1
|
Poudel N, Zheng S, Skrypnyk N, Sung SSJ, Goggins E, Nash WT, Pavelec C, Yee M, Balogun I, Medina CB, Yao J, Rosin DL, Leitinger N, Ravichandran KS, Okusa MD. Proximal tubule pannexin 1 contributes to mitochondrial dysfunction and cell death during acute kidney injury. Am J Physiol Renal Physiol 2025; 328:F830-F849. [PMID: 40241514 DOI: 10.1152/ajprenal.00226.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/04/2024] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Pannexin 1 (Panx1) is a membrane-associated channel that, when activated, facilitates the release of small metabolites into the extracellular environment. These metabolites signal as damage-associated molecular patterns (DAMP) and initiate inflammation. Upregulation and activation of Panx1 is one of the early events during inflammatory injury. Animal models show that a lack of Panx1 is protective against acute kidney injury (AKI). How Panx1 modulates AKI is poorly understood. We utilized both in vivo and in vitro models of PANX1 overexpression to study mitochondrial function, cell death, and inflammation to evaluate how Panx1 contributes to AKI. We used two models of AKI, ischemia-reperfusion injury (IRI) and cisplatin-induced AKI (cis-AKI), in animals that overexpress PANX1 globally or specifically in the proximal tubule or in the endothelium. Cisplatin-induced injury was investigated in vitro in PANX1-overexpressing proximal tubule cells in culture. Both global and proximal tubule-specific overexpression of PANX1 exacerbated AKI, whereas endothelium-specific overexpression had no effect. Panx1-dependent metabolite release and alterations in the intracellular compartment in proximal tubules independently contributed to cell death in vitro. PANX1 overexpression impaired mitochondrial function and increased mitochondrial reactive oxygen species (ROS) production. PANX1 overexpression resulted in increased inflammation in the kidneys during cis-AKI. We showed that PANX1 overexpression resulted in overt renal injury during AKI that is in part mediated by reduced mitochondrial function, increased cell death, and inflammation. Selective strategies to inhibit Panx1 could help prevent or treat AKI.NEW & NOTEWORTHY Despite the huge medical, economical, and quality of life burden that AKI poses to patients, there are no Food and Drug Administration (FDA)-approved therapeutic or pharmaceutical interventions for AKI. Pannexin 1 (Panx1), which is upregulated in patients with AKI as well as in animals that develop experimental AKI, plays a crucial role in mediating both inflammation and cell death during AKI. Our findings suggest clinical interventions with molecules that inhibit Panx1 channel activity could improve outcomes in AKI patients.
Collapse
Affiliation(s)
- Nabin Poudel
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Shuqiu Zheng
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Nataliya Skrypnyk
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Sun-Sang J Sung
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Eibhlin Goggins
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - William T Nash
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Caitlin Pavelec
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, United States
| | - Marissa Yee
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Ishaq Balogun
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Cristopher B Medina
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States
| | - Junlan Yao
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Diane L Rosin
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, United States
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, United States
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|
2
|
Henze E, Burkhardt RN, Fox BW, Schwertfeger TJ, Gelsleichter E, Michalski K, Kramer L, Lenfest M, Boesch JM, Lin H, Schroeder FC, Kawate T. ATP-release pannexin channels are gated by lysophospholipids. eLife 2025; 14:RP107067. [PMID: 40309905 PMCID: PMC12045621 DOI: 10.7554/elife.107067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
In addition to its role as cellular energy currency, adenosine triphosphate (ATP) serves as an extracellular messenger that mediates diverse cell-to-cell communication. Compelling evidence supports that ATP is released from cells through pannexins, a family of membrane proteins that form heptameric large-pore channels. However, the activation mechanisms that trigger ATP release by pannexins remain poorly understood. Here, we discover lysophospholipids as endogenous pannexin activators, using activity-guided fractionation of mouse tissue extracts combined with untargeted metabolomics and electrophysiology. We show that lysophospholipids directly and reversibly activate pannexins in the absence of other proteins. Secretomics experiments reveal that lysophospholipid-activated pannexin 1 leads to the release of not only ATP but also other signaling metabolites, such as 5'-methylthioadenosine, which is important for immunomodulation. We also demonstrate that lysophospholipids activate endogenous pannexin 1 in human monocytes, leading to the release of IL-1β through inflammasome activation. Our results provide a connection between lipid metabolism and purinergic signaling, both of which play major roles in immune responses.
Collapse
Affiliation(s)
- Erik Henze
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| | - Russell N Burkhardt
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Bennett William Fox
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Tyler J Schwertfeger
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Eric Gelsleichter
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Kevin Michalski
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| | - Lydia Kramer
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| | - Margret Lenfest
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Jordyn M Boesch
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
- Department of Molecular Biology and Genetics, Cornell UniversityIthacaUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Frank C Schroeder
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| |
Collapse
|
3
|
Cassavaugh J, Longhi MS, Robson SC. Impact of Estrogen on Purinergic Signaling in Microvascular Disease. Int J Mol Sci 2025; 26:2105. [PMID: 40076726 PMCID: PMC11900469 DOI: 10.3390/ijms26052105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Microvascular ischemia, especially in the heart and kidneys, is associated with inflammation and metabolic perturbation, resulting in cellular dysfunction and end-organ failure. Heightened production of adenosine from extracellular nucleotides released in response to inflammation results in protective effects, inclusive of adaptations to hypoxia, endothelial cell nitric oxide release with the regulation of vascular tone, and inhibition of platelet aggregation. Purinergic signaling is modulated by ectonucleoside triphosphate diphosphohydrolase-1 (NTPDase1)/CD39, which is the dominant factor dictating vascular metabolism of extracellular ATP to adenosine throughout the cardiovascular tissues. Excess levels of extracellular purine metabolites, however, have been associated with metabolic and cardiovascular diseases. Physiological estrogen signaling is anti-inflammatory with vascular protective effects, but pharmacological replacement use in transgender and postmenopausal individuals is associated with thrombosis and other side effects. Crucially, the loss of this important sex hormone following menopause or with gender reassignment is associated with worsened pro-inflammatory states linked to increased oxidative stress, myocardial fibrosis, and, ultimately, diastolic dysfunction, also known as Yentl syndrome. While there is a growing body of knowledge on distinctive purinergic or estrogen signaling and endothelial health, much less is known about the relationships between the two signaling pathways. Continued studies of the interactions between these pathways will allow further insight into future therapeutic targets to improve the cardiovascular health of aging women without imparting deleterious side effects.
Collapse
Affiliation(s)
- Jessica Cassavaugh
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (M.S.L.); (S.C.R.)
| | | | | |
Collapse
|
4
|
Henze E, Burkhardt RN, Fox BW, Schwertfeger TJ, Gelsleichter E, Michalski K, Kramer L, Lenfest M, Boesch JM, Lin H, Schroeder FC, Kawate T. ATP-release pannexin channels are gated by lysophospholipids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.23.563601. [PMID: 37961151 PMCID: PMC10634739 DOI: 10.1101/2023.10.23.563601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In addition to its role as cellular energy currency, adenosine triphosphate (ATP) serves as an extracellular messenger that mediates diverse cell-to-cell communication. Compelling evidence supports that ATP is released from cells through pannexins, a family of membrane proteins that form heptameric large-pore channels. However, the activation mechanisms that trigger ATP release by pannexins remain poorly understood. Here, we discover lysophospholipids as endogenous pannexin activators, using activity-guided fractionation of mouse tissue extracts combined with untargeted metabolomics and electrophysiology. We show that lysophospholipids directly and reversibly activate pannexins in the absence of other proteins. Secretomics experiments reveal that lysophospholipid-activated pannexin 1 leads to the release of not only ATP but also other signaling metabolites, such as 5'-methylthioadenosine, which is important for immunomodulation. We also demonstrate that lysophospholipids activate endogenous pannexin 1 in human monocytes, leading to the release of IL-1β through inflammasome activation. Our results provide a connection between lipid metabolism and purinergic signaling, both of which play major roles in immune responses.
Collapse
|
5
|
Hsueh W, Wu Y, Weng M, Liu S, Santavanond JP, Liu Y, Lin C, Lai C, Lu Y, Hsu JY, Gao H, Lee J, Wei S, Lyu P, Poon IKH, Hsieh H, Chiu Y. Novel Naphthyridones Targeting Pannexin 1 for Colitis Management. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411538. [PMID: 39739600 PMCID: PMC11831487 DOI: 10.1002/advs.202411538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/29/2024] [Indexed: 01/02/2025]
Abstract
Pannexin 1 (PANX1) forms cell-surface channels capable of releasing signaling metabolites for diverse patho-physiological processes. While inhibiting dysregulated PANX1 has been proposed as a therapeutic strategy for many pathological conditions, including inflammatory bowel disease (IBD), low efficacy, or poor specificity of classical PANX1 inhibitors introduces uncertainty for their applications in basic and translational research. Here, hit-to-lead optimization is performed and a naphthyridone, compound 12, is identified as a new PANX1 inhibitor with an IC50 of 0.73 µm that does not affect pannexin-homologous LRRC8/SWELL1 channels. Using structure-activity relationship analysis, mutagenesis, cell thermal shift assays, and molecular docking, it is revealed that compound 12 directly engages PANX1 Trp74 residue. Using a dextran sodium sulfate mouse model of IBD, it is found that compound 12 markedly reduced colitis severity, highlighting new PANX1 inhibitors as a proof-of-concept treatment for IBD. These data describe the mechanism of action for a new PANX1 inhibitor, uncover the binding site for future drug design, and present a targeted strategy for treating IBD.
Collapse
Affiliation(s)
- Wen‐Yun Hsueh
- Department of ChemistryNational Tsing Hua UniversityHsinchu300044Taiwan
- Institute of Biotechnology and Pharmaceutical ResearchNational Health Research InstitutesMiaoli County350401Taiwan
| | - Yi‐Ling Wu
- Institute of BiotechnologyNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Meng‐Tzu Weng
- Department of Internal MedicineNational Taiwan University HospitalTaipei City100229Taiwan
- Department of Medical ResearchNational Taiwan University HospitalHsin‐Chu BranchHsinchu302058Taiwan
| | - Shin‐Yun Liu
- Department of Internal MedicineNational Taiwan University HospitalTaipei City100229Taiwan
| | - Jascinta P Santavanond
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular ScienceLa Trobe UniversityMelbourneVIC3086Australia
- Research Centre for Extracellular VesiclesLa Trobe UniversityVIC3086Australia
| | - Yi‐Chung Liu
- Institute of Population Health SciencesNational Health Research InstitutesMiaoli County350401Taiwan
| | - Ching‐I Lin
- Department of Internal MedicineNational Taiwan University HospitalTaipei City100229Taiwan
| | - Cheng‐Nong Lai
- Institute of BiotechnologyNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Yi‐Ru Lu
- Institute of BiotechnologyNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Jing Yin Hsu
- Institute of BiotechnologyNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Hong‐Yu Gao
- Institute of BiotechnologyNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Jinq‐Chyi Lee
- Institute of Biotechnology and Pharmaceutical ResearchNational Health Research InstitutesMiaoli County350401Taiwan
| | - Shu‐Chen Wei
- Department of Internal MedicineNational Taiwan University HospitalTaipei City100229Taiwan
| | - Ping‐Chiang Lyu
- Institute of Bioinformatics and Structural BiologyNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Ivan K H Poon
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular ScienceLa Trobe UniversityMelbourneVIC3086Australia
- Research Centre for Extracellular VesiclesLa Trobe UniversityVIC3086Australia
| | - Hsing‐Pang Hsieh
- Department of ChemistryNational Tsing Hua UniversityHsinchu300044Taiwan
- Institute of Biotechnology and Pharmaceutical ResearchNational Health Research InstitutesMiaoli County350401Taiwan
| | - Yu‐Hsin Chiu
- Institute of BiotechnologyNational Tsing Hua UniversityHsinchu300044Taiwan
- Departments of Medical Science, Life Science, and MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| |
Collapse
|
6
|
Gebert JT, Scribano FJ, Engevik KA, Huleatt EM, Eledge MR, Dorn LE, Philip AA, Kawagishi T, Greenberg HB, Patton JT, Hyser JM. Viroporin activity is necessary for intercellular calcium signals that contribute to viral pathogenesis. SCIENCE ADVANCES 2025; 11:eadq8115. [PMID: 39823322 PMCID: PMC11740935 DOI: 10.1126/sciadv.adq8115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025]
Abstract
Viruses engage in a variety of processes to subvert host defenses and create an environment amenable to replication. Here, using rotavirus as a prototype, we show that calcium conductance out of the endoplasmic reticulum by the virus encoded ion channel, NSP4, induces intercellular calcium waves that extend beyond the infected cell and contribute to pathogenesis. Viruses that lack the ability to induce this signaling show diminished viral shedding and attenuated disease in a mouse model of rotavirus diarrhea. This implicates nonstructural protein 4 (NSP4) as a virulence factor and provides mechanistic insight into its mode of action. Critically, this signaling induces a transcriptional signature characteristic of interferon-independent innate immune activation, which is not observed in response to a mutant NSP4 that does not conduct calcium. This implicates calcium dysregulation as a means of pathogen recognition, a theme broadly applicable to calcium-altering pathogens beyond rotavirus.
Collapse
Affiliation(s)
- J. Thomas Gebert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Francesca J. Scribano
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kristen A. Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ethan M. Huleatt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael R. Eledge
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lauren E. Dorn
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Asha A. Philip
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Takahiro Kawagishi
- Departments of Medicine and Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Harry B. Greenberg
- Departments of Medicine and Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John T. Patton
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Joseph M. Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
7
|
Tsuyama T, Teramura R, Mitsuoka K, Kishikawa JI, Yokoyama K. Cryo-EM structure of the human Pannexin-3 channel. Biochem Biophys Res Commun 2025; 745:151227. [PMID: 39721314 DOI: 10.1016/j.bbrc.2024.151227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Pannexin-3 (PANX3) is a member of the pannexin family of large-pore, ATP-permeable channels conserved across vertebrates. PANX3 contributes to various developmental and pathophysiological processes by permeating ATP and Ca2+ ions; however, the structural basis of PANX3 channel function remains unclear. Here, we present the cryo-EM structure of human PANX3 at 2.9-3.2 Å. The PANX3 channel is heptameric and forms a transmembrane pore along the central symmetric axis. The narrowest constriction of the pore is composed of an isoleucine ring located in the extracellular region, and its size is comparable to that of other pannexins. A structural variability analysis revealed prominent structural dynamics in intracellular regions. Our structural studies provide a foundation for understanding the detailed properties of pannexin channels.
Collapse
Affiliation(s)
- Taiichi Tsuyama
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kita-ku, Kyoto, 603-8555, Japan
| | - Ryuga Teramura
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kita-ku, Kyoto, 603-8555, Japan
| | - Kaoru Mitsuoka
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Osaka, Japan
| | - Jun-Ichi Kishikawa
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kita-ku, Kyoto, 603-8555, Japan; Department of Applied Biology, Graduate School of Science and Technology, Kyoto Institute of Technology, Kyoto, Japan
| | - Ken Yokoyama
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kita-ku, Kyoto, 603-8555, Japan.
| |
Collapse
|
8
|
Luo J, Zhou Y, Wang M, Zhang J, Jiang E. Inflammasomes: potential therapeutic targets in hematopoietic stem cell transplantation. Cell Commun Signal 2024; 22:596. [PMID: 39695742 DOI: 10.1186/s12964-024-01974-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/30/2024] [Indexed: 12/20/2024] Open
Abstract
The realm of hematopoietic stem cell transplantation (HSCT) has witnessed remarkable advancements in elevating the cure and survival rates for patients with both malignant and non-malignant hematologic diseases. Nevertheless, a considerable number of patients continue to face challenges, including transplant-related complications, infection, graft failure, and mortality. Inflammasomes, the multi-protein complexes of the innate immune system, respond to various danger signals by releasing inflammatory cytokines and even mediating cell death. While moderate activation of inflammasomes is essential for immune defense and homeostasis maintenance, excessive activation precipitates inflammatory damage. The intricate interplay between HSCT and inflammasomes arises from their pivotal roles in immune responses and inflammation. This review examines the molecular architecture and composition of various types of inflammasomes, highlighting their activation and effector mechanisms within the context of the HSCT process and its associated complications. Additionally, we summarize the therapeutic implications of targeting inflammasomes and related factors in HSCT.
Collapse
Affiliation(s)
- Jieya Luo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yunxia Zhou
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Haihe Laboratory of Cell Ecosystem, Tianjin Medical University, Tianjin, 300051, China
| | - Mingyang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Junan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
9
|
Henze E, Ehrlich JJ, Robertson JL, Gelsleichter E, Kawate T. The C-terminal activating domain promotes pannexin 1 channel opening. Proc Natl Acad Sci U S A 2024; 121:e2411898121. [PMID: 39671183 DOI: 10.1073/pnas.2411898121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
Pannexin 1 (Panx1) constitutes a large pore channel responsible for the release of adenosine triphosphate (ATP) from apoptotic cells. Strong evidence indicates that caspase-mediated cleavage of the C-terminus promotes the opening of the Panx1 channel by unplugging the pore. However, this simple pore-plugging mechanism alone cannot account for the observation that a Panx1 construct ending before the caspase cleavage site remains closed. Here, we show that a helical region located immediately before the caspase cleavage site, referred to as the "C-terminal activating domain (CAD)", plays a pivotal role in facilitating Panx1 activation. Electrophysiology and mutagenesis studies uncovered that two conserved leucine residues within the CAD play a pivotal role. Cryoelectron microscopy (Cryo-EM) analysis of the construct ending before reaching the CAD demonstrated that the N terminus extends into an intracellular pocket. In contrast, the construct including the CAD revealed that this domain occupies the intracellular pocket, causing the N terminus to flip upward within the pore. Analysis of electrostatic free energy landscape in the closed conformation indicated that the intracellular side of the ion permeation pore may be occupied by anions like ATP, creating an electrostatic barrier for anions attempting to permeate the pore. When the N terminus flips up, it diminishes the positively charged surface, thereby reducing the drive to accumulate anions inside the pore. This dynamic change in the electrostatic landscape likely contributes to the selection of permeant ions. Collectively, these experiments put forth a mechanism in which C-terminal cleavage liberates the CAD, causing the repositioning of the N terminus to promote Panx1 channel opening.
Collapse
Affiliation(s)
- Erik Henze
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | | | - Janice L Robertson
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Eric Gelsleichter
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| |
Collapse
|
10
|
Abstract
Cardiovascular disease is the leading cause of death worldwide, and it commonly results from atherosclerotic plaque progression. One of the increasingly recognized drivers of atherosclerosis is dysfunctional efferocytosis, a homeostatic mechanism responsible for the clearance of dead cells and the resolution of inflammation. In atherosclerosis, the capacity of phagocytes to participate in efferocytosis is hampered, leading to the accumulation of apoptotic and necrotic tissue within the plaque, which results in enlargement of the necrotic core, increased luminal stenosis and plaque inflammation, and predisposition to plaque rupture or erosion. In this Review, we describe the different forms of programmed cell death that can occur in the atherosclerotic plaque and highlight the efferocytic machinery that is normally implicated in cardiovascular physiology. We then discuss the mechanisms by which efferocytosis fails in atherosclerosis and other cardiovascular and cardiometabolic diseases, including myocardial infarction and diabetes mellitus, and discuss therapeutic approaches that might reverse this pathological process.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
| |
Collapse
|
11
|
Casillas Martinez A, Wicki-Stordeur LE, Ariano AV, Swayne LA. Dual role for pannexin 1 at synapses: regulating functional and morphological plasticity. J Physiol 2024. [PMID: 39264228 DOI: 10.1113/jp285228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
Pannexin 1 (PANX1) is an ion and metabolite membrane channel and scaffold protein enriched in synaptic compartments of neurons in the central nervous system. In addition to a well-established link between PANX1 and synaptic plasticity, we recently identified a role for PANX1 in the regulation of dendritic spine stability. Notably, PANX1 and its interacting proteins are linked to neurological conditions involving dendritic spine loss. Understanding the dual role of PANX1 in synaptic function and morphology may help to shed light on these links. We explore potential mechanisms, including PANX1's interactions with postsynaptic receptors and cytoskeleton regulating proteins. Finally, we contextualize PANX1's dual role within neurological diseases involving dendritic spine and synapse dysfunction.
Collapse
Affiliation(s)
| | - Leigh E Wicki-Stordeur
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Annika V Ariano
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
12
|
Pavelec CM, Young AP, Luviano HL, Orrell EE, Szagdaj A, Poudel N, Wolpe AG, Thomas SH, Yeudall S, Upchurch CM, Okusa MD, Isakson BE, Wolf MJ, Leitinger N. Cardiomyocyte PANX1 Controls Glycolysis and Neutrophil Recruitment in Hypertrophy. Circ Res 2024; 135:503-517. [PMID: 38957990 PMCID: PMC11293983 DOI: 10.1161/circresaha.124.324650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND PANX1 (pannexin 1), a ubiquitously expressed ATP release membrane channel, has been shown to play a role in inflammation, blood pressure regulation, and myocardial infarction. However, the possible role of PANX1 in cardiomyocytes in the progression of heart failure has not yet been investigated. METHOD We generated a novel mouse line with constitutive deletion of PANX1 in cardiomyocytes (Panx1MyHC6). RESULTS PANX1 deletion in cardiomyocytes had no effect on unstressed heart function but increased the glycolytic metabolism and resulting glycolytic ATP production, with a concurrent decrease in oxidative phosphorylation, both in vivo and in vitro. In vitro, treatment of H9c2 (H9c2 rat myoblast cell line) cardiomyocytes with isoproterenol led to PANX1-dependent release of ATP and Yo-Pro-1 uptake, as assessed by pharmacological blockade with spironolactone and siRNA-mediated knockdown of PANX1. To investigate nonischemic heart failure and the preceding cardiac hypertrophy, we administered isoproterenol, and we demonstrated that Panx1MyHC6 mice were protected from systolic and diastolic left ventricle volume increases as a result of cardiomyocyte hypertrophy. Moreover, we found that Panx1MyHC6 mice showed decreased isoproterenol-induced recruitment of immune cells (CD45+), particularly neutrophils (CD11b+ [integrin subunit alpha M], Ly6g+ [lymphocyte antigen 6 family member G]), to the myocardium. CONCLUSIONS Together, these data demonstrate that PANX1 deficiency in cardiomyocytes increases glycolytic metabolism and protects against cardiac hypertrophy in nonischemic heart failure at least in part by reducing immune cell recruitment. Our study implies PANX1 channel inhibition as a therapeutic approach to ameliorate cardiac dysfunction in patients with heart failure.
Collapse
Affiliation(s)
- Caitlin M Pavelec
- Department of Pharmacology (C.M.P., H.L.L., E.E.O., A.S., S.H.T., S.Y., C.M.U., N.L.), University of Virginia School of Medicine, Charlottesville
- Robert M. Berne Cardiovascular Research Center (C.M.P., A.P.Y., B.E.I., M.J.W., N.L.), University of Virginia School of Medicine, Charlottesville
| | - Alexander P Young
- Cardiovascular Medicine, Department of Medicine (A.P.Y., M.J.W.), University of Virginia School of Medicine, Charlottesville
- Robert M. Berne Cardiovascular Research Center (C.M.P., A.P.Y., B.E.I., M.J.W., N.L.), University of Virginia School of Medicine, Charlottesville
| | - Hannah L Luviano
- Department of Pharmacology (C.M.P., H.L.L., E.E.O., A.S., S.H.T., S.Y., C.M.U., N.L.), University of Virginia School of Medicine, Charlottesville
| | - Emily E Orrell
- Department of Pharmacology (C.M.P., H.L.L., E.E.O., A.S., S.H.T., S.Y., C.M.U., N.L.), University of Virginia School of Medicine, Charlottesville
| | - Anna Szagdaj
- Department of Pharmacology (C.M.P., H.L.L., E.E.O., A.S., S.H.T., S.Y., C.M.U., N.L.), University of Virginia School of Medicine, Charlottesville
| | - Nabin Poudel
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine (N.P., M.D.O.), University of Virginia School of Medicine, Charlottesville
| | - Abigail G Wolpe
- Department of Cell Biology (A.G.W.), University of Virginia School of Medicine, Charlottesville
| | - Samantha H Thomas
- Department of Pharmacology (C.M.P., H.L.L., E.E.O., A.S., S.H.T., S.Y., C.M.U., N.L.), University of Virginia School of Medicine, Charlottesville
| | - Scott Yeudall
- Department of Pharmacology (C.M.P., H.L.L., E.E.O., A.S., S.H.T., S.Y., C.M.U., N.L.), University of Virginia School of Medicine, Charlottesville
| | - Clint M Upchurch
- Department of Pharmacology (C.M.P., H.L.L., E.E.O., A.S., S.H.T., S.Y., C.M.U., N.L.), University of Virginia School of Medicine, Charlottesville
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine (N.P., M.D.O.), University of Virginia School of Medicine, Charlottesville
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center (C.M.P., A.P.Y., B.E.I., M.J.W., N.L.), University of Virginia School of Medicine, Charlottesville
- Department of Molecular Physiology and Biological Physics (B.E.I.), University of Virginia School of Medicine, Charlottesville
| | - Matthew J Wolf
- Cardiovascular Medicine, Department of Medicine (A.P.Y., M.J.W.), University of Virginia School of Medicine, Charlottesville
- Robert M. Berne Cardiovascular Research Center (C.M.P., A.P.Y., B.E.I., M.J.W., N.L.), University of Virginia School of Medicine, Charlottesville
| | - Norbert Leitinger
- Department of Pharmacology (C.M.P., H.L.L., E.E.O., A.S., S.H.T., S.Y., C.M.U., N.L.), University of Virginia School of Medicine, Charlottesville
- Robert M. Berne Cardiovascular Research Center (C.M.P., A.P.Y., B.E.I., M.J.W., N.L.), University of Virginia School of Medicine, Charlottesville
| |
Collapse
|
13
|
Vardam-Kaur T, Banuelos A, Gabaldon-Parish M, Macedo BG, Salgado CL, Wanhainen KM, Zhou MH, van Dijk S, Santiago-Carvalho I, Beniwal AS, Leff CL, Peng C, Tran NL, Jameson SC, Borges da Silva H. The ATP-exporting channel Pannexin 1 promotes CD8 + T cell effector and memory responses. iScience 2024; 27:110290. [PMID: 39045105 PMCID: PMC11263643 DOI: 10.1016/j.isci.2024.110290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/17/2024] [Accepted: 06/14/2024] [Indexed: 07/25/2024] Open
Abstract
Sensing of extracellular ATP (eATP) controls CD8+ T cell function. Their accumulation can occur through export by specialized molecules, such as the release channel Pannexin 1 (Panx1). Whether Panx1 controls CD8+ T cell immune responses in vivo, however, has not been previously addressed. Here, we report that T-cell-specific Panx1 is needed for CD8+ T cell responses to viral infections and cancer. We found that CD8-specific Panx1 promotes both effector and memory CD8+ T cell responses. Panx1 favors initial effector CD8+ T cell activation through extracellular ATP (eATP) export and subsequent P2RX4 activation, which helps promote full effector differentiation through extracellular lactate accumulation and its subsequent recycling. In contrast, Panx1 promotes memory CD8+ T cell survival primarily through ATP export and subsequent P2RX7 engagement, leading to improved mitochondrial metabolism. In summary, Panx1-mediated eATP export regulates effector and memory CD8+ T cells through distinct purinergic receptors and different metabolic and signaling pathways.
Collapse
Affiliation(s)
- Trupti Vardam-Kaur
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
| | - Alma Banuelos
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
| | | | - Bruna Gois Macedo
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
| | | | | | - Maggie Hanqi Zhou
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
| | - Sarah van Dijk
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
| | | | - Angad S. Beniwal
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
| | - Chloe L. Leff
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
| | - Changwei Peng
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nhan L. Tran
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
| | - Stephen C. Jameson
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Henrique Borges da Silva
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, AZ 85255, USA
| |
Collapse
|
14
|
Qiao LY. Satellite Glial Cells Bridge Sensory Neuron Crosstalk in Visceral Pain and Cross-Organ Sensitization. J Pharmacol Exp Ther 2024; 390:213-221. [PMID: 38777604 PMCID: PMC11264254 DOI: 10.1124/jpet.123.002061] [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: 12/15/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Following colonic inflammation, the uninjured bladder afferent neurons are also activated. The mechanisms and pathways underlying this sensory neuron cross-activation (from injured neurons to uninjured neurons) are not fully understood. Colonic and bladder afferent neurons reside in the same spinal segments and are separated by satellite glial cells (SGCs) and extracellular matrix in dorsal root ganglia (DRG). SGCs communicate with sensory neurons in a bidirectional fashion. This review summarizes the differentially regulated genes/proteins in the injured and uninjured DRG neurons and explores the role of SGCs in regulation of sensory neuron crosstalk in visceral cross-organ sensitization. The review also highlights the paracrine pathways in mediating neuron-SGC and SGC-neuron coupling with an emphasis on the neurotrophins and purinergic systems. Finally, I discuss the results from recent RNAseq profiling of SGCs to reveal useful molecular markers for characterization, functional study, and therapeutic targets of SGCs. SIGNIFICANCE STATEMENT: Satellite glial cells (SGCs) are the largest glial subtypes in sensory ganglia and play a critical role in mediating sensory neuron crosstalk, an underlying mechanism in colon-bladder cross-sensitization. Identification of novel and unique molecular markers of SGCs can advance the discovery of therapeutic targets in treatment of chronic pain including visceral pain comorbidity.
Collapse
Affiliation(s)
- Liya Y Qiao
- Department of Physiology and Biophysics, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
15
|
Henze E, Ehrlich JJ, Robertson JL, Kawate T. The C-terminal activating domain promotes Panx1 channel opening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598903. [PMID: 38915727 PMCID: PMC11195165 DOI: 10.1101/2024.06.13.598903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Pannexin 1 (Panx1) constitutes a large pore channel responsible for the release of ATP from apoptotic cells. Strong evidence indicates that caspase-mediated cleavage of the C-terminus promotes the opening of the Panx1 channel by unplugging the pore. However, this simple pore-plugging mechanism alone cannot account for the observation that a Panx1 construct ending before the caspase cleavage site remains closed. Here, we show that a helical region located immediately before the caspase cleavage site, referred to as the "C-terminal activating domain (CAD)," plays a pivotal role in facilitating Panx1 activation. Electrophysiology and mutagenesis studies uncovered that two conserved leucine residues within the CAD plays a pivotal role. Cryo-EM analysis of the construct ending before reaching the CAD demonstrated that the N-terminus extends into an intracellular pocket. In contrast, the construct including the CAD revealed that this domain occupies the intracellular pocket, causing the N-terminus to flip upward within the pore. Analysis of electrostatic free energy landscape in the closed conformation indicated that the intracellular side of the ion permeation pore may be occupied by anions like ATP, creating an electrostatic barrier for anions attempting to permeate the pore. When the N-terminus flips up, it diminishes the positively charged surface, thereby reducing the drive to accumulate anions inside the pore. This dynamic change in the electrostatic landscape likely contributes to the selection of permeant ions. Collectively, these experiments put forth a novel mechanism in which C-terminal cleavage liberates the CAD, causing the repositioning of the N-terminus to promote Panx1 channel opening.
Collapse
Affiliation(s)
- Erik Henze
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | - Janice L. Robertson
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
16
|
Aslan A, Ari Yuka S. Therapeutic peptides for coronary artery diseases: in silico methods and current perspectives. Amino Acids 2024; 56:37. [PMID: 38822212 PMCID: PMC11143054 DOI: 10.1007/s00726-024-03397-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/06/2024] [Indexed: 06/02/2024]
Abstract
Many drug formulations containing small active molecules are used for the treatment of coronary artery disease, which affects a significant part of the world's population. However, the inadequate profile of these molecules in terms of therapeutic efficacy has led to the therapeutic use of protein and peptide-based biomolecules with superior properties, such as target-specific affinity and low immunogenicity, in critical diseases. Protein‒protein interactions, as a consequence of advances in molecular techniques with strategies involving the combined use of in silico methods, have enabled the design of therapeutic peptides to reach an advanced dimension. In particular, with the advantages provided by protein/peptide structural modeling, molecular docking for the study of their interactions, molecular dynamics simulations for their interactions under physiological conditions and machine learning techniques that can work in combination with all these, significant progress has been made in approaches to developing therapeutic peptides that can modulate the development and progression of coronary artery diseases. In this scope, this review discusses in silico methods for the development of peptide therapeutics for the treatment of coronary artery disease and strategies for identifying the molecular mechanisms that can be modulated by these designs and provides a comprehensive perspective for future studies.
Collapse
Affiliation(s)
- Ayca Aslan
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Esenler, Istanbul, Turkey
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler, Istanbul, Turkey
| | - Selcen Ari Yuka
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Esenler, Istanbul, Turkey.
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler, Istanbul, Turkey.
| |
Collapse
|
17
|
Luo Y, Zheng S, Xiao W, Zhang H, Li Y. Pannexins in the musculoskeletal system: new targets for development and disease progression. Bone Res 2024; 12:26. [PMID: 38705887 PMCID: PMC11070431 DOI: 10.1038/s41413-024-00334-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/04/2024] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
During cell differentiation, growth, and development, cells can respond to extracellular stimuli through communication channels. Pannexin (Panx) family and connexin (Cx) family are two important types of channel-forming proteins. Panx family contains three members (Panx1-3) and is expressed widely in bone, cartilage and muscle. Although there is no sequence homology between Panx family and Cx family, they exhibit similar configurations and functions. Similar to Cxs, the key roles of Panxs in the maintenance of physiological functions of the musculoskeletal system and disease progression were gradually revealed later. Here, we seek to elucidate the structure of Panxs and their roles in regulating processes such as osteogenesis, chondrogenesis, and muscle growth. We also focus on the comparison between Cx and Panx. As a new key target, Panxs expression imbalance and dysfunction in muscle and the therapeutic potentials of Panxs in joint diseases are also discussed.
Collapse
Affiliation(s)
- Yan Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, 410008, China
| | - Shengyuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, 410008, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hang Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
18
|
Vardam-Kaur T, Banuelos A, Gabaldon-Parish M, Macedo BG, Salgado CL, Wanhainen KM, Zhou MH, van Dijk S, Santiago-Carvalho I, Beniwal AS, Leff CL, Peng C, Tran NL, Jameson SC, da Silva HB. The ATP-exporting channel Pannexin-1 promotes CD8 + T cell effector and memory responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.19.537580. [PMID: 37131831 PMCID: PMC10153284 DOI: 10.1101/2023.04.19.537580] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Sensing of extracellular ATP (eATP) controls CD8+ T cell function. Their accumulation can occur through export by specialized molecules, such as the release channel Pannexin-1 (Panx1). Whether Panx1 controls CD8+ T cell immune responses in vivo, however, has not been previously addressed. Here, we report that T cell-specific Panx1 is needed for CD8+ T cell responses to viral infections and cancer. We found that CD8-specific Panx1 promotes both effector and memory CD8+ T cell responses. Panx1 favors initial effector CD8+ T cell activation through extracellular ATP (eATP) export and subsequent P2RX4 activation, which helps promote full effector differentiation through extracellular lactate accumulation and its subsequent recycling. In contrast, Panx1 promotes memory CD8+ T cell survival primarily through ATP export and subsequent P2RX7 engagement, leading to improved mitochondrial metabolism. In summary, Panx1-mediated eATP export regulates effector and memory CD8+ T cells through distinct purinergic receptors and different metabolic and signaling pathways.
Collapse
Affiliation(s)
- Trupti Vardam-Kaur
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
- Current address: Omeros Corporation, Seattle, Washington, United States
| | - Alma Banuelos
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Maria Gabaldon-Parish
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
- Current address: University of New Mexico, Albuquerque, New Mexico, United States
| | - Bruna Gois Macedo
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | | | | | - Maggie Hanqi Zhou
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Sarah van Dijk
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
- Current address: Biomedical Sciences Graduate Program, University of California, San Diego, California, United States
| | | | - Angad S. Beniwal
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Chloe L. Leff
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Changwei Peng
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States
- Current address: Department of Immunology & HMS Center for Immune Imaging, Harvard Medical School, Boston, Massachusetts, United States
| | - Nhan L. Tran
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Stephen C. Jameson
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States
| | - Henrique Borges da Silva
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| |
Collapse
|
19
|
Pavelec CM, Young AP, Luviano HL, Orrell EE, Szagdaj A, Poudel N, Wolpe AG, Thomas SH, Yeudall S, Upchurch CM, Okusa MD, Isakson BE, Wolf MJ, Leitinger N. Pannexin 1 Channels Control Cardiomyocyte Metabolism and Neutrophil Recruitment During Non-Ischemic Heart Failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.29.573679. [PMID: 38234768 PMCID: PMC10793433 DOI: 10.1101/2023.12.29.573679] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Pannexin 1 (PANX1), a ubiquitously expressed ATP release membrane channel, has been shown to play a role in inflammation, blood pressure regulation, and myocardial infarction. However, a possible role of PANX1 in cardiomyocytes in the progression of heart failure has not yet been investigated. We generated a novel mouse line with constitutive deletion of PANX1 in cardiomyocytes (Panx1 MyHC6 ). PANX1 deletion in cardiomyocytes had no effect on unstressed heart function but increased the glycolytic metabolism both in vivo and in vitro . In vitro , treatment of H9c2 cardiomyocytes with isoproterenol led to PANX1-dependent release of ATP and Yo-Pro-1 uptake, as assessed by pharmacological blockade with spironolactone and siRNA-mediated knock-down of PANX1. To investigate non-ischemic heart failure and the preceding cardiac hypertrophy we administered isoproterenol, and we demonstrate that Panx1 MyHC6 mice were protected from systolic and diastolic left ventricle volume increases and cardiomyocyte hypertrophy. Moreover, we found that Panx1 MyHC6 mice showed decreased isoproterenol-induced recruitment of immune cells (CD45 + ), particularly neutrophils (CD11b + , Ly6g + ), to the myocardium. Together these data demonstrate that PANX1 deficiency in cardiomyocytes impacts glycolytic metabolism and protects against cardiac hypertrophy in non-ischemic heart failure at least in part by reducing immune cell recruitment. Our study implies PANX1 channel inhibition as a therapeutic approach to ameliorate cardiac dysfunction in heart failure patients.
Collapse
|
20
|
McAllister BB, Stokes-Heck S, Harding EK, van den Hoogen NJ, Trang T. Targeting Pannexin-1 Channels: Addressing the 'Gap' in Chronic Pain. CNS Drugs 2024; 38:77-91. [PMID: 38353876 DOI: 10.1007/s40263-024-01061-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/07/2024] [Indexed: 02/22/2024]
Abstract
Chronic pain complicates many diseases and is notoriously difficult to treat. In search of new therapeutic targets, pannexin-1 (Panx1) channels have sparked intense interest as a key mechanism involved in a variety of chronic pain conditions. Panx1 channels are transmembrane proteins that release ions and small molecules, such as adenosine triphosphate (ATP). They are expressed along important nodes of the pain pathway, modulating activity of diverse cell types implicated in the development and progression of chronic pain caused by injury or pathology. This review highlights advances that have unlocked the core structure and machinery controlling Panx1 function with a focus on understanding and treating chronic pain.
Collapse
Affiliation(s)
- Brendan B McAllister
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Sierra Stokes-Heck
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Erika K Harding
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Nynke J van den Hoogen
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Tuan Trang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
21
|
O'Donnell BL, Penuela S. Skin in the game: pannexin channels in healthy and cancerous skin. Biochem J 2023; 480:1929-1949. [PMID: 38038973 DOI: 10.1042/bcj20230176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
The skin is a highly organized tissue composed of multiple layers and cell types that require coordinated cell to cell communication to maintain tissue homeostasis. In skin cancer, this organized structure and communication is disrupted, prompting the malignant transformation of healthy cells into melanoma, basal cell carcinoma or squamous cell carcinoma tumours. One such family of channel proteins critical for cellular communication is pannexins (PANX1, PANX2, PANX3), all of which are present in the skin. These heptameric single-membrane channels act as conduits for small molecules and ions like ATP and Ca2+ but have also been shown to have channel-independent functions through their interacting partners or action in signalling pathways. Pannexins have diverse roles in the skin such as in skin development, aging, barrier function, keratinocyte differentiation, inflammation, and wound healing, which were discovered through work with pannexin knockout mice, organotypic epidermis models, primary cells, and immortalized cell lines. In the context of cutaneous cancer, PANX1 is present at high levels in melanoma tumours and functions in melanoma carcinogenesis, and both PANX1 and PANX3 expression is altered in non-melanoma skin cancer. PANX2 has thus far not been implicated in any skin cancer. This review will discuss pannexin isoforms, structure, trafficking, post-translational modifications, interactome, and channel activity. We will also outline the expression, localization, and function of pannexin channels within the diverse cell types of the epidermis, dermis, hypodermis, and adnexal structures of the skin, and how these properties are exploited or abrogated in instances of skin cancer.
Collapse
Affiliation(s)
- Brooke L O'Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
- Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| |
Collapse
|
22
|
Gregory CD. Hijacking homeostasis: Regulation of the tumor microenvironment by apoptosis. Immunol Rev 2023; 319:100-127. [PMID: 37553811 PMCID: PMC10952466 DOI: 10.1111/imr.13259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023]
Abstract
Cancers are genetically driven, rogue tissues which generate dysfunctional, obdurate organs by hijacking normal, homeostatic programs. Apoptosis is an evolutionarily conserved regulated cell death program and a profoundly important homeostatic mechanism that is common (alongside tumor cell proliferation) in actively growing cancers, as well as in tumors responding to cytotoxic anti-cancer therapies. Although well known for its cell-autonomous tumor-suppressive qualities, apoptosis harbors pro-oncogenic properties which are deployed through non-cell-autonomous mechanisms and which generally remain poorly defined. Here, the roles of apoptosis in tumor biology are reviewed, with particular focus on the secreted and fragmentation products of apoptotic tumor cells and their effects on tumor-associated macrophages, key supportive cells in the aberrant homeostasis of the tumor microenvironment. Historical aspects of cell loss in tumor growth kinetics are considered and the impact (and potential impact) on tumor growth of apoptotic-cell clearance (efferocytosis) as well as released soluble and extracellular vesicle-associated factors are discussed from the perspectives of inflammation, tissue repair, and regeneration programs. An "apoptosis-centric" view is proposed in which dying tumor cells provide an important platform for intricate intercellular communication networks in growing cancers. The perspective has implications for future research and for improving cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Christopher D. Gregory
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarterEdinburghUK
| |
Collapse
|
23
|
Lamouroux A, Tournier M, Iaculli D, Caufriez A, Rusiecka OM, Martin C, Bes V, Carpio LE, Girardin Y, Loris R, Tabernilla A, Molica F, Gozalbes R, Mayán MD, Vinken M, Kwak BR, Ballet S. Structure-Based Design and Synthesis of Stapled 10Panx1 Analogues for Use in Cardiovascular Inflammatory Diseases. J Med Chem 2023; 66:13086-13102. [PMID: 37703077 PMCID: PMC10544015 DOI: 10.1021/acs.jmedchem.3c01116] [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: 06/20/2023] [Indexed: 09/14/2023]
Abstract
Following a rational design, a series of macrocyclic ("stapled") peptidomimetics of 10Panx1, the most established peptide inhibitor of Pannexin1 (Panx1) channels, were developed and synthesized. Two macrocyclic analogues SBL-PX1-42 and SBL-PX1-44 outperformed the linear native peptide. During in vitro adenosine triphosphate (ATP) release and Yo-Pro-1 uptake assays in a Panx1-expressing tumor cell line, both compounds were revealed to be promising bidirectional inhibitors of Panx1 channel function, able to induce a two-fold inhibition, as compared to the native 10Panx1 sequence. The introduction of triazole-based cross-links within the peptide backbones increased helical content and enhanced in vitro proteolytic stability in human plasma (>30-fold longer half-lives, compared to 10Panx1). In adhesion assays, a "double-stapled" peptide, SBL-PX1-206 inhibited ATP release from endothelial cells, thereby efficiently reducing THP-1 monocyte adhesion to a TNF-α-activated endothelial monolayer and making it a promising candidate for future in vivo investigations in animal models of cardiovascular inflammatory disease.
Collapse
Affiliation(s)
- Arthur Lamouroux
- Research
Group of Organic Chemistry, Departments of Chemistry and Bioengineering
Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Malaury Tournier
- Department
of Pathology and Immunology and Geneva Center for Inflammation Research,
Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Debora Iaculli
- Research
Group of Organic Chemistry, Departments of Chemistry and Bioengineering
Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Anne Caufriez
- Research
Group of Organic Chemistry, Departments of Chemistry and Bioengineering
Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
- Research
Unit of In Vitro Toxicology and Dermato-Cosmetology, Department of
Pharmaceutical Sciences, Vrije Universiteit
Brussel, Laarbeeklaan
103, 1090 Brussels, Belgium
| | - Olga M. Rusiecka
- Department
of Pathology and Immunology and Geneva Center for Inflammation Research,
Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Charlotte Martin
- Research
Group of Organic Chemistry, Departments of Chemistry and Bioengineering
Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Viviane Bes
- Department
of Pathology and Immunology and Geneva Center for Inflammation Research,
Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Laureano E. Carpio
- ProtoQSAR
SL, Centro Europeo de Empresas Innovadoras, Parque Tecnológico de Valencia, Avda. Benjamin Franklin 12, 46980 Paterna, Spain
| | - Yana Girardin
- Structural
Biology Brussels, Department of Biotechnology, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
- Centre for
Structural Biology, VIB, Pleinlaan 2, 1050 Brussels, Belgium
| | - Remy Loris
- Structural
Biology Brussels, Department of Biotechnology, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
- Centre for
Structural Biology, VIB, Pleinlaan 2, 1050 Brussels, Belgium
| | - Andrés Tabernilla
- Research
Unit of In Vitro Toxicology and Dermato-Cosmetology, Department of
Pharmaceutical Sciences, Vrije Universiteit
Brussel, Laarbeeklaan
103, 1090 Brussels, Belgium
| | - Filippo Molica
- Department
of Pathology and Immunology and Geneva Center for Inflammation Research,
Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Rafael Gozalbes
- ProtoQSAR
SL, Centro Europeo de Empresas Innovadoras, Parque Tecnológico de Valencia, Avda. Benjamin Franklin 12, 46980 Paterna, Spain
- MolDrug
AI Systems SL, c/Olimpia
Arozena 45, 46018 Valencia, Spain
| | - María D. Mayán
- CellCOM
Research Group, Instituto de Investigación Biomédica
de A Coruña, Servizo Galego de Saúde, Universidade da Coruña, 15071 A Coruña, Spain
| | - Mathieu Vinken
- Research
Unit of In Vitro Toxicology and Dermato-Cosmetology, Department of
Pharmaceutical Sciences, Vrije Universiteit
Brussel, Laarbeeklaan
103, 1090 Brussels, Belgium
| | - Brenda R. Kwak
- Department
of Pathology and Immunology and Geneva Center for Inflammation Research,
Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Steven Ballet
- Research
Group of Organic Chemistry, Departments of Chemistry and Bioengineering
Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| |
Collapse
|
24
|
Phan TTT, Truong NV, Wu WG, Su YC, Hsu TS, Lin LY. Tumor suppressor p53 mediates interleukin-6 expression to enable cancer cell evasion of genotoxic stress. Cell Death Discov 2023; 9:340. [PMID: 37696858 PMCID: PMC10495329 DOI: 10.1038/s41420-023-01638-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023] Open
Abstract
The tumor suppressor p53 primarily functions as a mediator of DNA damage-induced cell death, thereby contributing to the efficacy of genotoxic anticancer therapeutics. Here, we show, on the contrary, that cancer cells can employ genotoxic stress-induced p53 to acquire treatment resistance through the production of the pleiotropic cytokine interleukin (IL)-6. Mechanistically, DNA damage, either repairable or irreparable, activates p53 and stimulates Caspase-2-mediated cleavage of its negative regulator mouse double minute 2 (MDM2) creating a positive feedback loop that leads to elevated p53 protein accumulation. p53 transcriptionally controls the major adenosine triphosphate (ATP) release channel pannexin 1 (Panx1), which directs IL-6 induction via a mechanism dependent on the extracellular ATP-activated purinergic P2 receptors as well as their downstream intracellular calcium (iCa2+)/PI3K/Akt/NF-ĸB signaling pathway. Thus, p53 silencing impairs Panx1 and IL-6 expression and renders cancer cells sensitive to genotoxic stress. Moreover, we confirm that IL-6 hampers the effectiveness of genotoxic anticancer agents by mitigating DNA damage, driving the expression of anti-apoptotic Bcl-2 family genes, and maintaining the migratory and invasive properties of cancer cells. Analysis of patient survival and relevant factors in lung cancer and pan-cancer cohorts supports the prognostic and clinical values of Panx1 and IL-6. Notably, IL-6 secreted by cancer cells during genotoxic treatments promotes the polarization of monocytic THP-1-derived macrophages into an alternative (M2-like) phenotype that exhibits impaired anti-survival activities but enhanced pro-metastatic effects on cancer cells as compared to nonpolarized macrophages. Our study reveals the precise mechanism for genotoxic-induced IL-6 and suggests that targeting p53-mediated IL-6 may improve the responsiveness of cancer cells to genotoxic anticancer therapy.
Collapse
Affiliation(s)
- Trinh T T Phan
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Nam V Truong
- Institute of Bioinformatics and Structural Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Wen-Guey Wu
- Institute of Bioinformatics and Structural Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Yi-Chun Su
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC.
| | - Lih-Yuan Lin
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC.
| |
Collapse
|
25
|
Caufriez A, Lamouroux A, Martin C, Iaculli D, Ince Ergüç E, Gozalbes R, Mayan MD, Kwak BR, Tabernilla A, Vinken M, Ballet S. Determination of structural features that underpin the pannexin1 channel inhibitory activity of the peptide 10Panx1. Bioorg Chem 2023; 138:106612. [PMID: 37210827 DOI: 10.1016/j.bioorg.2023.106612] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/29/2023] [Accepted: 05/11/2023] [Indexed: 05/23/2023]
Abstract
Pannexin1 channels facilitate paracrine communication and are involved in a broad spectrum of diseases. Attempts to find appropriate pannexin1 channel inhibitors that showcase target-selective properties and in vivo applicability remain nonetheless scarce. However, a promising lead candidate, the ten amino acid long peptide mimetic 10Panx1 (H-Trp1-Arg2-Gln3-Ala4-Ala5-Phe6-Val7-Asp8-Ser9-Tyr10-OH), has shown potential as a pannexin1 channel inhibitor in both in vitro and in vivo studies. Nonetheless, structural optimization is critical for clinical use. One of the main hurdles to overcome along the optimization process consists of subduing the low biological stability (10Panx1 t1/2 = 2.27 ± 0.11 min). To tackle this issue, identification of important structural features within the decapeptide structure is warranted. For this reason, a structure-activity relationship study was performed to proteolytically stabilize the sequence. Through an Alanine scan, this study demonstrated that the side chains of Gln3 and Asp8 are crucial for 10Panx1's channel inhibitory capacity. Guided by plasma stability experiments, scissile amide bonds were identified and stabilized, while extracellular adenosine triphosphate release experiments, indicative of pannexin1 channel functionality, allowed to enhance the in vitro inhibitory capacity of 10Panx1.
Collapse
Affiliation(s)
- Anne Caufriez
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; Research Group of In Vitro Toxicology and Dermato-cosmetology, Department of Pharmaceutical and Pharmacological sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Arthur Lamouroux
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Charlotte Martin
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Debora Iaculli
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Elif Ince Ergüç
- Research Group of In Vitro Toxicology and Dermato-cosmetology, Department of Pharmaceutical and Pharmacological sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Rafael Gozalbes
- ProtoQSAR SL, Centro Europeo de Empresas Innovadoras, Parque Tecnológico de Valencia, Avda. Benjamin Franklin 12, 46980 Paterna, Spain
| | - Maria D Mayan
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña, Servizo Galego de Saúde, Universidade da Coruña, 15071 A Coruña, Spain
| | - Brenda R Kwak
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland
| | - Andrés Tabernilla
- Research Group of In Vitro Toxicology and Dermato-cosmetology, Department of Pharmaceutical and Pharmacological sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Mathieu Vinken
- Research Group of In Vitro Toxicology and Dermato-cosmetology, Department of Pharmaceutical and Pharmacological sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| |
Collapse
|
26
|
Wu YL, Yang AH, Chiu YH. Recent advances in the structure and activation mechanisms of metabolite-releasing Pannexin 1 channels. Biochem Soc Trans 2023; 51:1687-1699. [PMID: 37622532 DOI: 10.1042/bst20230038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023]
Abstract
Pannexin 1 (PANX1) is a widely expressed large-pore ion channel located in the plasma membrane of almost all vertebrate cells. It possesses a unique ability to act as a conduit for both inorganic ions (e.g. potassium or chloride) and bioactive metabolites (e.g. ATP or glutamate), thereby activating varying signaling pathways in an autocrine or paracrine manner. Given its crucial role in cell-cell interactions, the activity of PANX1 has been implicated in maintaining homeostasis of cardiovascular, immune, and nervous systems. Dysregulation of PANX1 has also been linked to numerous diseases, such as ischemic stroke, seizure, and inflammatory disorders. Therefore, the mechanisms underlying different modes of PANX1 activation and its context-specific channel properties have gathered significant attention. In this review, we summarize the roles of PANX1 in various physiological processes and diseases, and analyze the accumulated lines of evidence supporting diverse molecular mechanisms associated with different PANX1 activation modalities. We focus on examining recent discoveries regarding PANX1 regulations by reversible post-translational modifications, elevated intracellular calcium concentration, and protein-protein interactions, as well as by irreversible cleavage of its C-terminal tail. Additionally, we delve into the caveats in the proposed PANX1 gating mechanisms and channel open-closed configurations by critically analyzing the structural insights derived from cryo-EM studies and the unitary properties of PANX1 channels. By doing so, we aim to identify potential research directions for a better understanding of the functions and regulations of PANX1 channels.
Collapse
Affiliation(s)
- Yi-Ling Wu
- Department of Life Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
| | - Ai-Hsing Yang
- Department of Life Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
| | - Yu-Hsin Chiu
- Department of Life Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
| |
Collapse
|
27
|
Truong NV, Phan TTT, Hsu TS, Phu Duc P, Lin LY, Wu WG. Action mechanism of snake venom l-amino acid oxidase and its double-edged sword effect on cancer treatment: Role of pannexin 1-mediated interleukin-6 expression. Redox Biol 2023; 64:102791. [PMID: 37385076 PMCID: PMC10331595 DOI: 10.1016/j.redox.2023.102791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/16/2023] [Indexed: 07/01/2023] Open
Abstract
Snake venom l-amino acid oxidases (svLAAOs) have been recognized as promising candidates for anticancer therapeutics. However, multiple aspects of their catalytic mechanism and the overall responses of cancer cells to these redox enzymes remain ambiguous. Here, we present an analysis of the phylogenetic relationships and active site-related residues among svLAAOs and reveal that the previously proposed critical catalytic residue His 223 is highly conserved in the viperid but not the elapid svLAAO clade. To gain further insight into the action mechanism of the elapid svLAAOs, we purify and characterize the structural, biochemical, and anticancer therapeutic potentials of the Thailand elapid snake Naja kaouthia LAAO (NK-LAAO). We find that NK-LAAO, with Ser 223, exhibits high catalytic activity toward hydrophobic l-amino acid substrates. Moreover, NK-LAAO induces substantial oxidative stress-mediated cytotoxicity with the magnitude relying on both the levels of extracellular hydrogen peroxide (H2O2) and intracellular reactive oxygen species (ROS) generated during the enzymatic redox reactions, but not being influenced by the N-linked glycans on its surface. Unexpectedly, we discover a tolerant mechanism deployed by cancer cells to dampen the anticancer activities of NK-LAAO. NK-LAAO treatment amplifies interleukin (IL)-6 expression via the pannexin 1 (Panx1)-directed intracellular calcium (iCa2+) signaling pathway to confer adaptive and aggressive phenotypes on cancer cells. Accordingly, IL-6 silencing renders cancer cells vulnerable to NK-LAAO-induced oxidative stress together with abrogating NK-LAAO-stimulated metastatic acquisition. Collectively, our study urges caution when using svLAAOs in cancer treatment and identifies the Panx1/iCa2+/IL-6 axis as a therapeutic target for improving the effectiveness of svLAAOs-based anticancer therapies.
Collapse
Affiliation(s)
- Nam V Truong
- Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Trinh T T Phan
- Institute of Molecular and Cellular Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Phan Phu Duc
- Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Lih-Yuan Lin
- Institute of Molecular and Cellular Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC.
| | - Wen-Guey Wu
- Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC.
| |
Collapse
|
28
|
Van Campenhout R, Caufriez A, Tabernilla A, Maerten A, De Boever S, Sanz-Serrano J, Kadam P, Vinken M. Pannexin1 channels in the liver: an open enemy. Front Cell Dev Biol 2023; 11:1220405. [PMID: 37492223 PMCID: PMC10363690 DOI: 10.3389/fcell.2023.1220405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/23/2023] [Indexed: 07/27/2023] Open
Abstract
Pannexin1 proteins form communication channels at the cell plasma membrane surface, which allow the transfer of small molecules and ions between the intracellular compartment and extracellular environment. In this way, pannexin1 channels play an important role in various cellular processes and diseases. Indeed, a plethora of human pathologies is associated with the activation of pannexin1 channels. The present paper reviews and summarizes the structure, life cycle, regulation and (patho)physiological roles of pannexin1 channels, with a particular focus on the relevance of pannexin1 channels in liver diseases.
Collapse
|
29
|
Yi X, Yang Y, Li T, Li M, Yao T, Hu G, Wan G, Chang B. Signaling metabolite β-aminoisobutyric acid as a metabolic regulator, biomarker, and potential exercise pill. Front Endocrinol (Lausanne) 2023; 14:1192458. [PMID: 37313446 PMCID: PMC10258315 DOI: 10.3389/fendo.2023.1192458] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/15/2023] [Indexed: 06/15/2023] Open
Abstract
Signaling metabolites can effectively regulate the biological functions of many tissues and organs. β-Aminoisobutyric acid (BAIBA), a product of valine and thymine catabolism in skeletal muscle, has been reported to participate in the regulation of lipid, glucose, and bone metabolism, as well as in inflammation and oxidative stress. BAIBA is produced during exercise and is involved in the exercise response. No side effect has been observed in human and rat studies, suggesting that BAIBA can be developed as a pill that confers the benefits of exercise to subjects who, for some reason, are unable to do so. Further, BAIBA has been confirmed to participate in the diagnosis and prevention of diseases as an important biological marker of disease. The current review aimed to discuss the roles of BAIBA in multiple physiological processes and the possible pathways of its action, and assess the progress toward the development of BAIBA as an exercise mimic and biomarker with relevance to multiple disease states, in order to provide new ideas and strategies for basic research and disease prevention in related fields.
Collapse
|
30
|
Purohit R, Bera AK. Carboxyl terminus of Pannexin-1 plays a crucial role in P2X7 receptor-mediated signaling. Biochem Biophys Res Commun 2023; 664:20-26. [PMID: 37130457 DOI: 10.1016/j.bbrc.2023.04.081] [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: 03/27/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 05/04/2023]
Abstract
The cellular implications of the interaction between Pannexin-1 (Panx1) channel and P2X7 receptor (P2X7R) have not been fully elucidated. Evidence suggests that ATP, released through Panx1, activates P2X7R, which in turn promotes further activation of Panx1. In a previous study, we reported that the C-terminus of Panx1 (Panx1-CT) attenuates P2X7R-mediated Ca2+ influx and cell death. One of the distinctive features of P2X7R is the gradual increase in current with repetitive stimulation. In the current study, we report an effect of Panx1-CT (amino acid residues 350 to 426) on P2X7R current, which differs from the effect of full-length Panx1. Panx1-CT inhibited P2X7R current, which persisted in all consecutive agonist applications. However, full-length Panx1 reduced P2X7R current at initial stimulations, followed by gradual augmentation. When P2X7R was activated for an extended period, cells expressing Panx1-CT exhibited less mitochondrial depolarization, reactive oxygen species (ROS) generation, Caspase 3 activation and cell death, whereas cells overexpressing full-length Panx1 showed the opposite effect. Taken together, these findings suggest that Panx1 can either attenuate or augment P2X7R-mediated cellular processes depending on the degree of P2X7R activation.
Collapse
Affiliation(s)
- Rutambhara Purohit
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600 036, Tamil Nadu, India.
| | - Amal Kanti Bera
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600 036, Tamil Nadu, India.
| |
Collapse
|
31
|
Koval M, Schug WJ, Isakson BE. Pharmacology of pannexin channels. Curr Opin Pharmacol 2023; 69:102359. [PMID: 36858833 PMCID: PMC10023479 DOI: 10.1016/j.coph.2023.102359] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/18/2023] [Accepted: 01/29/2023] [Indexed: 03/02/2023]
Abstract
Pannexin channels play fundamental roles in regulating inflammation and have been implicated in many diseases including hypertension, stroke, and neuropathic pain. Thus, the ability to pharmacologically block these channels is a vital component of several therapeutic approaches. Pharmacologic interrogation of model systems also provides a means to discover new roles for pannexins in cell physiology. Here, we review the state of the art for agents that can be used to block pannexin channels, with a focus on chemical pharmaceuticals and peptide mimetics that act on pannexin 1. Guidance on interpreting results obtained with pannexin pharmacologics in experimental systems is discussed, as well as strengths and caveats of different agents, including specificity and feasibility of clinical application.
Collapse
Affiliation(s)
- Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Wyatt J Schug
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
32
|
Dale N, Butler J, Dospinescu VM, Nijjar S. Channel-mediated ATP release in the nervous system. Neuropharmacology 2023; 227:109435. [PMID: 36690324 DOI: 10.1016/j.neuropharm.2023.109435] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023]
Abstract
ATP is well established as a transmitter and modulator in the peripheral and central nervous system. While conventional exocytotic release of ATP at synapses occurs, this transmitter is unusual in also being released into the extracellular space via large-pored plasma membrane channels. This review considers the channels that are known to be permeable to ATP and some of the functions of channel-mediated ATP release. While the possibility of ATP release via channels mediating volume transmission has been known for some time, localised ATP release via channels at specialised synapses made by taste cells to the afferent nerve has recently been documented in taste buds. This raises the prospect that "channel synapses" may occur in other contexts. However, volume transmission and channel synapses are not necessarily mutually exclusive. We suggest that certain glial cells in the brain stem and hypothalamus, which possess long processes and are known to release ATP, may be candidates for both modes of ATP release -channel-mediated volume transmission in the region of their somata and more localised transmission possibly via either conventional or channel synapses from their processes at distal targets. Finally, we consider the different characteristics of vesicular and channel synapses and suggest that channel synapses may be advantageous in requiring less energy than their conventional vesicular counterparts. This article is part of the Special Issue on "Purinergic Signaling: 50 years".
Collapse
Affiliation(s)
- Nicholas Dale
- School of Life Sciences, University of Warwick, Coventry, CV4, AL, UK.
| | - Jack Butler
- School of Life Sciences, University of Warwick, Coventry, CV4, AL, UK
| | | | - Sarbjit Nijjar
- School of Life Sciences, University of Warwick, Coventry, CV4, AL, UK
| |
Collapse
|
33
|
Cryo-EM structure of human heptameric pannexin 2 channel. Nat Commun 2023; 14:1118. [PMID: 36869038 PMCID: PMC9984531 DOI: 10.1038/s41467-023-36861-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/19/2023] [Indexed: 03/05/2023] Open
Abstract
Pannexin 2 (Panx2) is a large-pore ATP-permeable channel with critical roles in various physiological processes, such as the inflammatory response, energy production and apoptosis. Its dysfunction is related to numerous pathological conditions including ischemic brain injury, glioma and glioblastoma multiforme. However, the working mechanism of Panx2 remains unclear. Here, we present the cryo-electron microscopy structure of human Panx2 at a resolution of 3.4 Å. Panx2 structure assembles as a heptamer, forming an exceptionally wide channel pore across the transmembrane and intracellular domains, which is compatible with ATP permeation. Comparing Panx2 with Panx1 structures in different states reveals that the Panx2 structure corresponds to an open channel state. A ring of seven arginine residues located at the extracellular entrance forms the narrowest site of the channel, which serves as the critical molecular filter controlling the permeation of substrate molecules. This is further verified by molecular dynamics simulations and ATP release assays. Our studies reveal the architecture of the Panx2 channel and provide insights into the molecular mechanism of its channel gating.
Collapse
|
34
|
Ladd Z, Su G, Hartman J, Lu G, Hensley S, Upchurch GR, Sharma AK. Pharmacologic inhibition by spironolactone attenuates experimental abdominal aortic aneurysms. Front Cardiovasc Med 2023; 10:1101389. [PMID: 36776267 PMCID: PMC9908993 DOI: 10.3389/fcvm.2023.1101389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/13/2023] [Indexed: 01/27/2023] Open
Abstract
Background Abdominal aortic aneurysms (AAA) are characterized by vascular inflammation and remodeling that can lead to aortic rupture resulting in significant mortality. Pannexin-1 channels on endothelial cells (ECs) can modulate ATP secretion to regulate the pathogenesis of AAA formation. Our hypothesis focused on potential of spironolactone to inhibit EC-mediated ATP release for the mitigation of AAA formation. Methods A topical elastase AAA model was used initially in C57BL/6 (wild-type; WT) male mice. Mice were administered either a vehicle control (saline) or spironolactone and analyzed on day 14. In a second chronic AAA model, mice were subjected to elastase and β-aminopropionitrile (BAPN) treatment with/without administration of spironolactone to pre-formed aneurysms starting on day 14 and analyzed on day 28. Aortic diameter was evaluated by video micrometry and aortic tissue was analyzed for cytokine expression and histology. ATP measurement and matrix metalloproteinase (MMP2) activity was evaluated in aortic tissue on days 14 or -28. In vitro studies were performed to evaluate the crosstalk between aortic ECs with macrophages or smooth muscle cells. Results In the elastase AAA model, spironolactone treatment displayed a significant decrease in aortic diameter compared to elastase-treated controls on day 14. A significant increase in smooth muscle α-actin expression as well as decrease in elastic fiber disruption and immune cell (macrophages and neutrophils) infiltration was observed in mice treated with spironolactone compared to saline-treated controls. Spironolactone treatment also significantly mitigated pro-inflammatory cytokine expression, MMP2 activity and ATP content in aortic tissue compared to controls. Moreover, in the chronic AAA model, spironolactone treatment of pre-formed aneurysms significantly attenuated vascular inflammation and remodeling to attenuate the progression of AAAs compared to controls. Mechanistically, in vitro data demonstrated that spironolactone treatment attenuates extracellular ATP release from endothelial cells to mitigate macrophage activation (IL-1β and HMGB1 expression) and smooth muscle cell-dependent vascular remodeling (MMP2 activity). Conclusion These results demonstrate that spironolactone can mitigate aortic inflammation and remodeling to attenuate AAA formation as well as decrease growth of pre-formed aneurysms via inhibition of EC-dependent ATP release. Therefore, this study implicates a therapeutic application of spironolactone in the treatment of AAAs.
Collapse
|
35
|
MSCs-derived apoptotic extracellular vesicles promote muscle regeneration by inducing Pannexin 1 channel-dependent creatine release by myoblasts. Int J Oral Sci 2023; 15:7. [PMID: 36646698 PMCID: PMC9842731 DOI: 10.1038/s41368-022-00205-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 01/18/2023] Open
Abstract
Severe muscle injury is hard to heal and always results in a poor prognosis. Recent studies found that extracellular vesicle-based therapy has promising prospects for regeneration medicine, however, whether extracellular vesicles have therapeutic effects on severe muscle injury is still unknown. Herein, we extracted apoptotic extracellular vesicles derived from mesenchymal stem cells (MSCs-ApoEVs) to treat cardiotoxin induced tibialis anterior (TA) injury and found that MSCs-ApoEVs promoted muscles regeneration and increased the proportion of multinucleated cells. Besides that, we also found that apoptosis was synchronized during myoblasts fusion and MSCs-ApoEVs promoted the apoptosis ratio as well as the fusion index of myoblasts. Furthermore, we revealed that MSCs-ApoEVs increased the relative level of creatine during myoblasts fusion, which was released via activated Pannexin 1 channel. Moreover, we also found that activated Pannexin 1 channel was highly expressed on the membrane of myoblasts-derived ApoEVs (Myo-ApoEVs) instead of apoptotic myoblasts, and creatine was the pivotal metabolite involved in myoblasts fusion. Collectively, our findings firstly revealed that MSCs-ApoEVs can promote muscle regeneration and elucidated that the new function of ApoEVs as passing inter-cell messages through releasing metabolites from activated Pannexin 1 channel, which will provide new evidence for extracellular vesicles-based therapy as well as improving the understanding of new functions of extracellular vesicles.
Collapse
|
36
|
Vega JL, Gutiérrez C, Rojas M, Güiza J, Sáez JC. Contribution of large-pore channels to inflammation induced by microorganisms. Front Cell Dev Biol 2023; 10:1094362. [PMID: 36699007 PMCID: PMC9868820 DOI: 10.3389/fcell.2022.1094362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
Plasma membrane ionic channels selectively permeate potassium, sodium, calcium, and chloride ions. However, large-pore channels are permeable to ions and small molecules such as ATP and glutamate, among others. Large-pore channels are structures formed by several protein families with little or no evolutionary linkages including connexins (Cxs), pannexins (Panxs), innexin (Inxs), unnexins (Unxs), calcium homeostasis modulator (CALHMs), and Leucine-rich repeat-containing 8 (LRRC8) proteins. Large-pore channels are key players in inflammatory cell response, guiding the activation of inflammasomes, the release of pro-inflammatory cytokines such as interleukin-1 beta (IL-1ß), and the release of adenosine-5'-triphosphate (ATP), which is considered a danger signal. This review summarizes our current understanding of large-pore channels and their contribution to inflammation induced by microorganisms, virulence factors or their toxins.
Collapse
Affiliation(s)
- José L. Vega
- Laboratory of Gap Junctions Proteins and Parasitic Diseases (GaPaL), Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile,Centro de Investigación en Inmunología y Biotecnología Biomédica de Antofagasta (CIIBBA), Universidad de Antofagasta, Antofagasta, Chile,Centro de Fisiología y Medicina de Altura (FIMEDALT), Universidad de Antofagasta, Antofagasta, Chile,*Correspondence: José L. Vega,
| | - Camila Gutiérrez
- Laboratory of Gap Junctions Proteins and Parasitic Diseases (GaPaL), Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile
| | - Mauro Rojas
- Laboratory of Gap Junctions Proteins and Parasitic Diseases (GaPaL), Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile
| | - Juan Güiza
- Laboratory of Gap Junctions Proteins and Parasitic Diseases (GaPaL), Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile
| | - Juan C. Sáez
- Centro Interdisciplinario de Neurociencias de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
37
|
Zhou J, Wang M, Hu J, Li Z, Zhu L, Jin L. A novel heterozygous variant in PANX1 causes primary infertility due to oocyte death. J Assist Reprod Genet 2023; 40:65-73. [PMID: 36469255 PMCID: PMC9840723 DOI: 10.1007/s10815-022-02666-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/18/2022] [Indexed: 12/08/2022] Open
Abstract
PURPOSE Variants in the pannexin1 (PANX1) gene have been reported to be associated with oocyte death and recurrent in vitro fertilization failure. In this study, we performed genetic analysis in the patient with female infertility due to oocyte death to identify the disease-causing gene variant in the patient. METHODS We characterized one patient from a non-consanguineous family who had suffered from oocyte death and female infertility. Whole-exome sequencing and Sanger sequencing were used to identify the variant in the family. Western blot analysis was used to check the effect of the variant on PANX1 glycosylation pattern in vitro. RESULTS We identified a novel heterozygous PANX1 variant (NM_015368.4 c.976_978del, (p.Asn326del)) associated with the phenotype of oocyte death in a non-consanguineous family, followed by an autosomal dominant (AD) mode. This variant showed a more delayed emergence of oocyte death than previously reported articles. Western blot analysis confirmed that the deletion variant of PANX1 (c.976_978del) altered the glycosylation pattern in HeLa cells. CONCLUSIONS Our findings expand the variant spectrum of PANX1 genes associated with oocyte death and provide new support for the genetic diagnosis of female infertility.
Collapse
Affiliation(s)
- Juepu Zhou
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030 China
| | - Meng Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030 China
| | - Juan Hu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030 China
| | - Zhou Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030 China
| | - Lixia Zhu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030 China
| | - Lei Jin
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030 China
| |
Collapse
|
38
|
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
|
39
|
Flores-Muñoz C, García-Rojas F, Pérez MA, Santander O, Mery E, Ordenes S, Illanes-González J, López-Espíndola D, González-Jamett AM, Fuenzalida M, Martínez AD, Ardiles ÁO. The Long-Term Pannexin 1 Ablation Produces Structural and Functional Modifications in Hippocampal Neurons. Cells 2022; 11:cells11223646. [PMID: 36429074 PMCID: PMC9688914 DOI: 10.3390/cells11223646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/29/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022] Open
Abstract
Enhanced activity and overexpression of Pannexin 1 (Panx1) channels contribute to neuronal pathologies such as epilepsy and Alzheimer's disease (AD). The Panx1 channel ablation alters the hippocampus's glutamatergic neurotransmission, synaptic plasticity, and memory flexibility. Nevertheless, Panx1-knockout (Panx1-KO) mice still retain the ability to learn, suggesting that compensatory mechanisms stabilize their neuronal activity. Here, we show that the absence of Panx1 in the adult brain promotes a series of structural and functional modifications in the Panx1-KO hippocampal synapses, preserving spontaneous activity. Compared to the wild-type (WT) condition, the adult hippocampal neurons of Panx1-KO mice exhibit enhanced excitability, a more complex dendritic branching, enhanced spine maturation, and an increased proportion of multiple synaptic contacts. These modifications seem to rely on the actin-cytoskeleton dynamics as an increase in the actin polymerization and an imbalance between the Rac1 and the RhoA GTPase activities were observed in Panx1-KO brain tissues. Our findings highlight a novel interaction between Panx1 channels, actin, and Rho GTPases, which appear to be relevant for synapse stability.
Collapse
Affiliation(s)
- Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Francisca García-Rojas
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Miguel A. Pérez
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Escuela de Ciencias de la Salud, Universidad de Viña del Mar, Viña del Mar 2572007, Chile
| | - Odra Santander
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Elena Mery
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Stefany Ordenes
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Javiera Illanes-González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Daniela López-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2529002, Chile
- Centro de Investigaciones Biomédicas, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2529002, Chile
| | - Arlek M. González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Correspondence: (M.F.); (A.D.M.); (Á.O.A.)
| | - Agustín D. Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Correspondence: (M.F.); (A.D.M.); (Á.O.A.)
| | - Álvaro O. Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro Interdisciplinario de estudios en salud, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2572007, Chile
- Correspondence: (M.F.); (A.D.M.); (Á.O.A.)
| |
Collapse
|
40
|
Rusiecka OM, Tournier M, Molica F, Kwak BR. Pannexin1 channels-a potential therapeutic target in inflammation. Front Cell Dev Biol 2022; 10:1020826. [PMID: 36438559 PMCID: PMC9682086 DOI: 10.3389/fcell.2022.1020826] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/20/2022] [Indexed: 08/11/2023] Open
Abstract
An exaggerated inflammatory response is the hallmark of a plethora of disorders. ATP is a central signaling molecule that orchestrates the initiation and resolution of the inflammatory response by enhancing activation of the inflammasome, leukocyte recruitment and activation of T cells. ATP can be released from cells through pannexin (Panx) channels, a family of glycoproteins consisting of three members, Panx1, Panx2, and Panx3. Panx1 is ubiquitously expressed and forms heptameric channels in the plasma membrane mediating paracrine and autocrine signaling. Besides their involvement in the inflammatory response, Panx1 channels have been shown to contribute to different modes of cell death (i.e., pyroptosis, necrosis and apoptosis). Both genetic ablation and pharmacological inhibition of Panx1 channels decrease inflammation in vivo and contribute to a better outcome in several animal models of inflammatory disease involving various organs, including the brain, lung, kidney and heart. Up to date, several molecules have been identified to inhibit Panx1 channels, for instance probenecid (Pbn), mefloquine (Mfq), flufenamic acid (FFA), carbenoxolone (Cbx) or mimetic peptides like 10Panx1. Unfortunately, the vast majority of these compounds lack specificity and/or serum stability, which limits their application. The recent availability of detailed structural information on the Panx1 channel from cryo-electron microscopy studies may open up innovative approaches to acquire new classes of synthetic Panx1 channel blockers with high target specificity. Selective inhibition of Panx1 channels may not only limit acute inflammatory responses but may also prove useful in chronic inflammatory diseases, thereby improving human health. Here, we reviewed the current knowledge on the role of Panx1 in the initiation and resolution of the inflammatory response, we summarized the effects of Panx1 inhibition in inflammatory pathologies and recapitulate current Panx1 channel pharmacology with an outlook towards future approaches.
Collapse
Affiliation(s)
- Olga M. Rusiecka
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Malaury Tournier
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Filippo Molica
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Brenda R. Kwak
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
41
|
King DR, Sedovy MW, Eaton X, Dunaway LS, Good ME, Isakson BE, Johnstone SR. Cell-To-Cell Communication in the Resistance Vasculature. Compr Physiol 2022; 12:3833-3867. [PMID: 35959755 DOI: 10.1002/cphy.c210040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The arterial vasculature can be divided into large conduit arteries, intermediate contractile arteries, resistance arteries, arterioles, and capillaries. Resistance arteries and arterioles primarily function to control systemic blood pressure. The resistance arteries are composed of a layer of endothelial cells oriented parallel to the direction of blood flow, which are separated by a matrix layer termed the internal elastic lamina from several layers of smooth muscle cells oriented perpendicular to the direction of blood flow. Cells within the vessel walls communicate in a homocellular and heterocellular fashion to govern luminal diameter, arterial resistance, and blood pressure. At rest, potassium currents govern the basal state of endothelial and smooth muscle cells. Multiple stimuli can elicit rises in intracellular calcium levels in either endothelial cells or smooth muscle cells, sourced from intracellular stores such as the endoplasmic reticulum or the extracellular space. In general, activation of endothelial cells results in the production of a vasodilatory signal, usually in the form of nitric oxide or endothelial-derived hyperpolarization. Conversely, activation of smooth muscle cells results in a vasoconstriction response through smooth muscle cell contraction. © 2022 American Physiological Society. Compr Physiol 12: 1-35, 2022.
Collapse
Affiliation(s)
- D Ryan King
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA
| | - Meghan W Sedovy
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA.,Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Blacksburg, Virginia, USA
| | - Xinyan Eaton
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA
| | - Luke S Dunaway
- Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Miranda E Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Scott R Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
42
|
Burboa PC, Puebla M, Gaete PS, Durán WN, Lillo MA. Connexin and Pannexin Large-Pore Channels in Microcirculation and Neurovascular Coupling Function. Int J Mol Sci 2022; 23:ijms23137303. [PMID: 35806312 PMCID: PMC9266979 DOI: 10.3390/ijms23137303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 01/27/2023] Open
Abstract
Microcirculation homeostasis depends on several channels permeable to ions and/or small molecules that facilitate the regulation of the vasomotor tone, hyperpermeability, the blood–brain barrier, and the neurovascular coupling function. Connexin (Cxs) and Pannexin (Panxs) large-pore channel proteins are implicated in several aspects of vascular physiology. The permeation of ions (i.e., Ca2+) and key metabolites (ATP, prostaglandins, D-serine, etc.) through Cxs (i.e., gap junction channels or hemichannels) and Panxs proteins plays a vital role in intercellular communication and maintaining vascular homeostasis. Therefore, dysregulation or genetic pathologies associated with these channels promote deleterious tissue consequences. This review provides an overview of current knowledge concerning the physiological role of these large-pore molecule channels in microcirculation (arterioles, capillaries, venules) and in the neurovascular coupling function.
Collapse
Affiliation(s)
- Pía C. Burboa
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA; (P.C.B.); (W.N.D.)
- Departamento de Morfología y Función, Facultad de Salud y Ciencias Sociales, Sede Santiago Centro, Universidad de las Américas, Avenue República 71, Santiago 8370040, Chile;
| | - Mariela Puebla
- Departamento de Morfología y Función, Facultad de Salud y Ciencias Sociales, Sede Santiago Centro, Universidad de las Américas, Avenue República 71, Santiago 8370040, Chile;
| | - Pablo S. Gaete
- Department of Physiology and Membrane Biology, University of California at Davis, Davis, CA 95616, USA;
| | - Walter N. Durán
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA; (P.C.B.); (W.N.D.)
- Rutgers School of Graduate Studies, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Mauricio A. Lillo
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA; (P.C.B.); (W.N.D.)
- Correspondence:
| |
Collapse
|
43
|
NMDA and P2X7 Receptors Require Pannexin 1 Activation to Initiate and Maintain Nociceptive Signaling in the Spinal Cord of Neuropathic Rats. Int J Mol Sci 2022; 23:ijms23126705. [PMID: 35743148 PMCID: PMC9223805 DOI: 10.3390/ijms23126705] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 02/01/2023] Open
Abstract
Pannexin 1 (Panx1) is involved in the spinal central sensitization process in rats with neuropathic pain, but its interaction with well-known, pain-related, ligand-dependent receptors, such as NMDA receptors (NMDAR) and P2X7 purinoceptors (P2X7R), remains largely unexplored. Here, we studied whether NMDAR- and P2X7R-dependent nociceptive signaling in neuropathic rats require the activation of Panx1 channels to generate spinal central sensitization, as assessed by behavioral (mechanical hyperalgesia) and electrophysiological (C-reflex wind-up potentiation) indexes. Administration of either a selective NMDAR agonist i.t. (NMDA, 2 mM) or a P2X7R agonist (BzATP, 150 μM) significantly increased both the mechanical hyperalgesia and the C-reflex wind-up potentiation, effects that were rapidly reversed (minutes) by i.t. administration of a selective pannexin 1 antagonist (10panx peptide, 300 μM), with the scores even reaching values of rats without neuropathy. Accordingly, 300 μM 10panx completely prevented the effects of NMDA and BzATP administered 1 h later, on mechanical hyperalgesia and C-reflex wind-up potentiation. Confocal immunofluorescence imaging revealed coexpression of Panx1 with NeuN protein in intrinsic dorsal horn neurons of neuropathic rats. The results indicate that both NMDAR- and P2X7R-mediated increases in mechanical hyperalgesia and C-reflex wind-up potentiation require neuronal Panx1 channel activation to initiate and maintain nociceptive signaling in neuropathic rats.
Collapse
|
44
|
Caufriez A, Tabernilla A, Van Campenhout R, Cooreman A, Leroy K, Sanz Serrano J, Kadam P, dos Santos Rodrigues B, Lamouroux A, Ballet S, Vinken M. Effects of Drugs Formerly Suggested for COVID-19 Repurposing on Pannexin1 Channels. Int J Mol Sci 2022; 23:ijms23105664. [PMID: 35628472 PMCID: PMC9146942 DOI: 10.3390/ijms23105664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
Although many efforts have been made to elucidate the pathogenesis of COVID-19, the underlying mechanisms are yet to be fully uncovered. However, it is known that a dysfunctional immune response and the accompanying uncontrollable inflammation lead to troublesome outcomes in COVID-19 patients. Pannexin1 channels are put forward as interesting drug targets for the treatment of COVID-19 due to their key role in inflammation and their link to other viral infections. In the present study, we selected a panel of drugs previously tested in clinical trials as potential candidates for the treatment of COVID-19 early on in the pandemic, including hydroxychloroquine, chloroquine, azithromycin, dexamethasone, ribavirin, remdesivir, favipiravir, lopinavir, and ritonavir. The effect of the drugs on pannexin1 channels was assessed at a functional level by means of measurement of extracellular ATP release. Immunoblot analysis and real-time quantitative reversetranscription polymerase chain reaction analysis were used to study the potential of the drugs to alter pannexin1 protein and mRNA expression levels, respectively. Favipiravir, hydroxychloroquine, lopinavir, and the combination of lopinavir with ritonavir were found to inhibit pannexin1 channel activity without affecting pannexin1 protein or mRNA levels. Thusthree new inhibitors of pannexin1 channels were identified that, though currently not being used anymore for the treatment of COVID-19 patients, could be potential drug candidates for other pannexin1-related diseases.
Collapse
Affiliation(s)
- Anne Caufriez
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (A.C.); (A.T.); (R.V.C.); (A.C.); (K.L.); (J.S.S.); (P.K.); (B.d.S.R.)
- Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; (A.L.); (S.B.)
| | - Andrés Tabernilla
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (A.C.); (A.T.); (R.V.C.); (A.C.); (K.L.); (J.S.S.); (P.K.); (B.d.S.R.)
| | - Raf Van Campenhout
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (A.C.); (A.T.); (R.V.C.); (A.C.); (K.L.); (J.S.S.); (P.K.); (B.d.S.R.)
| | - Axelle Cooreman
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (A.C.); (A.T.); (R.V.C.); (A.C.); (K.L.); (J.S.S.); (P.K.); (B.d.S.R.)
| | - Kaat Leroy
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (A.C.); (A.T.); (R.V.C.); (A.C.); (K.L.); (J.S.S.); (P.K.); (B.d.S.R.)
| | - Julen Sanz Serrano
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (A.C.); (A.T.); (R.V.C.); (A.C.); (K.L.); (J.S.S.); (P.K.); (B.d.S.R.)
| | - Prashant Kadam
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (A.C.); (A.T.); (R.V.C.); (A.C.); (K.L.); (J.S.S.); (P.K.); (B.d.S.R.)
| | - Bruna dos Santos Rodrigues
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (A.C.); (A.T.); (R.V.C.); (A.C.); (K.L.); (J.S.S.); (P.K.); (B.d.S.R.)
| | - Arthur Lamouroux
- Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; (A.L.); (S.B.)
| | - Steven Ballet
- Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; (A.L.); (S.B.)
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (A.C.); (A.T.); (R.V.C.); (A.C.); (K.L.); (J.S.S.); (P.K.); (B.d.S.R.)
- Correspondence: ; Tel.: +32-2477-4587
| |
Collapse
|
45
|
Whyte-Fagundes P, Taskina D, Safarian N, Zoidl C, Carlen PL, Donaldson LW, Zoidl GR. Panx1 channels promote both anti- and pro-seizure-like activities in the zebrafish via p2rx7 receptors and ATP signaling. Commun Biol 2022; 5:472. [PMID: 35585187 PMCID: PMC9117279 DOI: 10.1038/s42003-022-03356-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/12/2022] [Indexed: 11/08/2022] Open
Abstract
The molecular mechanisms of excitation/inhibition imbalances promoting seizure generation in epilepsy patients are not fully understood. Evidence suggests that Pannexin1 (Panx1), an ATP release channel, modulates the excitability of the brain. In this report, we performed electrophysiological, behavioral, and molecular phenotyping experiments on zebrafish larvae bearing genetic or pharmacological knockouts of Panx1a and Panx1b channels, each homologous to human PANX1. When Panx1a function is lost, or both channels are under pharmacological blockade, seizures with ictal-like events and seizure-like locomotion are reduced in the presence of pentylenetetrazol. Transcriptome profiling by RNA-seq demonstrates a spectrum of distinct metabolic and cell signaling states which correlate with the loss of Panx1a. Furthermore, the pro- and anticonvulsant activities of both Panx1 channels affect ATP release and involve the purinergic receptor P2rx7. Our findings suggest a subfunctionalization of Panx1 enabling dual roles in seizures, providing a unique and comprehensive perspective to understanding seizure mechanisms in the context of this channel.
Collapse
Affiliation(s)
- Paige Whyte-Fagundes
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada.
- Center of Vision Research (CVR), York University, Toronto, ON, M3J1P3, Canada.
- Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 1M8, Canada.
| | - Daria Taskina
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada
- Center of Vision Research (CVR), York University, Toronto, ON, M3J1P3, Canada
| | - Nickie Safarian
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada
- Center of Vision Research (CVR), York University, Toronto, ON, M3J1P3, Canada
| | - Christiane Zoidl
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada
- Center of Vision Research (CVR), York University, Toronto, ON, M3J1P3, Canada
| | - Peter L Carlen
- Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 1M8, Canada
- Department of Medicine, Physiology and BME, University of Toronto, 399 Bathurst St., 5w442, Toronto, ON, M5T 2S8, Canada
| | | | - Georg R Zoidl
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada.
- Center of Vision Research (CVR), York University, Toronto, ON, M3J1P3, Canada.
- Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 1M8, Canada.
| |
Collapse
|
46
|
Lucas CD, Medina CB, Bruton FA, Dorward DA, Raymond MH, Tufan T, Etchegaray JI, Barron B, Oremek ME, Arandjelovic S, Farber E, Onngut-Gumuscu S, Ke E, Whyte MKB, Rossi AG, Ravichandran KS. Pannexin 1 drives efficient epithelial repair after tissue injury. Sci Immunol 2022; 7:eabm4032. [PMID: 35559667 PMCID: PMC7612772 DOI: 10.1126/sciimmunol.abm4032] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Epithelial tissues such as lung and skin are exposed to the environment and therefore particularly vulnerable to damage during injury or infection. Rapid repair is therefore essential to restore function and organ homeostasis. Dysregulated epithelial tissue repair occurs in several human disease states, yet how individual cell types communicate and interact to coordinate tissue regeneration is incompletely understood. Here, we show that pannexin 1 (Panx1), a cell membrane channel activated by caspases in dying cells, drives efficient epithelial regeneration after tissue injury by regulating injury-induced epithelial proliferation. Lung airway epithelial injury promotes the Panx1-dependent release of factors including ATP, from dying epithelial cells, which regulates macrophage phenotype after injury. This process, in turn, induces a reparative response in tissue macrophages that includes the induction of the soluble mitogen amphiregulin, which promotes injury-induced epithelial proliferation. Analysis of regenerating lung epithelium identified Panx1-dependent induction of Nras and Bcas2, both of which positively promoted epithelial proliferation and tissue regeneration in vivo. We also established that this role of Panx1 in boosting epithelial repair after injury is conserved between mouse lung and zebrafish tailfin. These data identify a Panx1-mediated communication circuit between epithelial cells and macrophages as a key step in promoting epithelial regeneration after injury.
Collapse
Affiliation(s)
- Christopher D. Lucas
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
- Institute for Regeneration and Repair, Edinburgh BioQuarter, UK
| | - Christopher B. Medina
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Finnius A. Bruton
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
| | - David A. Dorward
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
| | - Michael H. Raymond
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Turan Tufan
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - J. Iker Etchegaray
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Brady Barron
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Magdalena E.M. Oremek
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
| | - Sanja Arandjelovic
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Emily Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Suna Onngut-Gumuscu
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Eugene Ke
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Moira KB Whyte
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
| | - Adriano G. Rossi
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
| | - Kodi S. Ravichandran
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Inflammation Research Centre, VIB, and the Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
47
|
Crocetti L, Guerrini G, Giovannoni MP, Melani F, Lamanna S, Di Cesare Mannelli L, Lucarini E, Ghelardini C, Wang J, Dahl G. New Panx-1 Blockers: Synthesis, Biological Evaluation and Molecular Dynamic Studies. Int J Mol Sci 2022; 23:4827. [PMID: 35563213 PMCID: PMC9103715 DOI: 10.3390/ijms23094827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/25/2022] Open
Abstract
The channel protein Panx-1 is involved in some pathologies, such as epilepsy, ischemic stroke, cancer and Parkinson's disease, as well as in neuropathic pain. These observations make Panx-1 an interesting biological target. We previously published some potent indole derivatives as Panx-1 blockers, and as continuation of the research in this field we report here the studies on additional chemical scaffolds, naphthalene and pyrazole, appropriately substituted with those functions that gave the best results as in our indole series (sulphonamide functions and one/two carboxylic groups) and in Panx-1 blockers reported in the literature (sulphonic acid). Compounds 4 and 13, the latter being an analogue of the drug Probenecid, are the most potent Panx-1 blockers obtained in this study, with I = 97% and I = 93.7% at 50 µM, respectively. Both compounds, tested in a mouse model of oxaliplatin-induced neuropathic pain, showed a similar anti-hypersensitivity profile and are able to significantly increase the mouse pain threshold 45 min after the injection of the doses of 1 nmol and 3 nmol. Finally, the molecular dynamic studies and the PCA analysis have made it possible to identify a discriminating factor able to separate active compounds from inactive ones.
Collapse
Affiliation(s)
- Letizia Crocetti
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Gabriella Guerrini
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Maria Paola Giovannoni
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Fabrizio Melani
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Silvia Lamanna
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Lorenzo Di Cesare Mannelli
- NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Elena Lucarini
- NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Carla Ghelardini
- NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Junjie Wang
- Department of Physiology and Biophysics, University of Miami School of Medicine, 1600 N.W. 10th Avenue, Miami, FL 33136, USA
| | - Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami School of Medicine, 1600 N.W. 10th Avenue, Miami, FL 33136, USA
| |
Collapse
|
48
|
Liu C, Shen Y, Huang L, Wang J. TLR2/caspase-5/Panx1 pathway mediates necrosis-induced NLRP3 inflammasome activation in macrophages during acute kidney injury. Cell Death Dis 2022; 8:232. [PMID: 35473933 PMCID: PMC9042857 DOI: 10.1038/s41420-022-01032-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 11/19/2022]
Abstract
Acute kidney injury (AKI) is characterized by necroinflammation formed by necrotic tubular epithelial cells (TECs) and interstitial inflammation. In necroinflammation, macrophages are key inflammatory cells and can be activated and polarized into proinflammatory macrophages. Membranous Toll-like receptors (TLRs) can cooperate with intracellular NOD-like receptor protein 3 (NLRP3) to recognize danger signals from necrotic TECs and activate proinflammatory macrophages by assembling NLRP3 inflammasome. However, the cooperation between TLRs and NLRP3 is still unclear. Using conditioned medium from necrotic TECs, we confirmed that necrotic TECs could release danger signals to activate NLRP3 inflammasome in macrophages. We further identified that necrotic TECs-induced NLRP3 inflammasome activation was dependent on ATP secretion via Pannexin-1 (Panx1) channel in macrophages. Next, we verified that TLR2 was required for the activation of Panx1 and NLRP3 in macrophages. Mechanistically, we indicated that caspase-5 mediated TLR2-induced Panx1 activation. In addition, we showed that necrotic TECs-induced activation of TLR2/caspase-5/Panx1 axis could be decreased in macrophages when TECs was protected by N-acetylcysteine (NAC). Overall, we demonstrate that danger signals from necrotic TECs could activate NLRP3 inflammasome in macrophages via TLR2/caspase-5/Panx1 axis during AKI.
Collapse
Affiliation(s)
- Chongbin Liu
- Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Yanting Shen
- Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Liuwei Huang
- Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jun Wang
- Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China. .,Department of Nephrology, The First People's Hospital of Kashi, Kashi, PR China.
| |
Collapse
|
49
|
Filiberto AC, Spinosa MD, Elder CT, Su G, Leroy V, Ladd Z, Lu G, Mehaffey JH, Salmon MD, Hawkins RB, Ravichandran KS, Isakson BE, Upchurch GR, Sharma AK. Endothelial pannexin-1 channels modulate macrophage and smooth muscle cell activation in abdominal aortic aneurysm formation. Nat Commun 2022; 13:1521. [PMID: 35315432 PMCID: PMC8938517 DOI: 10.1038/s41467-022-29233-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 03/07/2022] [Indexed: 01/17/2023] Open
Abstract
Pannexin-1 (Panx1) channels have been shown to regulate leukocyte trafficking and tissue inflammation but the mechanism of Panx1 in chronic vascular diseases like abdominal aortic aneurysms (AAA) is unknown. Here we demonstrate that Panx1 on endothelial cells, but not smooth muscle cells, orchestrate a cascade of signaling events to mediate vascular inflammation and remodeling. Mechanistically, Panx1 on endothelial cells acts as a conduit for ATP release that stimulates macrophage activation via P2X7 receptors and mitochondrial DNA release to increase IL-1β and HMGB1 secretion. Secondly, Panx1 signaling regulates smooth muscle cell-dependent intracellular Ca2+ release and vascular remodeling via P2Y2 receptors. Panx1 blockade using probenecid markedly inhibits leukocyte transmigration, aortic inflammation and remodeling to mitigate AAA formation. Panx1 expression is upregulated in human AAAs and retrospective clinical data demonstrated reduced mortality in aortic aneurysm patients treated with Panx1 inhibitors. Collectively, these data identify Panx1 signaling as a contributory mechanism of AAA formation. Pannexin-1 ion channels on endothelial cells regulate vascular inflammation and remodeling to mediate aortic aneurysm formation. Pharmacological blockade of Pannexin-1 channels may offer translational therapeutic mitigation of aneurysmal pathology.
Collapse
|
50
|
Erol A. Importance of Efferocytosis in COVID-19 Mortality. Infect Drug Resist 2022; 15:995-1007. [PMID: 35299855 PMCID: PMC8922362 DOI: 10.2147/idr.s348639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/03/2022] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is a generally benign coronavirus disease that can spread rapidly, except for those with a group of risk factors. Since the pathogenesis responsible for the severity of the disease has not been clearly revealed, effective treatment alternatives has not been developed. The hallmark of the SARS-CoV-2-infected cells is apoptosis. Apoptotic cells are cleared through a sterile process defined as efferocytosis by professional and nonprofessional phagocytic cells. The disease would be rapidly brought under control in the organism that can achieve effective efferocytosis, which is also a kind of innate immune response. In the risk group, the efferocytic process is defective. With the addition of the apoptotic cell load associated with SARS-COV-2 infection, failure to achieve efferocytosis of dying cells can initiate secondary necrosis, which is a highly destructive process. Uncontrolled inflammation and coagulation abnormalities caused by secondary necrosis reason in various organ failures, lung in particular, which are responsible for the poor prognosis. Following the short and simplified information, this opinion paper aims to present possible treatment options that can control the severity of COVID-19 by detailing the mechanisms that can cause defective efferocytosis.
Collapse
Affiliation(s)
- Adnan Erol
- Independent Researcher, Not Affiliated to Any Institution, Silivri-Istanbul, Turkey
| |
Collapse
|