathy. Accordingly, 300 μM 10panx completely prevented the effects of NMDA and BzATP administered 1 h later, on mechanical hyperalgesia and C-reflex wind-up potentiation. Confocal immunofluorescence imaging revealed coexpression of Panx1 with NeuN protein in intrinsic dorsal horn neurons of neuropathic rats. The results indicate that both NMDAR- and P2X7R-mediated increases in mechanical hyperalgesia and C-reflex wind-up potentiation require neuronal Panx1 channel activation to initiate and maintain nociceptive signaling in neuropathic rats.
Collapse
|
40
|
Omolaoye TS, Jalaleddine N, Cardona Maya WD, du Plessis SS. Mechanisms of SARS-CoV-2 and Male Infertility: Could Connexin and Pannexin Play a Role? Front Physiol 2022; 13:866675. [PMID: 35721552 PMCID: PMC9205395 DOI: 10.3389/fphys.2022.866675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
The impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on male infertility has lately received significant attention. SARS-CoV-2, the virus that causes coronavirus disease (COVID-19) in humans, has been shown to impose adverse effects on both the structural components and function of the testis, which potentially impact spermatogenesis. These adverse effects are partially explained by fever, systemic inflammation, oxidative stress, and an increased immune response leading to impaired blood-testis barrier. It has been well established that efficient cellular communication via gap junctions or functional channels is required for tissue homeostasis. Connexins and pannexins are two protein families that mediate autocrine and paracrine signaling between the cells and the extracellular environment. These channel-forming proteins have been shown to play a role in coordinating cellular communication in the testis and epididymis. Despite their role in maintaining a proper male reproductive milieu, their function is disrupted under pathological conditions. The involvement of these channels has been well documented in several physiological and pathological conditions and their designated function in infectious diseases. However, their role in COVID-19 and their meaningful contribution to male infertility remains to be elucidated. Therefore, this review highlights the multivariate pathophysiological mechanisms of SARS-CoV-2 involvement in male reproduction. It also aims to shed light on the role of connexin and pannexin channels in disease progression, emphasizing their unexplored role and regulation of SARS-CoV-2 pathophysiology. Finally, we hypothesize the possible involvement of connexins and pannexins in SARS-CoV-2 inducing male infertility to assist future research ideas targeting therapeutic approaches.
Collapse
Affiliation(s)
- Temidayo S. Omolaoye
- Department of Basic Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Nour Jalaleddine
- Department of Basic Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Walter D. Cardona Maya
- Reproduction Group, Department of Microbiology and Parasitology, Faculty of Medicine, Universidad de Antioquia, Medellin, Colombia
| | - Stefan S. du Plessis
- Department of Basic Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
- *Correspondence: Stefan S. du Plessis,
| |
Collapse
|
41
|
Whyte-Fagundes P, Taskina D, Safarian N, Zoidl C, Carlen PL, Donaldson LW, Zoidl GR. Panx1 channels promote both anti- and pro-seizure-like activities in the zebrafish via p2rx7 receptors and ATP signaling. Commun Biol 2022; 5:472. [PMID: 35585187 PMCID: PMC9117279 DOI: 10.1038/s42003-022-03356-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/12/2022] [Indexed: 11/08/2022] Open
Abstract
The molecular mechanisms of excitation/inhibition imbalances promoting seizure generation in epilepsy patients are not fully understood. Evidence suggests that Pannexin1 (Panx1), an ATP release channel, modulates the excitability of the brain. In this report, we performed electrophysiological, behavioral, and molecular phenotyping experiments on zebrafish larvae bearing genetic or pharmacological knockouts of Panx1a and Panx1b channels, each homologous to human PANX1. When Panx1a function is lost, or both channels are under pharmacological blockade, seizures with ictal-like events and seizure-like locomotion are reduced in the presence of pentylenetetrazol. Transcriptome profiling by RNA-seq demonstrates a spectrum of distinct metabolic and cell signaling states which correlate with the loss of Panx1a. Furthermore, the pro- and anticonvulsant activities of both Panx1 channels affect ATP release and involve the purinergic receptor P2rx7. Our findings suggest a subfunctionalization of Panx1 enabling dual roles in seizures, providing a unique and comprehensive perspective to understanding seizure mechanisms in the context of this channel.
Collapse
Affiliation(s)
- Paige Whyte-Fagundes
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada.
- Center of Vision Research (CVR), York University, Toronto, ON, M3J1P3, Canada.
- Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 1M8, Canada.
| | - Daria Taskina
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada
- Center of Vision Research (CVR), York University, Toronto, ON, M3J1P3, Canada
| | - Nickie Safarian
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada
- Center of Vision Research (CVR), York University, Toronto, ON, M3J1P3, Canada
| | - Christiane Zoidl
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada
- Center of Vision Research (CVR), York University, Toronto, ON, M3J1P3, Canada
| | - Peter L Carlen
- Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 1M8, Canada
- Department of Medicine, Physiology and BME, University of Toronto, 399 Bathurst St., 5w442, Toronto, ON, M5T 2S8, Canada
| | | | - Georg R Zoidl
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada.
- Center of Vision Research (CVR), York University, Toronto, ON, M3J1P3, Canada.
- Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 1M8, Canada.
| |
Collapse
|
42
|
Endogenous pannexin1 channels form functional intercellular cell-cell channels with characteristic voltage-dependent properties. Proc Natl Acad Sci U S A 2022; 119:e2202104119. [PMID: 35486697 PMCID: PMC9171361 DOI: 10.1073/pnas.2202104119] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pannexin1 is a glycoprotein that has been shown to form functional plasma membrane channels and mediate many cellular signaling pathways. However, the formation and function of pannexin1-based intercellular cell–cell channels in mammalian cells and vertebrate tissue is a question of substantial debate. This work provides robust electrophysiological evidence to demonstrate that endogenously expressed human pannexin1 forms cell–cell channels and lays the groundwork for studying a potential new type of electrical synapses between many mammalian cell types that endogenously express pannexin1. The occurrence of intercellular channels formed by pannexin1 has been challenged for more than a decade. Here, we provide an electrophysiological characterization of exogenous human pannexin1 (hPanx1) cell–cell channels expressed in HeLa cells knocked out for connexin45. The observed hPanx1 cell–cell channels show two phenotypes: O-state and S-state. The former displayed low transjunctional voltage (Vj) sensitivity and single-channel conductance of ∼175 pS, with a substate of ∼35 pS; the latter showed a peculiar dynamic asymmetry in Vj dependence and single-channel conductance identical to the substate conductance of the O-state. S-state hPanx1 cell–cell channels were also identified between TC620 cells, a human oligodendroglioma cell line that endogenously expresses hPanx1. In these cells, dye and electrical coupling increased with temperature and were strongly reduced after hPanx1 expression was knocked down by small interfering RNA or inhibited with Panx1 mimetic inhibitory peptide. Moreover, cell–cell coupling was augmented when hPanx1 levels were increased with a doxycycline-inducible expression system. Application of octanol, a connexin gap junction (GJ) channel inhibitor, was not sufficient to block electrical coupling between HeLa KO Cx45-hPanx1 or TC620 cell pairs. In silico studies suggest that several arginine residues inside the channel pore may be neutralized by hydrophobic interactions, allowing the passage of DAPI, consistent with dye coupling observed between TC620 cells. These findings demonstrate that endogenously expressed hPanx1 forms intercellular cell–cell channels and their unique properties resemble those described in innexin-based GJ channels. Since Panx1 is ubiquitously expressed, finding conditions to recognize Panx1 cell–cell channels in different cell types might require special attention.
Collapse
|
43
|
Mechanisms of Pannexin 1 (PANX1) Channel Mechanosensitivity and Its Pathological Roles. Int J Mol Sci 2022; 23:ijms23031523. [PMID: 35163442 PMCID: PMC8836264 DOI: 10.3390/ijms23031523] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
Pannexins (PANX) were cloned based on their sequence homology to innexins (Inx), invertebrate gap junction proteins. Although there is no sequence homology between PANX and connexins (Cx), these proteins exhibit similar configurations. The PANX family has three members, PANX1, PANX2 and PANX3. Among them, PANX1 has been the most extensively studied. The PANX1 channels are activated by many factors, including high extracellular K+ ([K+]e), high intracellular Ca2+ ([Ca2+]i), Src family kinase (SFK)-mediated phosphorylation, caspase cleavage and mechanical stimuli. However, the mechanisms mediating this mechanosensitivity of PANX1 remain unknown. Both force-from-lipids and force-from-filaments models are proposed to explain the gating mechanisms of PANX1 channel mechanosensitivity. Finally, both the physiological and pathological roles of mechanosensitive PANX1 are discussed.
Collapse
|
44
|
Jaque-Fernández F, Jorquera G, Troc-Gajardo J, Pietri-Rouxel F, Gentil C, Buvinic S, Allard B, Jaimovich E, Jacquemond V, Casas M. Pannexin-1 and CaV1.1 show reciprocal interaction during excitation-contraction and excitation-transcription coupling in skeletal muscle. J Gen Physiol 2021; 153:212695. [PMID: 34636893 PMCID: PMC8515650 DOI: 10.1085/jgp.202012635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/24/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023] Open
Abstract
One of the most important functions of skeletal muscle is to respond to nerve stimuli by contracting. This function ensures body movement but also participates in other important physiological roles, like regulation of glucose homeostasis. Muscle activity is closely regulated to adapt to different demands and shows a plasticity that relies on both transcriptional activity and nerve stimuli. These two processes, both dependent on depolarization of the plasma membrane, have so far been regarded as separated and independent processes due to a lack of evidence of common protein partners or molecular mechanisms. In this study, we reveal intimate functional interactions between the process of excitation-induced contraction and the process of excitation-induced transcriptional activity in skeletal muscle. We show that the plasma membrane voltage-sensing protein CaV1.1 and the ATP-releasing channel Pannexin-1 (Panx1) regulate each other in a reciprocal manner, playing roles in both processes. Specifically, knockdown of CaV1.1 produces chronically elevated extracellular ATP concentrations at rest, consistent with disruption of the normal control of Panx1 activity. Conversely, knockdown of Panx1 affects not only activation of transcription but also CaV1.1 function on the control of muscle fiber contraction. Altogether, our results establish the presence of bidirectional functional regulations between the molecular machineries involved in the control of contraction and transcription induced by membrane depolarization of adult muscle fibers. Our results are important for an integrative understanding of skeletal muscle function and may impact our understanding of several neuromuscular diseases.
Collapse
Affiliation(s)
- Francisco Jaque-Fernández
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Gonzalo Jorquera
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jennifer Troc-Gajardo
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - France Pietri-Rouxel
- Université Pierre et Marie Curie, Université Paris 06, Institut National de la Santé et de la Recherche Médicale/Centre National de la Recherche Scientifique/Institut de Myologie/Centre de Recherche en Myologie, Groupement hospitalier universitaire Pitié Salpêtrière, Paris, France
| | - Christel Gentil
- Université Pierre et Marie Curie, Université Paris 06, Institut National de la Santé et de la Recherche Médicale/Centre National de la Recherche Scientifique/Institut de Myologie/Centre de Recherche en Myologie, Groupement hospitalier universitaire Pitié Salpêtrière, Paris, France
| | - Sonja Buvinic
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Bruno Allard
- Université Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR-5310, Institut National de la Santé et de la Recherche Médicale U-1217, Institut NeuroMyoGène, Lyon, France
| | - Enrique Jaimovich
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Vincent Jacquemond
- Université Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR-5310, Institut National de la Santé et de la Recherche Médicale U-1217, Institut NeuroMyoGène, Lyon, France
| | - Mariana Casas
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
45
|
Koval M, Cwiek A, Carr T, Good ME, Lohman AW, Isakson BE. Pannexin 1 as a driver of inflammation and ischemia-reperfusion injury. Purinergic Signal 2021; 17:521-531. [PMID: 34251590 PMCID: PMC8273370 DOI: 10.1007/s11302-021-09804-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023] Open
Abstract
Pannexin 1 (Panx1) is a ubiquitously expressed protein forming large conductance channels that are central to many distinct inflammation and injury responses. There is accumulating evidence showing ATP released from Panx1 channels, as well as metabolites, provide effective paracrine and autocrine signaling molecules that regulate different elements of the injury response. As channels with a broad range of permselectivity, Panx1 channels mediate the secretion and uptake of multiple solutes, ranging from calcium to bacterial derived molecules. In this review, we describe how Panx1 functions in response to different pro-inflammatory stimuli, focusing mainly on signaling coordinated by the vasculature. How Panx1 mediates ATP release by injured cells is also discussed. The ability of Panx1 to serve as a central component of many diverse physiologic responses has proven to be critically dependent on the context of expression, post-translational modification, interacting partners, and the mode of stimulation.
Collapse
Affiliation(s)
- Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA.
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Aleksandra Cwiek
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Thomas Carr
- Department of Cell Biology and Anatomy, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Miranda E Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Alexander W Lohman
- Department of Cell Biology and Anatomy, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, School of Medicine, University of Virginia, PO Box 801394, Charlottesville, VA, 22908, USA.
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
| |
Collapse
|
46
|
Erdener ŞE, Kaya Z, Dalkara T. Parenchymal neuroinflammatory signaling and dural neurogenic inflammation in migraine. J Headache Pain 2021; 22:138. [PMID: 34794382 PMCID: PMC8600694 DOI: 10.1186/s10194-021-01353-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background Pain is generally concomitant with an inflammatory reaction at the site where the nociceptive fibers are activated. Rodent studies suggest that a sterile meningeal inflammatory signaling cascade may play a role in migraine headache as well. Experimental studies also suggest that a parenchymal inflammatory signaling cascade may report the non-homeostatic conditions in brain to the meninges to induce headache. However, how these signaling mechanisms function in patients is unclear and debated. Our aim is to discuss the role of inflammatory signaling in migraine pathophysiology in light of recent developments. Body Rodent studies suggest that a sterile meningeal inflammatory reaction can be initiated by release of peptides from active trigeminocervical C-fibers and stimulation of resident macrophages and dendritic/mast cells. This inflammatory reaction might be needed for sustained stimulation and sensitization of meningeal nociceptors after initial activation along with ganglionic and central mechanisms. Most migraines likely have cerebral origin as suggested by prodromal neurologic symptoms. Based on rodent studies, a parenchymal inflammatory signaling cascade has been proposed as a potential mechanism linking cortical spreading depolarization (CSD) to meningeal nociception. A recent PET/MRI study using a sensitive inflammation marker showed the presence of meningeal inflammatory activity in migraine with aura patients over the occipital cortex generating the visual aura. These studies also suggest the presence of a parenchymal inflammatory activity, supporting the experimental findings. In rodents, parenchymal inflammatory signaling has also been shown to be activated by migraine triggers such as sleep deprivation without requiring a CSD because of the resultant transcriptional changes, predisposing to inadequate synaptic energy supply during intense excitatory transmission. Thus, it may be hypothesized that neuronal stress created by either CSD or synaptic activity-energy mismatch could both initiate a parenchymal inflammatory signaling cascade, propagating to the meninges, where it is converted to a lasting headache with or without aura. Conclusion Experimental studies in animals and emerging imaging findings from patients warrant further research to gain deeper insight to the complex role of inflammatory signaling in headache generation in migraine.
Collapse
Affiliation(s)
- Şefik Evren Erdener
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Zeynep Kaya
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, and Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
47
|
Mao XY, Yin XX, Guan QW, Xia QX, Yang N, Zhou HH, Liu ZQ, Jin WL. Dietary nutrition for neurological disease therapy: Current status and future directions. Pharmacol Ther 2021; 226:107861. [PMID: 33901506 DOI: 10.1016/j.pharmthera.2021.107861] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023]
Abstract
Adequate food intake and relative abundance of dietary nutrients have undisputed effects on the brain function. There is now substantial evidence that dietary nutrition aids in the prevention and remediation of neurologic symptoms in diverse pathological conditions. The newly described influences of dietary factors on the alterations of mitochondrial dysfunction, epigenetic modification and neuroinflammation are important mechanisms that are responsible for the action of nutrients on the brain health. In this review, we discuss the state of evidence supporting that distinct dietary interventions including dietary supplement and dietary restriction have the ability to tackle neurological disorders using Alzheimer's disease, Parkinson's disease, stroke, epilepsy, traumatic brain injury, amyotrophic lateral sclerosis, Huntington's disease and multiple sclerosis as examples. Additionally, it is also highlighting that diverse potential mechanisms such as metabolic control, epigenetic modification, neuroinflammation and gut-brain axis are of utmost importance for nutrient supply to the risk of neurologic condition and therapeutic response. Finally, we also highlight the novel concept that dietary nutrient intervention reshapes metabolism-epigenetics-immunity cycle to remediate brain dysfunction. Targeting metabolism-epigenetics-immunity network will delineate a new blueprint for combating neurological weaknesses.
Collapse
Affiliation(s)
- Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China.
| | - Xi-Xi Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Qi-Wen Guan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Qin-Xuan Xia
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Nan Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China.
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
48
|
Dossi E, Rouach N. Pannexin 1 channels and ATP release in epilepsy: two sides of the same coin : The contribution of pannexin-1, connexins, and CALHM ATP-release channels to purinergic signaling. Purinergic Signal 2021; 17:533-548. [PMID: 34495463 DOI: 10.1007/s11302-021-09818-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/08/2021] [Indexed: 11/29/2022] Open
Abstract
Purinergic signaling mediated by ATP and its metabolites contributes to various brain physiological processes as well as to several pathological conditions, including neurodegenerative and neurological disorders, such as epilepsy. Among the different ATP release pathways, pannexin 1 channels represent one of the major conduits being primarily activated in pathological contexts. Investigations on in vitro and in vivo models of epileptiform activity and seizures in mice and human tissues revealed pannexin 1 involvement in aberrant network activity and epilepsy, and highlighted that pannexin 1 exerts a complex role. Pannexin 1 can indeed either sustain seizures through release of ATP that can directly activate purinergic receptors, or tune down epileptic activity via ATP-derived adenosine that decreases neuronal excitability. Interestingly, in-depth analysis of the literature unveils that this dichotomy is only apparent, as it depends on the model of seizure induction and the type of evoked epileptiform activity, two factors that can differentially activate pannexin 1 channels and trigger distinct intracellular signaling cascades. Here, we review the general properties and ATP permeability of pannexin 1 channels, and discuss their impact on acute epileptiform activity and chronic epilepsy according to the regime of activity and disease state. These data pave the way for the development of new antiepileptic strategies selectively targeting pannexin 1 channels in a context-dependent manner.
Collapse
Affiliation(s)
- Elena Dossi
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Centre National de la Recherche Scientifique UMR 7241, Institut National de la Santé Et de la Recherche Médicale U1050, Collège de France, Labex Memolife, Université PSL, Paris, France.
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Centre National de la Recherche Scientifique UMR 7241, Institut National de la Santé Et de la Recherche Médicale U1050, Collège de France, Labex Memolife, Université PSL, Paris, France.
| |
Collapse
|
49
|
García-Rodríguez C, Bravo-Tobar ID, Duarte Y, Barrio LC, Sáez JC. Contribution of non-selective membrane channels and receptors in epilepsy. Pharmacol Ther 2021; 231:107980. [PMID: 34481811 DOI: 10.1016/j.pharmthera.2021.107980] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/14/2022]
Abstract
Overcoming refractory epilepsy's resistance to the combination of antiepileptic drugs (AED), mitigating side effects, and preventing sudden unexpected death in epilepsy are critical goals for therapy of this disorder. Current therapeutic strategies are based primarily on neurocentric mechanisms, overlooking the participation of astrocytes and microglia in the pathophysiology of epilepsy. This review is focused on a set of non-selective membrane channels (permeable to ions and small molecules), including channels and ionotropic receptors of neurons, astrocytes, and microglia, such as: the hemichannels formed by Cx43 and Panx1; the purinergic P2X7 receptors; the transient receptor potential vanilloid (TRPV1 and TRPV4) channels; calcium homeostasis modulators (CALHMs); transient receptor potential canonical (TRPC) channels; transient receptor potential melastatin (TRPM) channels; voltage-dependent anion channels (VDACs) and volume-regulated anion channels (VRACs), which all have in common being activated by epileptic activity and the capacity to exacerbate seizure intensity. Specifically, we highlight evidence for the activation of these channels/receptors during epilepsy including neuroinflammation and oxidative stress, discuss signaling pathways and feedback mechanisms, and propose the functions of each of them in acute and chronic epilepsy. Studying the role of these non-selective membrane channels in epilepsy and identifying appropriate blockers for one or more of them could provide complementary therapies to better alleviate the disease.
Collapse
Affiliation(s)
- Claudia García-Rodríguez
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Chile.
| | - Iván D Bravo-Tobar
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Chile
| | - Yorley Duarte
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Luis C Barrio
- Hospital Ramon y Cajal-IRYCIS, Centro de Tecnología Biomédica de la Universidad Politécnica, Madrid, Spain
| | - Juan C Sáez
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Chile.
| |
Collapse
|
50
|
Prieto-Villalobos J, Alvear TF, Liberona A, Lucero CM, Martínez-Araya CJ, Balmazabal J, Inostroza CA, Ramírez G, Gómez GI, Orellana JA. Astroglial Hemichannels and Pannexons: The Hidden Link between Maternal Inflammation and Neurological Disorders. Int J Mol Sci 2021; 22:ijms22179503. [PMID: 34502412 PMCID: PMC8430734 DOI: 10.3390/ijms22179503] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 12/11/2022] Open
Abstract
Maternal inflammation during pregnancy causes later-in-life alterations of the offspring’s brain structure and function. These abnormalities increase the risk of developing several psychiatric and neurological disorders, including schizophrenia, intellectual disability, bipolar disorder, autism spectrum disorder, microcephaly, and cerebral palsy. Here, we discuss how astrocytes might contribute to postnatal brain dysfunction following maternal inflammation, focusing on the signaling mediated by two families of plasma membrane channels: hemi-channels and pannexons. [Ca2+]i imbalance linked to the opening of astrocytic hemichannels and pannexons could disturb essential functions that sustain astrocytic survival and astrocyte-to-neuron support, including energy and redox homeostasis, uptake of K+ and glutamate, and the delivery of neurotrophic factors and energy-rich metabolites. Both phenomena could make neurons more susceptible to the harmful effect of prenatal inflammation and the experience of a second immune challenge during adulthood. On the other hand, maternal inflammation could cause excitotoxicity by producing the release of high amounts of gliotransmitters via astrocytic hemichannels/pannexons, eliciting further neuronal damage. Understanding how hemichannels and pannexons participate in maternal inflammation-induced brain abnormalities could be critical for developing pharmacological therapies against neurological disorders observed in the offspring.
Collapse
Affiliation(s)
- Juan Prieto-Villalobos
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Tanhia F. Alvear
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Andrés Liberona
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Claudia M. Lucero
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.M.L.); (G.I.G.)
| | - Claudio J. Martínez-Araya
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Javiera Balmazabal
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Carla A. Inostroza
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Gigliola Ramírez
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Gonzalo I. Gómez
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.M.L.); (G.I.G.)
| | - Juan A. Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
- Correspondence: ; Tel.: +56-23548105
| |
Collapse
|