1
|
Santiago-Carvalho I, Ishikawa M, Borges da Silva H. Channel plan: control of adaptive immune responses by pannexins. Trends Immunol 2024; 45:892-902. [PMID: 39393945 PMCID: PMC11560585 DOI: 10.1016/j.it.2024.09.009] [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: 07/16/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024]
Abstract
The development of mammalian adaptive (i.e., B and T cell-mediated) immune responses is tightly controlled at transcriptional, epigenetic, and metabolic levels. Signals derived from the extracellular milieu are crucial regulators of adaptive immunity. Beyond the traditionally studied cytokines and chemokines, many other extracellular metabolites can bind to specialized receptors and regulate T and B cell immune responses. These molecules often accumulate extracellularly through active export by plasma membrane transporters. For example, mammalian immune and non-immune cells express pannexin (PANX)1-3 channels on the plasma membrane, which release many distinct small molecules, notably intracellular ATP. Here, we review novel findings defining PANXs as crucial regulators of T and B cell immune responses in disease contexts such as cancer or viral infections.
Collapse
Affiliation(s)
| | - Masaki Ishikawa
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | | |
Collapse
|
2
|
Sanchez‐Pupo RE, Finch GA, Johnston DE, Craig H, Abdo R, Barr K, Kerfoot S, Dagnino L, Penuela S. Global pannexin 1 deletion increases tumor-infiltrating lymphocytes in the BRAF/Pten mouse melanoma model. Mol Oncol 2024; 18:969-987. [PMID: 38327091 PMCID: PMC10994229 DOI: 10.1002/1878-0261.13596] [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: 07/28/2023] [Revised: 11/20/2023] [Accepted: 01/18/2024] [Indexed: 02/09/2024] Open
Abstract
Immunotherapies for malignant melanoma seek to boost the anti-tumoral response of CD8+ T cells, but have a limited patient response rate, in part due to limited tumoral immune cell infiltration. Genetic or pharmacological inhibition of the pannexin 1 (PANX1) channel-forming protein is known to decrease melanoma cell tumorigenic properties in vitro and ex vivo. Here, we crossed Panx1 knockout (Panx1-/-) mice with the inducible melanoma model BrafCA, PtenloxP, Tyr::CreERT2 (BPC). We found that deleting the Panx1 gene in mice does not reduce BRAF(V600E)/Pten-driven primary tumor formation or improve survival. However, tumors in BPC-Panx1-/- mice exhibited a significant increase in the infiltration of CD8+ T lymphocytes, with no changes in the expression of early T-cell activation marker CD69, lymphocyte activation gene 3 protein (LAG-3) checkpoint receptor, or programmed cell death ligand-1 (PD-L1) in tumors when compared to the BPC-Panx1+/+ genotype. Our results suggest that, although Panx1 deletion does not overturn the aggressive BRAF/Pten-driven melanoma progression in vivo, it does increase the infiltration of effector immune T-cell populations in the tumor microenvironment. We propose that PANX1-targeted therapy could be explored as a strategy to increase tumor-infiltrating lymphocytes to boost anti-tumor immunity.
Collapse
Affiliation(s)
| | - Garth A. Finch
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
| | | | - Heather Craig
- Department of Microbiology and ImmunologyWestern UniversityLondonCanada
| | - Rober Abdo
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
| | - Kevin Barr
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
| | - Steven Kerfoot
- Department of Microbiology and ImmunologyWestern UniversityLondonCanada
| | - Lina Dagnino
- Department of Physiology and PharmacologyWestern UniversityLondonCanada
- Division of Experimental Oncology, Department of Oncology, Schulich School of Medicine and DentistryWestern UniversityLondonCanada
| | - Silvia Penuela
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
- Division of Experimental Oncology, Department of Oncology, Schulich School of Medicine and DentistryWestern UniversityLondonCanada
| |
Collapse
|
3
|
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: 0] [Impact Index Per Article: 0] [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
|
4
|
Hernandez CA, Eugenin EA. The role of Pannexin-1 channels, ATP, and purinergic receptors in the pathogenesis of HIV and SARS-CoV-2. Curr Opin Pharmacol 2023; 73:102404. [PMID: 37734241 PMCID: PMC10838406 DOI: 10.1016/j.coph.2023.102404] [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: 03/15/2023] [Accepted: 08/25/2023] [Indexed: 09/23/2023]
Abstract
Infectious agents such as human immune deficiency virus-1 (HIV) and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) use host proteins to infect, replicate, and induce inflammation within the host. A critical component of these diseases is the axis between pannexin-1 channels, extracellular ATP, and purinergic receptors. Here, we describe the potential therapeutic role of Pannexin-1/purinergic approaches to prevent or reduce the devastating consequences of these pathogens.
Collapse
Affiliation(s)
- Cristian A Hernandez
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Eliseo A Eugenin
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA.
| |
Collapse
|
5
|
Badaoui M, Chanson M. Intercellular Communication in Airway Epithelial Cell Regeneration: Potential Roles of Connexins and Pannexins. Int J Mol Sci 2023; 24:16160. [PMID: 38003349 PMCID: PMC10671439 DOI: 10.3390/ijms242216160] [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: 09/26/2023] [Revised: 10/19/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Connexins and pannexins are transmembrane proteins that can form direct (gap junctions) or indirect (connexons, pannexons) intercellular communication channels. By propagating ions, metabolites, sugars, nucleotides, miRNAs, and/or second messengers, they participate in a variety of physiological functions, such as tissue homeostasis and host defense. There is solid evidence supporting a role for intercellular signaling in various pulmonary inflammatory diseases where alteration of connexin/pannexin channel functional expression occurs, thus leading to abnormal intercellular communication pathways and contributing to pathophysiological aspects, such as innate immune defense and remodeling. The integrity of the airway epithelium, which is the first line of defense against invading microbes, is established and maintained by a repair mechanism that involves processes such as proliferation, migration, and differentiation. Here, we briefly summarize current knowledge on the contribution of connexins and pannexins to necessary processes of tissue repair and speculate on their possible involvement in the shaping of the airway epithelium integrity.
Collapse
Affiliation(s)
| | - Marc Chanson
- Department of Cell Physiology & Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
| |
Collapse
|
6
|
Van Campenhout R, De Groof TWM, Kadam P, Kwak BR, Muyldermans S, Devoogdt N, Vinken M. Nanobody-based pannexin1 channel inhibitors reduce inflammation in acute liver injury. J Nanobiotechnology 2023; 21:371. [PMID: 37821897 PMCID: PMC10566086 DOI: 10.1186/s12951-023-02137-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND The opening of pannexin1 channels is considered as a key event in inflammation. Pannexin1 channel-mediated release of adenosine triphosphate triggers inflammasome signaling and activation of immune cells. By doing so, pannexin1 channels play an important role in several inflammatory diseases. Although pannexin1 channel inhibition could represent a novel clinical strategy for treatment of inflammatory disorders, therapeutic pannexin1 channel targeting is impeded by the lack of specific, potent and/or in vivo-applicable inhibitors. The goal of this study is to generate nanobody-based inhibitors of pannexin1 channels. RESULTS Pannexin1-targeting nanobodies were developed as potential new pannexin1 channel inhibitors. We identified 3 cross-reactive nanobodies that showed affinity for both murine and human pannexin1 proteins. Flow cytometry experiments revealed binding capacities in the nanomolar range. Moreover, the pannexin1-targeting nanobodies were found to block pannexin1 channel-mediated release of adenosine triphosphate. The pannexin1-targeting nanobodies were also demonstrated to display anti-inflammatory effects in vitro through reduction of interleukin 1 beta amounts. This anti-inflammatory outcome was reproduced in vivo using a human-relevant mouse model of acute liver disease relying on acetaminophen overdosing. More specifically, the pannexin1-targeting nanobodies lowered serum levels of inflammatory cytokines and diminished liver damage. These effects were linked with alteration of the expression of several NLRP3 inflammasome components. CONCLUSIONS This study introduced for the first time specific, potent and in vivo-applicable nanobody-based inhibitors of pannexin1 channels. As demonstrated for the case of liver disease, the pannexin1-targeting nanobodies hold great promise as anti-inflammatory agents, yet this should be further tested for extrahepatic inflammatory disorders. Moreover, the pannexin1-targeting nanobodies represent novel tools for fundamental research regarding the role of pannexin1 channels in pathological and physiological processes.
Collapse
Affiliation(s)
- Raf Van Campenhout
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Timo W M De Groof
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Molecular Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Prashant Kadam
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Brenda R Kwak
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, CH-1211, Geneva, Switzerland
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Bioengineering Sciences Department, Vrije Universiteit Brussel, 1050, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Molecular Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Mathieu Vinken
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090, Brussels, Belgium.
| |
Collapse
|
7
|
Thakur A, Sharma V, Averbek S, Liang L, Pandya N, Kumar G, Cili A, Zhang K. Immune landscape and redox imbalance during neurological disorders in COVID-19. Cell Death Dis 2023; 14:593. [PMID: 37673862 PMCID: PMC10482955 DOI: 10.1038/s41419-023-06102-6] [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: 03/21/2023] [Revised: 08/13/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023]
Abstract
The outbreak of Coronavirus Disease 2019 (COVID-19) has prompted the scientific community to explore potential treatments or vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes the illness. While SARS-CoV-2 is mostly considered a respiratory pathogen, several neurological complications have been reported, raising questions about how it may enter the Central Nervous System (CNS). Receptors such as ACE2, CD147, TMPRSS2, and NRP1 have been identified in brain cells and may be involved in facilitating SARS-CoV-2 entry into the CNS. Moreover, proteins like P2X7 and Panx-1 may contribute to the pathogenesis of COVID-19. Additionally, the role of the immune system in the gravity of COVID-19 has been investigated with respect to both innate and adaptive immune responses caused by SARS-CoV-2 infection, which can lead to a cytokine storm, tissue damage, and neurological manifestations. A redox imbalance has also been linked to the pathogenesis of COVID-19, potentially causing mitochondrial dysfunction, and generating proinflammatory cytokines. This review summarizes different mechanisms of reactive oxygen species and neuro-inflammation that may contribute to the development of severe COVID-19, and recent progress in the study of immunological events and redox imbalance in neurological complications of COVID-19, and the role of bioinformatics in the study of neurological implications of COVID-19.
Collapse
Affiliation(s)
- Abhimanyu Thakur
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation-CAS Limited, Hong Kong SAR, Hong Kong.
| | - Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sera Averbek
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
- Technische Universität Darmstadt, Darmstadt, Germany
| | - Lifan Liang
- University of Pittsburgh, Pittsburgh, PA, USA
| | - Nirali Pandya
- Department of Chemistry, Faculty of Sciences, National University of Singapore, Singapore, Singapore
| | - Gaurav Kumar
- School of Biosciences and Biomedical Engineering, Department of Clinical Research, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Alma Cili
- Clinic of Hematology, University of Medicine, University Hospital center "Mother Teresa", Tirane, Albania
| | - Kui Zhang
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass sciences, Southwest University, Chongqing, China.
- Cancer Centre, Medical Research Institute, Southwest University, Chongqing, China.
| |
Collapse
|
8
|
Kobayashi D, Umemoto E, Miyasaka M. The role of extracellular ATP in homeostatic immune cell migration. Curr Opin Pharmacol 2023; 68:102331. [PMID: 36535235 DOI: 10.1016/j.coph.2022.102331] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/30/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
Antigen stimulation induces adenosine triphosphate (ATP) release from naïve lymphocytes in lymphoid tissues. However, previous studies indicated that the non-lytic release of ATP also occurs in most tissues and cell types under physiological conditions. Here, we show that extracellular ATP (eATP) is indeed constitutively produced by naïve T cells in response to lymphoid chemokines in uninflamed lymph nodes and is involved in the regulation of immune cell migration. In this review, we briefly summarize the homeostatic role of extracellular ATP in immune cell migration in vivo.
Collapse
Affiliation(s)
- Daichi Kobayashi
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Eiji Umemoto
- Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Masayuki Miyasaka
- Immunology Frontier Research Center, Osaka University, Suita, Japan.
| |
Collapse
|
9
|
Obot P, Subah G, Schonwald A, Pan J, Velíšek L, Velíšková J, Stanton PK, Scemes E. Astrocyte and neuronal Panx1 support long-term reference memory in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.16.524236. [PMID: 36711845 PMCID: PMC9882221 DOI: 10.1101/2023.01.16.524236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Pannexin 1 (Panx1) are ubiquitously expressed proteins that form plasma membrane channels permeable to anions and moderate sized signaling molecules (e.g., ATP, glutamate). In the nervous system, activation of Panx1 channels have been extensively shown to contribute to distinct neurological disorders (epilepsy, chronic pain, migraine, neuroAIDS, etc.) but knowledge of extent to which these channels have a physiological role remains restricted to three studies supporting their involvement in hippocampus dependent learning. Given that Panx1 channels may provide an important mechanism for activity-dependent neuron-glia interaction, we used Panx1 transgenic mice with global and cell-type specific deletions of Panx1 to interrogate their participation in working and reference memory. Using the 8-arm radial maze, we show that long-term spatial reference memory, but not spatial working memory, is deficient in Panx1-null mice and that both astrocyte and neuronal Panx1 contribute to the consolidation of long-term spatial memory. Field potential recordings in hippocampal slices of Panx1-null mice revealed an attenuation of both long-term potentiation (LTP) of synaptic strength and long-term depression (LTD) at Schaffer collateral - CA1 synapses without alterations basal synaptic transmission or pre-synaptic paired-pulse facilitation. Our results implicate both neuronal and astrocyte Panx1 channels as critical players for the development and maintenance of long-term spatial reference memory in mice.
Collapse
|
10
|
Obot P, Subah G, Schonwald A, Pan J, Velíšek L, Velíšková J, Stanton PK, Scemes E. Astrocyte and Neuronal Panx1 Support Long-Term Reference Memory in Mice. ASN Neuro 2023; 15:17590914231184712. [PMID: 37365910 DOI: 10.1177/17590914231184712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Pannexin 1 (Panx1) is an ubiquitously expressed protein that forms plasma membrane channels permeable to anions and moderate-sized signaling molecules (e.g., ATP, glutamate). In the nervous system, activation of Panx1 channels has been extensively shown to contribute to distinct neurological disorders (epilepsy, chronic pain, migraine, neuroAIDS, etc.), but knowledge of the extent to which these channels have a physiological role remains restricted to three studies supporting their involvement in hippocampus dependent learning. Given that Panx1 channels may provide an important mechanism for activity-dependent neuron-glia interaction, we used Panx1 transgenic mice with global and cell-type specific deletions of Panx1 to interrogate their participation in working and reference memory. Using the eight-arm radial maze, we show that long-term spatial reference memory, but not spatial working memory, is deficient in Panx1-null mice and that both astrocyte and neuronal Panx1 contribute to the consolidation of long-term spatial memory. Field potential recordings in hippocampal slices of Panx1-null mice revealed an attenuation of both long-term potentiation (LTP) of synaptic strength and long-term depression (LTD) at Schaffer collateral-CA1 synapses without alterations of basal synaptic transmission or pre-synaptic paired-pulse facilitation. Our results implicate both neuronal and astrocyte Panx1 channels as critical players for the development and maintenance of long-term spatial reference memory in mice.
Collapse
Affiliation(s)
- Price Obot
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Emergency Medicine, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Galadu Subah
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Antonia Schonwald
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Jian Pan
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Libor Velíšek
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Pediatrics, Penn State Hershey Medical Center, Hershey, PA, USA
- Department of Neurology, New York Medical College, Valhalla, NY, USA
| | - Jana Velíšková
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Obstetrics and Gynecology, New York Medical College, Valhalla, NY, USA
| | - Patric K Stanton
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Neurology, New York Medical College, Valhalla, NY, USA
| | - Eliana Scemes
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
11
|
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
|
12
|
Hernandez CA, Eliseo E. The Role of Pannexin-1 Channels in HIV and NeuroHIV Pathogenesis. Cells 2022; 11:2245. [PMID: 35883688 PMCID: PMC9323506 DOI: 10.3390/cells11142245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 02/06/2023] Open
Abstract
The human immunodeficiency virus-1 (HIV) enters the brain shortly after infection, leading to long-term neurological complications in half of the HIV-infected population, even in the current anti-retroviral therapy (ART) era. Despite decades of research, no biomarkers can objectively measure and, more importantly, predict the onset of HIV-associated neurocognitive disorders. Several biomarkers have been proposed; however, most of them only reflect late events of neuronal damage. Our laboratory recently identified that ATP and PGE2, inflammatory molecules released through Pannexin-1 channels, are elevated in the serum of HIV-infected individuals compared to uninfected individuals and other inflammatory diseases. More importantly, high circulating ATP levels, but not PGE2, can predict a decline in cognition, suggesting that HIV-infected individuals have impaired ATP metabolism and associated signaling. We identified that Pannexin-1 channel opening contributes to the high serological ATP levels, and ATP in the circulation could be used as a biomarker of HIV-associated cognitive impairment. In addition, we believe that ATP is a major contributor to chronic inflammation in the HIV-infected population, even in the anti-retroviral era. Here, we discuss the mechanisms associated with Pannexin-1 channel opening within the circulation, as well as within the resident viral reservoirs, ATP dysregulation, and cognitive disease observed in the HIV-infected population.
Collapse
Affiliation(s)
| | - Eugenin Eliseo
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA;
| |
Collapse
|
13
|
Harcha PA, López-López T, Palacios AG, Sáez PJ. Pannexin Channel Regulation of Cell Migration: Focus on Immune Cells. Front Immunol 2022; 12:750480. [PMID: 34975840 PMCID: PMC8716617 DOI: 10.3389/fimmu.2021.750480] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
The role of Pannexin (PANX) channels during collective and single cell migration is increasingly recognized. Amongst many functions that are relevant to cell migration, here we focus on the role of PANX-mediated adenine nucleotide release and associated autocrine and paracrine signaling. We also summarize the contribution of PANXs with the cytoskeleton, which is also key regulator of cell migration. PANXs, as mechanosensitive ATP releasing channels, provide a unique link between cell migration and purinergic communication. The functional association with several purinergic receptors, together with a plethora of signals that modulate their opening, allows PANX channels to integrate physical and chemical cues during inflammation. Ubiquitously expressed in almost all immune cells, PANX1 opening has been reported in different immunological contexts. Immune activation is the epitome coordination between cell communication and migration, as leukocytes (i.e., T cells, dendritic cells) exchange information while migrating towards the injury site. In the current review, we summarized the contribution of PANX channels during immune cell migration and recruitment; although we also compile the available evidence for non-immune cells (including fibroblasts, keratinocytes, astrocytes, and cancer cells). Finally, we discuss the current evidence of PANX1 and PANX3 channels as a both positive and/or negative regulator in different inflammatory conditions, proposing a general mechanism of these channels contribution during cell migration.
Collapse
Affiliation(s)
- Paloma A Harcha
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Tamara López-López
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Adrián G Palacios
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo J Sáez
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
Luu R, Valdebenito S, Scemes E, Cibelli A, Spray DC, Rovegno M, Tichauer J, Cottignies-Calamarte A, Rosenberg A, Capron C, Belouzard S, Dubuisson J, Annane D, de la Grandmaison GL, Cramer-Bordé E, Bomsel M, Eugenin E. Pannexin-1 channel opening is critical for COVID-19 pathogenesis. iScience 2021; 24:103478. [PMID: 34841222 PMCID: PMC8603863 DOI: 10.1016/j.isci.2021.103478] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/30/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) rapidly rampaged worldwide, causing a pandemic of coronavirus disease (COVID -19), but the biology of SARS-CoV-2 remains under investigation. We demonstrate that both SARS-CoV-2 spike protein and human coronavirus 229E (hCoV-229E) or its purified S protein, one of the main viruses responsible for the common cold, induce the transient opening of Pannexin-1 (Panx-1) channels in human lung epithelial cells. However, the Panx-1 channel opening induced by SARS-CoV-2 is greater and more prolonged than hCoV-229E/S protein, resulting in an enhanced ATP, PGE2, and IL-1β release. Analysis of lung lavages and tissues indicate that Panx-1 mRNA expression is associated with increased ATP, PGE2, and IL-1β levels. Panx-1 channel opening induced by SARS-CoV-2 spike protein is angiotensin-converting enzyme 2 (ACE-2), endocytosis, and furin dependent. Overall, we demonstrated that Panx-1 channel is a critical contributor to SARS-CoV-2 infection and should be considered as an alternative therapy.
Collapse
Affiliation(s)
- Ross Luu
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, 105 11th Street, Galveston, TX 77555, USA
| | - Silvana Valdebenito
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, 105 11th Street, Galveston, TX 77555, USA
| | - Eliana Scemes
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| | - Antonio Cibelli
- Dominick P. Purpura Department of Neuroscience & Department of Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - David C Spray
- Dominick P. Purpura Department of Neuroscience & Department of Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Maximiliano Rovegno
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Tichauer
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrea Cottignies-Calamarte
- Hôpital Cochin, Service de Virologie, Hôpital Cochin (AP-HP), Paris, France.,Service d'Hématologie Hôpital Ambroise Paré (AP-HP), Boulogne-Billancourt, France
| | - Arielle Rosenberg
- Hôpital Cochin, Service de Virologie, Hôpital Cochin (AP-HP), Paris, France.,Service d'Hématologie Hôpital Ambroise Paré (AP-HP), Boulogne-Billancourt, France.,Virologie Moléculaire et Cellulaire des Coronavirus, Centre d'infection et d'immunité de Lille, Institut Pasteur de Lille, Université de Lille, CNRS, Inserm, CHRU, 59000 Lille, France
| | - Calude Capron
- Service des Maladies Infectieuses, Centre Hospitalier Universitaire Raymond Poincaré, AP-HP, Garches, France
| | | | - Jean Dubuisson
- Intensive Care Unit, Raymond Poincaré Hospital (AP-HP), Paris, France
| | - Djillali Annane
- Simone Veil School of Medicine, Université of Versailles, Versailles, France.,University Paris Saclay, Garches, France
| | - Geoffroy Lorin de la Grandmaison
- Department of Forensic Medicine and Pathology, Versailles Saint-Quentin Université, AP-HP, Raymond Poincaré Hospital, Garches, France
| | | | - Morgane Bomsel
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Université de Paris, Paris, France.,INSERM U1016, Paris, France
| | - Eliseo Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, 105 11th Street, Galveston, TX 77555, USA
| |
Collapse
|
15
|
Muñoz MF, Griffith TN, Contreras JE. Mechanisms of ATP release in pain: role of pannexin and connexin channels. Purinergic Signal 2021; 17:549-561. [PMID: 34792743 PMCID: PMC8677853 DOI: 10.1007/s11302-021-09822-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Pain is a physiological response to bodily damage and serves as a warning of potential threat. Pain can also transform from an acute response to noxious stimuli to a chronic condition with notable emotional and psychological components that requires treatment. Indeed, the management of chronic pain is currently an important unmet societal need. Several reports have implicated the release of the neurotransmitter adenosine triphosphate (ATP) and subsequent activation of purinergic receptors in distinct pain etiologies. Purinergic receptors are broadly expressed in peripheral neurons and the spinal cord; thus, purinergic signaling in sensory neurons or in spinal circuits may be critical for pain processing. Nevertheless, an outstanding question remains: what are the mechanisms of ATP release that initiate nociceptive signaling? Connexin and pannexin channels are established conduits of ATP release and have been suggested to play important roles in a variety of pathologies, including several models of pain. As such, these large-pore channels represent a new and exciting putative pharmacological target for pain treatment. Herein, we will review the current evidence for a role of connexin and pannexin channels in ATP release during nociceptive signaling, such as neuropathic and inflammatory pain. Collectively, these studies provide compelling evidence for an important role of connexins and pannexins in pain processing.
Collapse
Affiliation(s)
- Manuel F. Muñoz
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, USA
| | - Theanne N. Griffith
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, USA
| | - Jorge E. Contreras
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, USA
| |
Collapse
|
16
|
G-protein-coupled receptor P2Y10 facilitates chemokine-induced CD4 T cell migration through autocrine/paracrine mediators. Nat Commun 2021; 12:6798. [PMID: 34815397 PMCID: PMC8611058 DOI: 10.1038/s41467-021-26882-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
G-protein-coupled receptors (GPCRs), especially chemokine receptors, play a central role in the regulation of T cell migration. Various GPCRs are upregulated in activated CD4 T cells, including P2Y10, a putative lysophospholipid receptor that is officially still considered an orphan GPCR, i.e., a receptor with unknown endogenous ligand. Here we show that in mice lacking P2Y10 in the CD4 T cell compartment, the severity of experimental autoimmune encephalomyelitis and cutaneous contact hypersensitivity is reduced. P2Y10-deficient CD4 T cells show normal activation, proliferation and differentiation, but reduced chemokine-induced migration, polarization, and RhoA activation upon in vitro stimulation. Mechanistically, CD4 T cells release the putative P2Y10 ligands lysophosphatidylserine and ATP upon chemokine exposure, and these mediators induce P2Y10-dependent RhoA activation in an autocrine/paracrine fashion. ATP degradation impairs RhoA activation and migration in control CD4 T cells, but not in P2Y10-deficient CD4 T cells. Importantly, the P2Y10 pathway appears to be conserved in human T cells. Taken together, P2Y10 mediates RhoA activation in CD4 T cells in response to auto-/paracrine-acting mediators such as LysoPS and ATP, thereby facilitating chemokine-induced migration and, consecutively, T cell-mediated diseases.
Collapse
|
17
|
D'Amico D, Valdebenito S, Eugenin EA. The role of Pannexin-1 channels and extracellular ATP in the pathogenesis of the human immunodeficiency virus. Purinergic Signal 2021; 17:563-576. [PMID: 34542793 DOI: 10.1007/s11302-021-09817-3] [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: 06/03/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022] Open
Abstract
Only recently, the role of large ionic channels such as Pannexin-1 channels and Connexin hemichannels has been implicated in several physiological and pathological conditions, including HIV infection and associated comorbidities. These channels are in a closed stage in healthy conditions, but in pathological conditions including HIV, Pannexin-1 channels and Connexin hemichannels become open. Our data demonstrate that acute and chronic HIV infection induces channel opening (Pannexin and Connexin channels), ATP release into the extracellular space, and subsequent activation of purinergic receptors in immune and non-immune cells. We demonstrated that Pannexin and Connexin channels contribute to HIV infection and replication, the long-term survival of viral reservoirs, and comorbidities such as NeuroHIV. Here, we discuss the available data to support the participation of these channels in the HIV life cycle and the potential therapeutic approach to prevent HIV-associated comorbidities.
Collapse
Affiliation(s)
- Daniela D'Amico
- Department of Neuroscience , Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, 105 11th Street, Galveston, TX, 77555, USA
| | - Silvana Valdebenito
- Department of Neuroscience , Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, 105 11th Street, Galveston, TX, 77555, USA
| | - Eliseo A Eugenin
- Department of Neuroscience , Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, 105 11th Street, Galveston, TX, 77555, USA.
| |
Collapse
|
18
|
Laird DW, Penuela S. Pannexin biology and emerging linkages to cancer. Trends Cancer 2021; 7:1119-1131. [PMID: 34389277 DOI: 10.1016/j.trecan.2021.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/18/2022]
Abstract
Pannexins are a family of glycoproteins that comprises three members, PANX1, PANX2, and PANX3. The widely expressed and interrogated PANX1 forms heptameric membrane channels that primarily serve to connect the cytoplasm to the extracellular milieu by being selectively permeable to small signaling molecules when activated. Apart from notable exceptions, PANX1 in many tumor cells appears to facilitate tumor growth and metastasis, suggesting that pannexin-blocking therapeutics may have utility in cancer. Attenuation of PANX1 function must also consider the fact that PANX1 is found in stromal cells of the tumor microenvironment (TME), including immune cells. This review highlights the key discoveries of the past 5 years that suggest pannexins facilitate, or in some cases inhibit, tumor cell growth and metastasis via direct protein interactions and through the regulated efflux of signaling molecules.
Collapse
Affiliation(s)
- Dale W Laird
- Department of Anatomy and Cell Biology, and Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Department of Oncology, Divisions of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
19
|
Medina CB, Chiu YH, Stremska ME, Lucas CD, Poon I, Tung KS, Elliott MR, Desai B, Lorenz UM, Bayliss DA, Ravichandran KS. Pannexin 1 channels facilitate communication between T cells to restrict the severity of airway inflammation. Immunity 2021; 54:1715-1727.e7. [PMID: 34283971 PMCID: PMC8363584 DOI: 10.1016/j.immuni.2021.06.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/21/2021] [Accepted: 06/21/2021] [Indexed: 01/14/2023]
Abstract
Allergic airway inflammation is driven by type-2 CD4+ T cell inflammatory responses. We uncover an immunoregulatory role for the nucleotide release channel, Panx1, in T cell crosstalk during airway disease. Inverse correlations between Panx1 and asthmatics and our mouse models revealed the necessity, specificity, and sufficiency of Panx1 in T cells to restrict inflammation. Global Panx1-/- mice experienced exacerbated airway inflammation, and T-cell-specific deletion phenocopied Panx1-/- mice. A transgenic designed to re-express Panx1 in T cells reversed disease severity in global Panx1-/- mice. Panx1 activation occurred in pro-inflammatory T effector (Teff) and inhibitory T regulatory (Treg) cells and mediated the extracellular-nucleotide-based Treg-Teff crosstalk required for suppression of Teff cell proliferation. Mechanistic studies identified a Salt-inducible kinase-dependent phosphorylation of Panx1 serine 205 important for channel activation. A genetically targeted mouse expressing non-phosphorylatable Panx1S205A phenocopied the exacerbated inflammation in Panx1-/- mice. These data identify Panx1-dependent Treg:Teff cell communication in restricting airway disease.
Collapse
Affiliation(s)
- Christopher B Medina
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA; Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yu-Hsin Chiu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu, 30071, Taiwan
| | - Marta E Stremska
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Christopher D Lucas
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA
| | - Ivan Poon
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, VIC 3083, Australia
| | - Kenneth S Tung
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Michael R Elliott
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA; Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Bimal Desai
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ulrike M Lorenz
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kodi S Ravichandran
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA; Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA; VIB/UGent Inflammation Research Centre and the Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
20
|
Chiu YH, Medina CB, Doyle CA, Zhou M, Narahari AK, Sandilos JK, Gonye EC, Gao HY, Guo SY, Parlak M, Lorenz UM, Conrads TP, Desai BN, Ravichandran KS, Bayliss DA. Deacetylation as a receptor-regulated direct activation switch for pannexin channels. Nat Commun 2021; 12:4482. [PMID: 34301959 PMCID: PMC8302610 DOI: 10.1038/s41467-021-24825-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 07/12/2021] [Indexed: 02/07/2023] Open
Abstract
Activation of Pannexin 1 (PANX1) ion channels causes release of intercellular signaling molecules in a variety of (patho)physiological contexts. PANX1 can be activated by G protein-coupled receptors (GPCRs), including α1-adrenergic receptors (α1-ARs), but how receptor engagement leads to channel opening remains unclear. Here, we show that GPCR-mediated PANX1 activation can occur via channel deacetylation. We find that α1-AR-mediated activation of PANX1 channels requires Gαq but is independent of phospholipase C or intracellular calcium. Instead, α1-AR-mediated PANX1 activation involves RhoA, mammalian diaphanous (mDia)-related formin, and a cytosolic lysine deacetylase activated by mDia - histone deacetylase 6. HDAC6 associates with PANX1 and activates PANX1 channels, even in excised membrane patches, suggesting direct deacetylation of PANX1. Substitution of basally-acetylated intracellular lysine residues identified on PANX1 by mass spectrometry either prevents HDAC6-mediated activation (K140/409Q) or renders the channels constitutively active (K140R). These data define a non-canonical RhoA-mDia-HDAC6 signaling pathway for GαqPCR activation of PANX1 channels and uncover lysine acetylation-deacetylation as an ion channel silencing-activation mechanism.
Collapse
Affiliation(s)
- Yu-Hsin Chiu
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
| | - Christopher B Medina
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Catherine A Doyle
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Ming Zhou
- Inova Center for Personalized Health, Inova Schar Cancer Institute, Fairfax, VA, USA
| | - Adishesh K Narahari
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Joanna K Sandilos
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Elizabeth C Gonye
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Hong-Yu Gao
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Shih Yi Guo
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Mahmut Parlak
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Ulrike M Lorenz
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Thomas P Conrads
- Inova Center for Personalized Health, Inova Schar Cancer Institute, Fairfax, VA, USA
| | - Bimal N Desai
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
21
|
Silvana V, Paul C, Ajasin D, Eugenin EA. Astrocytes are HIV reservoirs in the brain: A cell type with poor HIV infectivity and replication but efficient cell-to-cell viral transfer. J Neurochem 2021; 158:429-443. [PMID: 33655498 PMCID: PMC11102126 DOI: 10.1111/jnc.15336] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/06/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022]
Abstract
The major barrier to eradicating Human immunodeficiency virus-1 (HIV) infection is the generation of tissue-associated quiescent long-lasting viral reservoirs refractory to therapy. Upon interruption of anti-retroviral therapy (ART), HIV replication can be reactivated. Within the brain, microglia/macrophages and a small population of astrocytes are infected with HIV. However, the role of astrocytes as a potential viral reservoir is becoming more recognized because of the improved detection and quantification of HIV viral reservoirs. In this report, we examined the infectivity of human primary astrocytes in vivo and in vitro, and their capacity to maintain HIV infection, become latently infected, be reactivated, and transfer new HIV virions into neighboring cells. Analysis of human brain tissue sections obtained from HIV-infected individuals under effective and prolonged ART indicates that a small population of astrocytes has integrated HIV-DNA. In vitro experiments using HIV-infected human primary astrocyte cultures confirmed a low percentage of astrocytes had integrated HIV-DNA, with poor to undetectable replication. Even in the absence of ART, long-term culture results in latency that could be transiently reactivated with histone deacetylase inhibitor, tumor necrosis factor-alpha (TNF-α), or methamphetamine. Reactivation resulted in poor viral production but efficient cell-to-cell viral transfer into cells that support high viral replication. Together, our data provide a new understanding of astrocytes' role as viral reservoirs within the central nervous system (CNS).
Collapse
Affiliation(s)
- Valdebenito Silvana
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, Texas, USA
| | - Castellano Paul
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, Texas, USA
| | - David Ajasin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, Texas, USA
| | - Eliseo A. Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, Texas, USA
| |
Collapse
|
22
|
Ledderose C, Junger WG. Mitochondria Synergize With P2 Receptors to Regulate Human T Cell Function. Front Immunol 2020; 11:549889. [PMID: 33133068 PMCID: PMC7550529 DOI: 10.3389/fimmu.2020.549889] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
Intracellular ATP is the universal energy carrier that fuels many cellular processes. However, immune cells can also release a portion of their ATP into the extracellular space. There, ATP activates purinergic receptors that mediate autocrine and paracrine signaling events needed for the initiation, modulation, and termination of cell functions. Mitochondria contribute to these processes by producing ATP that is released. Here, we summarize the synergistic interplay between mitochondria and purinergic signaling that regulates T cell functions. Specifically, we discuss how mitochondria interact with P2X1, P2X4, and P2Y11 receptors to regulate T cell metabolism, cell migration, and antigen recognition. These mitochondrial and purinergic signaling mechanisms are indispensable for host immune defense. However, they also represent an Achilles heel that can render the host susceptible to infections and inflammatory disorders. Hypoxia and mitochondrial dysfunction deflate the purinergic signaling mechanisms that regulate T cells, while inflammation and tissue damage generate excessive systemic ATP levels that distort autocrine purinergic signaling and impair T cell function. An improved understanding of the metabolic and purinergic signaling mechanisms that regulate T cells may lead to novel strategies for the diagnosis and treatment of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
23
|
Ledderose C, Bromberger S, Slubowski CJ, Sueyoshi K, Aytan D, Shen Y, Junger WG. The purinergic receptor P2Y11 choreographs the polarization, mitochondrial metabolism, and migration of T lymphocytes. Sci Signal 2020; 13:13/651/eaba3300. [PMID: 32994212 DOI: 10.1126/scisignal.aba3300] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
T cells must migrate to encounter antigen-presenting cells and perform their roles in host defense. Here, we found that autocrine stimulation of the purinergic receptor P2Y11 regulates the migration of human CD4 T cells. P2Y11 receptors redistributed from the front to the back of polarized cells where they triggered intracellular cAMP/PKA signals that attenuated mitochondrial metabolism at the back. The absence of P2Y11 receptors at the front of cells resulted in hotspots of mitochondrial metabolism and localized ATP production that stimulated P2X4 receptors, Ca2+ influx, and pseudopod protrusion at the front. This regulatory function of P2Y11 receptors depended on their subcellular redistribution and autocrine stimulation by cellular ATP release and was perturbed by indiscriminate global stimulation. We conclude that excessive extracellular ATP-such as in response to inflammation, sepsis, and cancer-disrupts this autocrine feedback mechanism, which results in defective T cell migration, impaired T cell function, and loss of host immune defense.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Sophie Bromberger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christian J Slubowski
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Koichiro Sueyoshi
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dilan Aytan
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yong Shen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
24
|
Myeloid Pannexin-1 mediates acute leukocyte infiltration and leads to worse outcomes after brain trauma. J Neuroinflammation 2020; 17:245. [PMID: 32819386 PMCID: PMC7441665 DOI: 10.1186/s12974-020-01917-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 08/04/2020] [Indexed: 01/09/2023] Open
Abstract
Background Neuroinflammation is a major component of secondary damage after traumatic brain injury (TBI). We recently reported that pharmacological inhibition of Pannexin-1 (Panx1) channels markedly reduced the inflammatory response after TBI. Panx1 channels have been shown to be important conduits for adenosine 5′-triphosphate (ATP) release and are associated with leukocyte infiltration and pyroptosis. Because Panx1 blockers significantly decrease ATP release and migration of activated microglia and other myeloid cells (such as monocyte-derived macrophages and dendritic cells) in vitro, we hypothesized that myeloid Panx1 channels play a specific role in immune cell infiltration promoting tissue damage following TBI. Methods The murine-controlled cortical impact (CCI) model was used on myeloid-specific Panx1 conditional knockout (Cx3cr1-Cre::Panx1fl/fl) mice to determine whether myeloid Panx1 mediates neuroinflammation and brain damage. Immune cell infiltration was measured using flow cytometry. Locomotor and memory functions were measured using the rotarod and Barnes maze test, respectively. The levels of biomarkers for tissue damage and blood–brain barrier leakage were measured using western blot and magnetic resonance imaging. Panx1 channel activity was measured with ex vivo dye uptake assays, using flow cytometry and confocal microscopy. Results CCI-injured Cx3cr1-Cre::Panx1fl/fl mice showed markedly reduced immune cell infiltration to the brain parenchyma compared with Panx1fl/fl mice. As expected, Panx1 dependent activity, assessed by dye uptake, was markedly reduced only in myeloid cells from Cx3cr1-Cre::Panx1fl/fl mice. The expression of biomarkers of tissue damage was significantly reduced in the CCI-injured Cx3cr1-Cre::Panx1fl/fl mice compared with Panx1fl/fl mice. In line with this, magnetic resonance imaging showed reduced blood–brain barrier leakage in CCI-injured Cx3cr1-Cre::Panx1fl/fl mice. There was also a significant improvement in motor and memory function in Cx3cr1-Cre::Panx1fl/fl mice when compared with Panx1fl/fl mice within a week post-CCI injury. Conclusion Our data demonstrate that CCI-related outcomes correlate with Panx1 channel function in myeloid cells, indicating that activation of Panx1 channels in myeloid cells is a major contributor to acute brain inflammation following TBI. Importantly, our data indicate myeloid Panx1 channels could serve as an effective therapeutic target to improve outcome after TBI.
Collapse
|
25
|
Yeung AK, Patil CS, Jackson MF. Pannexin‐1 in the CNS: Emerging concepts in health and disease. J Neurochem 2020; 154:468-485. [DOI: 10.1111/jnc.15004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/26/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Albert K. Yeung
- Department of Pharmacology and Therapeutics Max Rady College of Medicine Rady Faculty of Health Sciences University of Manitoba Winnipeg Manitoba Canada
- Neuroscience Research Program Kleysen Institute for Advanced Medicine University of Manitoba Winnipeg Manitoba Canada
| | - Chetan S. Patil
- Department of Pharmacology and Therapeutics Max Rady College of Medicine Rady Faculty of Health Sciences University of Manitoba Winnipeg Manitoba Canada
- Neuroscience Research Program Kleysen Institute for Advanced Medicine University of Manitoba Winnipeg Manitoba Canada
| | - Michael F. Jackson
- Department of Pharmacology and Therapeutics Max Rady College of Medicine Rady Faculty of Health Sciences University of Manitoba Winnipeg Manitoba Canada
- Neuroscience Research Program Kleysen Institute for Advanced Medicine University of Manitoba Winnipeg Manitoba Canada
| |
Collapse
|
26
|
Circulating levels of ATP is a biomarker of HIV cognitive impairment. EBioMedicine 2019; 51:102503. [PMID: 31806564 PMCID: PMC7000317 DOI: 10.1016/j.ebiom.2019.10.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/14/2019] [Accepted: 10/16/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND In developed countries, Human Immunodeficiency Virus type-1 (HIV-1) infection has become a chronic disease despite the positive effects of anti-retroviral therapies (ART), but still at least half of the HIV infected population shown signs of cognitive impairment. Therefore, biomarkers of HIV cognitive decline are urgently needed. METHODS We analyze the opening of one of the larger channels expressed by humans, pannexin-1 (Panx-1) channels, in the uninfected and HIV infected population (n = 175). We determined channel opening and secretion of intracellular second messengers released through the channel such as PGE2 and ATP. Also, we correlated the opening of Panx-1 channels with the circulating levels of PGE2 and ATP as well as cogntive status of the individuals analyzed. FINDINGS Here, we demonstrate that Panx-1 channels on fresh PBMCs obtained from uninfected individuals are closed and no significant amounts of PGE2 and ATP are detected in the circulation. In contrast, in all HIV-infected individuals analyzed, even the ones under effective ART, a spontaneous opening of Panx-1 channels and increased circulating levels of PGE2 and ATP were detected. Circulating levels of ATP were correlated with cognitive decline in the HIV-infected population supporting that ATP is a biomarker of cognitive disease in the HIV-infected population. INTERPRETATION We propose that circulating levels of ATP could predict CNS compromise and lead to the breakthroughs necessary to detect and prevent brain compromise in the HIV-infected population.
Collapse
|
27
|
Novielli-Kuntz NM, Jelen M, Barr K, DeLalio LJ, Feng Q, Isakson BE, Gros R, Laird DW. Ablation of both Cx40 and Panx1 results in similar cardiovascular phenotypes exhibited in Cx40 knockout mice. Biosci Rep 2019; 39:BSR20182350. [PMID: 30745457 PMCID: PMC6393227 DOI: 10.1042/bsr20182350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/10/2019] [Accepted: 02/05/2019] [Indexed: 11/30/2022] Open
Abstract
Connexins (Cxs) and pannexins (Panxs) are highly regulated large-pore channel-forming proteins that participate in cellular communication via small molecular exchange with the extracellular microenvironment, or in the case of connexins, directly between cells. Given the putative functional overlap between single membrane-spanning connexin hemichannels and Panx channels, and cardiovascular system prevalence, we generated the first Cx40-/-Panx1-/- mouse with the anticipation that this genetic modification would lead to a severe cardiovascular phenotype. Mice null for both Cx40 and Panx1 produced litter sizes and adult growth progression similar to wild-type (WT), Cx40-/- and Panx1-/- mice. Akin to Cx40-/- mice, Cx40-/-Panx1-/- mice exhibited cardiac hypertrophy and elevated systolic, diastolic, and mean arterial blood pressure compared with WT and Panx1-/- mice; however assessment of left ventricular ejection fraction and fractional shortening revealed no evidence of cardiac dysfunction between groups. Furthermore, Cx40-/-, Panx1-/-, and Cx40-/-Panx1-/- mice demonstrated impaired endothelial-mediated vasodilation of aortic segments to increasing concentrations of methacholine (MCh) compared with WT, highlighting roles for both Cx40 and Panx1 in vascular endothelial cell (EC) function. Surprisingly, elevated kidney renin mRNA expression, plasma renin activity, and extraglomerular renin-producing cell populations found in Cx40-/- mice was further exaggerated in double knockout mice. Thus, while gestation and gross development were conserved in Cx40-/-Panx1-/- mice, they exhibit cardiac hypertrophy, hypertension, and impaired endothelial-mediated vasodilation that phenocopies Cx40-/- mice. Nevertheless, the augmented renin homeostasis observed in the double knockout mice suggests that both Cx40 and Panx1 may play an integrative role.
Collapse
Affiliation(s)
| | - Meghan Jelen
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Canada
| | - Kevin Barr
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Canada
| | - Leon J DeLalio
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, U.S.A
| | - Qingping Feng
- Department of Physiology and Pharmacology London, ON, Canada
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, U.S.A
| | - Robert Gros
- Department of Physiology and Pharmacology London, ON, Canada
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Canada
- Department of Physiology and Pharmacology London, ON, Canada
| |
Collapse
|
28
|
Li S, Li J, Wang N, Zhang T, Xu Y, Sun J. Expression analysis of Pannexin1 channel gene in response to immune challenges and its role in infection-induced ATP release in tilapia (Oreochromis niloticus). FISH & SHELLFISH IMMUNOLOGY 2018; 81:470-475. [PMID: 30064016 DOI: 10.1016/j.fsi.2018.07.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/21/2018] [Accepted: 07/27/2018] [Indexed: 06/08/2023]
Abstract
ATP released from immune cells plays an important role in activation of host innate immunity. However, the molecular mechanisms for pathogen infection-induced ATP release in fish remains unclear. Pannexin1 (Panx1) is a recently identified ATP release channel important for controlling immune responses. The immune relevance of Panx1 in fish, however, is still poorly understood. In this study, we characterized a Panx1 gene homologue (termed tPanx1) from Nile tilapia (Oreochromis niloticus) and analyzed its expression in response to different immune challenges. We also investigated the role of tPanx1 channel in bacterial infection-induced ATP release. Real-time quantitative PCR analysis revealed that tPanx1 gene is expressed in all tested tissues with predominant expression in intestine. Immune challenges with lipopolysaccharide, polyinosinic-polycytidylic acid and zymosan led to increased gene expression of tPanx1 in tilapia head kidney cells and peripheral blood leucocytes. In addition, tPanx1 gene was up-regulated in hepatopancreas, muscle, spleen, gill, head kidney and blood after Aeromonas hydrophila infection. Furthermore, pharmacological inhibition of tPanx1 channel activity with Panx1 channel inhibitor, carbenoxolone, significantly attenuated A. hydrophila infection-induced ATP release in tilapia head kidney cells. Taken together, our findings suggested that tPanx1 is an important immune response gene involved in bacterial infection-induced ATP release in tilapia O. niloticus.
Collapse
Affiliation(s)
- Shuo Li
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China.
| | - Jiafang Li
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China
| | - Nan Wang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China
| | - Tianxu Zhang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China
| | - Yaqi Xu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China
| | - Jinsheng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China.
| |
Collapse
|
29
|
Ledderose C, Liu K, Kondo Y, Slubowski CJ, Dertnig T, Denicoló S, Arbab M, Hubner J, Konrad K, Fakhari M, Lederer JA, Robson SC, Visner GA, Junger WG. Purinergic P2X4 receptors and mitochondrial ATP production regulate T cell migration. J Clin Invest 2018; 128:3583-3594. [PMID: 29894310 DOI: 10.1172/jci120972] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
T cells must migrate in order to encounter antigen-presenting cells (APCs) and to execute their varied functions in immune defense and inflammation. ATP release and autocrine signaling through purinergic receptors contribute to T cell activation at the immune synapse that T cells form with APCs. Here, we show that T cells also require ATP release and purinergic signaling for their migration to APCs. We found that the chemokine stromal-derived factor-1α (SDF-1α) triggered mitochondrial ATP production, rapid bursts of ATP release, and increased migration of primary human CD4+ T cells. This process depended on pannexin-1 ATP release channels and autocrine stimulation of P2X4 receptors. SDF-1α stimulation caused localized accumulation of mitochondria with P2X4 receptors near the front of cells, resulting in a feed-forward signaling mechanism that promotes cellular Ca2+ influx and sustains mitochondrial ATP synthesis at levels needed for pseudopod protrusion, T cell polarization, and cell migration. Inhibition of P2X4 receptors blocked the activation and migration of T cells in vitro. In a mouse lung transplant model, P2X4 receptor antagonist treatment prevented the recruitment of T cells into allograft tissue and the rejection of lung transplants. Our findings suggest that P2X4 receptors are therapeutic targets for immunomodulation in transplantation and inflammatory diseases.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kaifeng Liu
- Department of Medicine/Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yutaka Kondo
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian J Slubowski
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Dertnig
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara Denicoló
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mona Arbab
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Johannes Hubner
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kirstin Konrad
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mahtab Fakhari
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Simon C Robson
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Gary A Visner
- Department of Medicine/Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Ludwig Boltzmann Institute for Traumatology, Vienna, Austria
| |
Collapse
|
30
|
Makarenkova HP, Shah SB, Shestopalov VI. The two faces of pannexins: new roles in inflammation and repair. J Inflamm Res 2018; 11:273-288. [PMID: 29950881 PMCID: PMC6016592 DOI: 10.2147/jir.s128401] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Pannexins belong to a family of ATP-release channels expressed in almost all cell types. An increasing body of literature on pannexins suggests that these channels play dual and sometimes contradictory roles, contributing to normal cell function, as well as to the pathological progression of disease. In this review, we summarize our understanding of pannexin "protective" and "harmful" functions in inflammation, regeneration and mechanical signaling. We also suggest a possible basis for pannexin's dual roles, related to extracellular ATP and K+ levels and the activation of various types of P2 receptors that are associated with pannexin. Finally, we speculate upon therapeutic strategies related to pannexin using eyes, lacrimal glands, and peripheral nerves as examples of interesting therapeutic targets.
Collapse
Affiliation(s)
| | - Sameer B Shah
- Departments of Orthopaedic Surgery and Bioengineering, University of California.,Research Division, Veterans Affairs San Diego Healthcare System, San Diego, CA
| | - Valery I Shestopalov
- Bascom Eye Institute, Department of Ophthalmology, University of Miami, Miami, FL, USA.,Vavilov Institute for General Genetics, Russian Academy of Sciences.,Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
31
|
Chiu YH, Schappe MS, Desai BN, Bayliss DA. Revisiting multimodal activation and channel properties of Pannexin 1. J Gen Physiol 2017; 150:19-39. [PMID: 29233884 PMCID: PMC5749114 DOI: 10.1085/jgp.201711888] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 11/09/2017] [Indexed: 12/23/2022] Open
Abstract
Pannexin 1 (Panx1) forms plasma membrane ion channels that are widely expressed throughout the body. Panx1 activation results in the release of nucleotides such as adenosine triphosphate and uridine triphosphate. Thus, these channels have been implicated in diverse physiological and pathological functions associated with purinergic signaling, such as apoptotic cell clearance, blood pressure regulation, neuropathic pain, and excitotoxicity. In light of this, substantial attention has been directed to understanding the mechanisms that regulate Panx1 channel expression and activation. Here we review accumulated evidence for the various activation mechanisms described for Panx1 channels and, where possible, the unitary channel properties associated with those forms of activation. We also emphasize current limitations in studying Panx1 channel function and propose potential directions to clarify the exciting and expanding roles of Panx1 channels.
Collapse
Affiliation(s)
- Yu-Hsin Chiu
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA
| | - Michael S Schappe
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA
| | - Bimal N Desai
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
32
|
Hainz N, Wolf S, Beck A, Wagenpfeil S, Tschernig T, Meier C. Probenecid arrests the progression of pronounced clinical symptoms in a mouse model of multiple sclerosis. Sci Rep 2017; 7:17214. [PMID: 29222419 PMCID: PMC5722901 DOI: 10.1038/s41598-017-17517-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 11/27/2017] [Indexed: 12/05/2022] Open
Abstract
While it has been established that Probenecid (PBN) prevents the onset of experimental autoimmune encephalomyelitis (EAE) in mice, it is not clear whether it has any effect on already manifest EAE. The aim of this study was therefore to analyze the therapeutic effect of PBN in pronounced EAE. Mice with manifest clinical symptoms of EAE were either treated with PBN or solvent for 20 days, or they were left untreated. The clinical symptoms were monitored daily. Inflammation, demyelination and oligodendrocyte numbers were determined in the spinal cord. We were able to demonstrate that PBN not only significantly prolonged survival but also prevented the progression of clinical symptoms in the EAE model of multiple sclerosis. In addition, we were able to show that PBN reduced inflammation, T cell infiltration and oligodendrocyte cell loss. PBN was previously shown to inhibit – among other targets – pannexin channels. As pannexin channels provide conduits for ATP, are associated with the inflammasome, and act as “find me-signals” in the process of apoptosis, inhibition of pannexins via PBN might contribute to the PBN-effects observed in this study. The beneficial and therapeutic effects of PBN in the context of EAE demonstrate an intriguing link between PBN and neuroinflammation, which might foster translational interest.
Collapse
Affiliation(s)
- Nadine Hainz
- Dept. of Anatomy and Cell Biology, Saarland University, Homburg/Saar, Germany
| | - Sandra Wolf
- Dept. of Anatomy and Cell Biology, Saarland University, Homburg/Saar, Germany
| | - Artjom Beck
- Dept. of Anatomy and Cell Biology, Saarland University, Homburg/Saar, Germany
| | - Stefan Wagenpfeil
- Institute for Medical Biometry, Epidemiology & Medical Informatics, Saarland University, Homburg/Saar, Germany
| | - Thomas Tschernig
- Dept. of Anatomy and Cell Biology, Saarland University, Homburg/Saar, Germany
| | - Carola Meier
- Dept. of Anatomy and Cell Biology, Saarland University, Homburg/Saar, Germany.
| |
Collapse
|
33
|
Willebrords J, Maes M, Crespo Yanguas S, Vinken M. Inhibitors of connexin and pannexin channels as potential therapeutics. Pharmacol Ther 2017; 180:144-160. [PMID: 28720428 PMCID: PMC5802387 DOI: 10.1016/j.pharmthera.2017.07.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
While gap junctions support the exchange of a number of molecules between neighboring cells, connexin hemichannels provide communication between the cytosol and the extracellular environment of an individual cell. The latter equally holds true for channels composed of pannexin proteins, which display an architecture reminiscent of connexin hemichannels. In physiological conditions, gap junctions are usually open, while connexin hemichannels and, to a lesser extent, pannexin channels are typically closed, yet they can be activated by a number of pathological triggers. Several agents are available to inhibit channels built up by connexin and pannexin proteins, including alcoholic substances, glycyrrhetinic acid, anesthetics and fatty acids. These compounds not always strictly distinguish between gap junctions, connexin hemichannels and pannexin channels, and may have effects on other targets as well. An exception lies with mimetic peptides, which reproduce specific amino acid sequences in connexin or pannexin primary protein structure. In this paper, a state-of-the-art overview is provided on inhibitors of cellular channels consisting of connexins and pannexins with specific focus on their mode-of-action and therapeutic potential.
Collapse
Affiliation(s)
- Joost Willebrords
- Department of In Vitro Toxicology and Dermato-cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, Belgium
| | - Michaël Maes
- Department of In Vitro Toxicology and Dermato-cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, Belgium
| | - Sara Crespo Yanguas
- Department of In Vitro Toxicology and Dermato-cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, Belgium
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, Belgium.
| |
Collapse
|
34
|
Sáez PJ, Vargas P, Shoji KF, Harcha PA, Lennon-Duménil AM, Sáez JC. ATP promotes the fast migration of dendritic cells through the activity of pannexin 1 channels and P2X 7 receptors. Sci Signal 2017; 10:10/506/eaah7107. [PMID: 29162744 DOI: 10.1126/scisignal.aah7107] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Upon its release from injured cells, such as infected, transformed, inflamed, or necrotic cells, extracellular adenosine-5'-triphosphate (ATP) acts as a danger signal that recruits phagocytes, such as neutrophils, macrophages, and dendritic cells (DCs), to the site of injury. The sensing of extracellular ATP occurs through purinergic (P2) receptors. We investigated the cellular mechanisms linking purinergic signaling to DC motility. We found that ATP stimulated fast DC motility through an autocrine signaling loop, which was initiated by the activation of P2X7 receptors and further amplified by pannexin 1 (Panx1) channels. Upon stimulation of the P2X7 receptor by ATP, Panx1 contributed to fast DC motility by increasing the permeability of the plasma membrane, which resulted in supplementary ATP release. In the absence of Panx1, DCs failed to increase their speed of migration in response to ATP, despite exhibiting a normal P2X7 receptor-mediated Ca2+ response. In addition to DC migration, Panx1 channel- and P2X7 receptor-dependent signaling was further required to stimulate the reorganization of the actin cytoskeleton. In vivo, functional Panx1 channels were required for the homing of DCs to lymph nodes, although they were dispensable for DC maturation. These data suggest that P2X7 receptors and Panx1 channels are crucial players in the regulation of DC migration to endogenous danger signals.
Collapse
Affiliation(s)
- Pablo J Sáez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 6513677, Chile. .,INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 12 Rue Lhomond, Paris 75005, France
| | - Pablo Vargas
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 12 Rue Lhomond, Paris 75005, France.,CNRS UMR144, Institut Curie, PSL Research University, 12 Rue Lhomond, Paris 75005, France
| | - Kenji F Shoji
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 6513677, Chile
| | - Paloma A Harcha
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 6513677, Chile.,Instituto Milenio, Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso 2360103, Chile
| | - Ana-María Lennon-Duménil
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 12 Rue Lhomond, Paris 75005, France.
| | - Juan C Sáez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 6513677, Chile. .,Instituto Milenio, Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso 2360103, Chile
| |
Collapse
|
35
|
Pannexin-1 channels in epilepsy. Neurosci Lett 2017; 695:71-75. [PMID: 28886985 DOI: 10.1016/j.neulet.2017.09.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 07/20/2017] [Accepted: 09/01/2017] [Indexed: 02/07/2023]
Abstract
Pannexin-1 (Panx1) expression is raised in several animal seizure models and in resected human epileptic brain tissue, suggesting relevance to epilepsy. Multiple factors that are characteristic of seizures are thought to regulate Panx1 channel opening, including elevated levels of extracellular K+. Panx1, when open, 1) releases ATP, glutamate, and other metabolites into the extracellular medium, and 2) may depolarize the membrane due to a channel reversal potential around 0mV. Resultant ATP release from stimulated Panx1 can activate purinergic receptors, including P2X7 receptors. Glutamate and other signaling molecules released by Panx1 opening may have both excitatory and inhibitory actions on seizure generation. This review examines the critical and complex roles of Panx1 channels in epilepsy, which could provide a basis for future therapeutics.
Collapse
|
36
|
Valdebenito S, Barreto A, Eugenin EA. The role of connexin and pannexin containing channels in the innate and acquired immune response. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:154-165. [PMID: 28559189 DOI: 10.1016/j.bbamem.2017.05.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/17/2017] [Accepted: 05/25/2017] [Indexed: 12/20/2022]
Abstract
Connexin (Cx) and pannexin (Panx) containing channels - gap junctions (GJs) and hemichannels (HCs) - are present in virtually all cells and tissues. Currently, the role of these channels under physiological conditions is well defined. However, their role in the immune response and pathological conditions has only recently been explored. Data from several laboratories demonstrates that infectious agents, including HIV, have evolved to take advantage of GJs and HCs to improve viral/bacterial replication, enhance inflammation, and help spread toxicity into neighboring areas. In the current review, we discuss the role of Cx and Panx containing channels in immune activation and the pathogenesis of several infectious diseases. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Public Health Research Institute (PHRI), Newark, NJ, USA; Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA
| | - Andrea Barreto
- Public Health Research Institute (PHRI), Newark, NJ, USA; Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA
| | - Eliseo A Eugenin
- Public Health Research Institute (PHRI), Newark, NJ, USA; Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA.
| |
Collapse
|
37
|
Jiang LH, Mousawi F, Yang X, Roger S. ATP-induced Ca 2+-signalling mechanisms in the regulation of mesenchymal stem cell migration. Cell Mol Life Sci 2017; 74:3697-3710. [PMID: 28534085 PMCID: PMC5597679 DOI: 10.1007/s00018-017-2545-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/03/2017] [Accepted: 05/18/2017] [Indexed: 12/13/2022]
Abstract
The ability of cells to migrate to the destined tissues or lesions is crucial for physiological processes from tissue morphogenesis, homeostasis and immune responses, and also for stem cell-based regenerative medicines. Cytosolic Ca2+ is a primary second messenger in the control and regulation of a wide range of cell functions including cell migration. Extracellular ATP, together with the cognate receptors on the cell surface, ligand-gated ion channel P2X receptors and a subset of G-protein-coupled P2Y receptors, represents common autocrine and/or paracrine Ca2+ signalling mechanisms. The P2X receptor ion channels mediate extracellular Ca2+ influx, whereas stimulation of the P2Y receptors triggers intracellular Ca2+ release from the endoplasmic reticulum (ER), and activation of both type of receptors thus can elevate the cytosolic Ca2+ concentration ([Ca2+]c), albeit with different kinetics and capacity. Reduction in the ER Ca2+ level following the P2Y receptor activation can further induce store-operated Ca2+ entry as a distinct Ca2+ influx pathway that contributes in ATP-induced increase in the [Ca2+]c. Mesenchymal stem cells (MSC) are a group of multipotent stem cells that grow from adult tissues and hold promising applications in tissue engineering and cell-based therapies treating a great and diverse number of diseases. There is increasing evidence to show constitutive or evoked ATP release from stem cells themselves or mature cells in the close vicinity. In this review, we discuss the mechanisms for ATP release and clearance, the receptors and ion channels participating in ATP-induced Ca2+ signalling and the roles of such signalling mechanisms in mediating ATP-induced regulation of MSC migration.
Collapse
Affiliation(s)
- Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK. .,Sino-UK Joint Laboratory of Brain Function and Injury, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China. .,Inserm UMR1069, Nutrition, Croissance et Cancer, Université François-Rabelais de Tours, 37032, Tours, France.
| | - Fatema Mousawi
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| | - Xuebin Yang
- Department of Oral Biology, University of Leeds, WTBB, St James University Hospital, Leeds, LS97TF, UK
| | - Sėbastien Roger
- Inserm UMR1069, Nutrition, Croissance et Cancer, Université François-Rabelais de Tours, 37032, Tours, France
| |
Collapse
|