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
|
Cen YY, Gao XL, Feng YH, Zhou C, Li CJ, Liu F, Shen JF, Zhang YY. The Double-Edged Effect of Connexins and Pannexins of Glial Cells in Central and Peripheral Nervous System After Nerve Injury. Mol Neurobiol 2025:10.1007/s12035-025-04991-6. [PMID: 40310549 DOI: 10.1007/s12035-025-04991-6] [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: 01/25/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
Glial cells play pivotal roles in homeostatic regulation and driving reactive pathologic changes after nerve injury. Connexins (Cxs) and pannexins (Panxs) have emerged as seminal proteins implicated in cell-cell communication, exerting a profound impact on the response processes of glial cell activation, proliferation, protein synthesis and secretion, as well as apoptosis following nerve injury. These influences are mediated through various forms, including protein monomers, hemichannel (HC), and gap junction (GJ), mainly by regulating intercellular or intracellular signaling pathways. Multiple Cx and Panx isoforms have been detected in central nervous system (CNS) or peripheral nervous system (PNS). Each isoform exhibits distinct cellular and subcellular localization, and the differential regulation and functional roles of various protein isoforms are observed post-injury. The quantitative and functional alterations of the same protein isoform in different studies remain inconsistent, attributable to factors such as the predominant mode of protein polymerization, the specific injury model, and the injury site. Similarly, the same protein isoforms have different roles in regulating the response processes after nerve injury, thus exerting a double-edged sword effect. This review describes the regulatory mechanisms and bidirectional effects of Cxs and Panxs. Additionally, it surveys the current status of research and application of drugs as therapeutic targets for neuropathic injuries. We summarize comprehensive and up-to-date information on these proteins in the glial cell response to nerve injury, providing new perspectives for future mechanistic exploration and development of targeted therapeutic approaches.
Collapse
Affiliation(s)
- Yue-Yan Cen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Xin-Lin Gao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Yu-Heng Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China
| | - Chun-Jie Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
| |
Collapse
|
3
|
Harris AL. PANX1 hexamers work but cells prefer heptamers. J Gen Physiol 2025; 157:e202413727. [PMID: 39907692 PMCID: PMC11797006 DOI: 10.1085/jgp.202413727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025] Open
Abstract
Gupta et al. (https://doi.org/10.1085/jgp.202413676) reconcile a disconnect between structural and functional data regarding stoichiometry of PANX1 channels and provide new insights about channel activation.
Collapse
Affiliation(s)
- Andrew L. Harris
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
4
|
Xu S, Li H, Gao Y, Wang Y, Zhu B, Shi H, Wang J, Wu X, Wang Y, Shi B, Su Z, Zhang Y, Peng Z, Yu X. Blocking donor liver Pannexin 1 channels facilitates mitochondria protection during liver transplantation. Am J Transplant 2025; 25:489-500. [PMID: 39481542 DOI: 10.1016/j.ajt.2024.10.021] [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: 05/18/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024]
Abstract
Static cold storage (SCS) is the standard technique for organ preservation during transplantation, resulting in cold ischemic injury. Hypoxia can induce pannexin 1 (Panx1) channels to open, leading to release of adenosine triphosphate. However, it is unknown if Panx1 plays a role in SCS. In this study, livers from Panx1-/- mice exhibited reduced adenosine triphosphate release, resulting in hepatocyte protection during preservation. The donor liver damage was decreased during SCS when Panx1 activity was blocked. Transmission electron microscopy revealed a decrease in mitochondria-associated endoplasmic reticulum membranes and improved mitochondria morphology. Mechanistically, Panx1 blockade upregulated the phosphatidylinositol 3-kinase-protein kinase B pathway and increased B cell leukemia/lymphoma 2 levels to combat apoptosis during liver preservation. The data indicate that blocking Panx1 during preservation of the donor liver can effectively improve mitochondrial function and reduce cellular stress damage thereby decreasing cold ischemia and reperfusion-related injuries in liver transplantation.
Collapse
Affiliation(s)
- Shiquan Xu
- Organ Transplantation Clinical Medical Center of Xiamen University, Department of Organ Transplantation, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Hao Li
- Organ Transplantation Clinical Medical Center of Xiamen University, Department of Organ Transplantation, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Yuxue Gao
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing, China
| | - Yaohui Wang
- School of Basic Medical Sciences, Henan University, Henan Province, China
| | - Bo Zhu
- Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen Key Laboratory of Genetic Testing, Fujian Province, Xiamen, China
| | - He Shi
- Orthopedics and Sports Medicine Center, Qingdao Municipal Hospital, Shandong, China
| | - Jie Wang
- Organ Transplantation Clinical Medical Center of Xiamen University, Department of Organ Transplantation, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Xia Wu
- Organ Transplantation Clinical Medical Center of Xiamen University, Department of Organ Transplantation, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Ying Wang
- Organ Transplantation Clinical Medical Center of Xiamen University, Department of Organ Transplantation, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Baojie Shi
- Organ Transplantation Clinical Medical Center of Xiamen University, Department of Organ Transplantation, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Zhaojie Su
- Organ Transplantation Clinical Medical Center of Xiamen University, Department of Organ Transplantation, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Yang Zhang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing, China.
| | - Zhihai Peng
- Organ Transplantation Clinical Medical Center of Xiamen University, Department of Organ Transplantation, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.
| | - Xiaoyu Yu
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
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
|
Tichauer JE, Rovegno M. Role of astrocytes connexins - pannexins in acute brain injury. Neurotherapeutics 2025; 22:e00523. [PMID: 39848901 PMCID: PMC11840357 DOI: 10.1016/j.neurot.2025.e00523] [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: 12/11/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025] Open
Abstract
Acute brain injuries (ABIs) encompass a broad spectrum of primary injuries such as ischemia, hypoxia, trauma, and hemorrhage that converge into secondary injury where some mechanisms show common determinants. In this regard, astroglial connexin and pannexin channels have been shown to play an important role. These channels are transmembrane proteins sharing similar topology and form gateways between adjacent cells named gap junctions (GJs) and pores into unopposed membranes named hemichannels (HCs). In astrocytes, GJs and HCs enable intercellular communication and have active participation in normal brain physiological processes, such as calcium waves, synapsis modulation, regional blood flow regulation, and homeostatic control of the extracellular environment, among others. However, after acute brain injury, astrocytes can change their phenotype and modify the activity of both channels and hemichannels, which can result in the amplification of danger signals, increased mediators of inflammation, and neuronal death, contributing to the expansion of brain damage and neurological deterioration. This is known as secondary brain damage. In this review, we discussed the main biological mechanism of secondary brain damage with a particular focus on astroglial connexin and pannexin participation during acute brain injuries.
Collapse
Affiliation(s)
- Juan E Tichauer
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile.
| | - Maximiliano Rovegno
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile.
| |
Collapse
|
7
|
Ehrlich A, Pelli G, Foglia B, Molica F, Kwak BR. Protective role of Pannexin1 in lymphatic endothelial cells in the progression of atherosclerosis in female mice. PLoS One 2024; 19:e0315511. [PMID: 39775604 PMCID: PMC11684638 DOI: 10.1371/journal.pone.0315511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Atherosclerosis is a progressive arterial disease arising from imbalanced lipid metabolism and a maladaptive immune response. The lymphatic system ensures tissue fluid homeostasis, absorption of dietary fats and trafficking of immune cells to draining lymph nodes, thereby potentially affecting atherogenesis. Endothelial cell-specific deletion of Pannexin1 (Panx1) in apolipoprotein E-deficient (Apoe-/-) mice increased atherosclerosis, suggesting a protective role for Panx1 channels in arterial endothelial function. Here, we investigated the role of Panx1 in lymphatic endothelial cells (LECs) in the initiation and the progression of atherosclerosis. Male or female Prox1-CreERT2+Panx1fl/flApoe-/- and Panx1fl/flApoe-/- mice were fed a high cholesterol diet (HCD) for 6 or 10 weeks. Tamoxifen-induced deletion of Panx1 was performed before or after 4 weeks of HCD. Body weight and serum lipid profiles were determined. The atherosclerotic plaque burden was assessed by Sudan-IV staining on thoracic-abdominal aortas and in aortic roots. Plaque composition was determined by immunohistochemistry. No differences in serum cholesterol, LDL and HDL were observed between genotypes and between sexes after HCD. Bodyweight, serum triglycerides and free fatty acid levels were higher before and after 6 weeks of HCD in male Prox1-CreERT2+Panx1fl/flApoe-/- and control Panx1fl/flApoe-/- mice compared to females of the same genotypes, which was associated with more lipids and inflammatory cells in their atherosclerotic plaques. In contrast, the atherosclerotic plaque burden was higher in female mice. The progression of atherosclerosis in male mice was not different between genotypes. However, female Prox1-CreERT2+Panx1fl/flApoe-/- mice showed enhanced progression of atherosclerosis compared to Panx1fl/flApoe-/- controls of the same sex. In addition, atherosclerotic lesions in female, but not in male, Prox1-CreERT2+Panx1fl/flApoe-/- mice showed T cell enrichment. Altogether, our results reveal differential sex-dependent effects of Panx1 in lymphatic endothelium on the progression of atherosclerosis.
Collapse
Affiliation(s)
- Avigail Ehrlich
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Graziano Pelli
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Bernard Foglia
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Filippo Molica
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Brenda R. Kwak
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
8
|
Ma Q, Cai L, Zhou Y, Zhang C. HuMSC-EVs Protect Endothelial Cells Against Hypoxia/Reoxygenation Injury by Inhibiting the Pannexin 1/p38-MAPK Pathway. Transplant Proc 2024; 56:1659-1664. [PMID: 39147615 DOI: 10.1016/j.transproceed.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024]
Abstract
Vascular endothelial cell dysfunction plays an important role in myocardial ischemia-reperfusion (I/R) injury, and pannexin 1 (Panx1), an ATP-permeable channel, is closely associated with the pathophysiological processes of I/R injury. The purpose of this study was to investigate the protective effects of human umbilical cord mesenchymal stromal cell-derived extracellular vesicles (HuMSC-EVs) and the underlying mechanism in a model of I/R injury. For the cellular model of I/R injury, human umbilical vein endothelial cells (HuVECs) were exposed to hypoxia/reoxygenation (H/R) conditions. The model cells were then treated with HuMSC-EVs, and the effects on cell survival and specific signaling activities were observed. The results showed that after H/R exposure, Panx1 expression and other markers of cellular damage were increased in HuVECs. However, treatment with HuMSC-EVs inhibited the H/R-induced increase in Panx1 expression and improved HuVEC survival. Mechanistically, HuMSC-EVs were found to inhibit the p38 mitogen-activated protein kinase (MAPK)-dependent apoptosis pathway, as evidenced by increased Bcl2 expression and reductions in p38 MAPK phosphorylation, Bax expression, and cleaved-caspase 3 expression. Together our data suggest that HuMSC-EVs alleviate H/R-induced apoptosis among HuVECs by inhibiting activity of the Panx1/p38-MAPK-dependent apoptosis pathway.
Collapse
Affiliation(s)
- Qian Ma
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Liwei Cai
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yu Zhou
- Department of Neurology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Changyi Zhang
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
9
|
Dalkara T, Kaya Z, Erdener ŞE. Unraveling the interplay of neuroinflammatory signaling between parenchymal and meningeal cells in migraine headache. J Headache Pain 2024; 25:124. [PMID: 39080518 PMCID: PMC11290240 DOI: 10.1186/s10194-024-01827-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The initiation of migraine headaches and the involvement of neuroinflammatory signaling between parenchymal and meningeal cells remain unclear. Experimental evidence suggests that a cascade of inflammatory signaling originating from neurons may extend to the meninges, thereby inducing neurogenic inflammation and headache. This review explores the role of parenchymal inflammatory signaling in migraine headaches, drawing upon recent advancements. BODY: Studies in rodents have demonstrated that sterile meningeal inflammation can stimulate and sensitize meningeal nociceptors, culminating in headaches. The efficacy of relatively blood-brain barrier-impermeable anti-calcitonin gene-related peptide antibodies and triptans in treating migraine attacks, both with and without aura, supports the concept of migraine pain originating in meninges. Additionally, PET studies utilizing inflammation markers have revealed meningeal inflammatory activity in patients experiencing migraine with aura, particularly over the occipital cortex generating visual auras. The parenchymal neuroinflammatory signaling involving neurons, astrocytes, and microglia, which eventually extends to the meninges, can link non-homeostatic perturbations in the insensate brain to pain-sensitive meninges. Recent experimental research has brought deeper insight into parenchymal signaling mechanisms: Neuronal pannexin-1 channels act as stress sensors, initiating the inflammatory signaling by inflammasome formation and high-mobility group box-1 release in response to transient perturbations such as cortical spreading depolarization (CSD) or synaptic metabolic insufficiency caused by transcriptional changes induced by migraine triggers like sleep deprivation and stress. After a single CSD, astrocytes respond by upregulating the transcription of proinflammatory enzymes and mediators, while microglia are involved in restoring neuronal structural integrity; however, repeated CSDs may prompt microglia to adopt a pro-inflammatory state. Transcriptional changes from pro- to anti-inflammatory within 24 h may serve to dampen the inflammatory signaling. The extensive coverage of brain surface and perivascular areas by astrocyte endfeet suggests their role as an interface for transporting inflammatory mediators to the cerebrospinal fluid to contribute to meningeal nociception. CONCLUSION We propose that neuronal stress induced by CSD or synaptic activity-energy mismatch may initiate a parenchymal inflammatory signaling cascade, transmitted to the meninges, thereby triggering lasting headaches characteristic of migraine, with or without aura. This neuroinflammatory interplay between parenchymal and meningeal cells points to the potential for novel targets for migraine treatment and prophylaxis.
Collapse
Affiliation(s)
- Turgay Dalkara
- Departments of Neuroscience and, Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey.
| | - Zeynep Kaya
- Department of Neurology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Şefik Evren Erdener
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
10
|
Fu S, Liu S, Li J, Dong Q, Fu Y, Luo R, Sun Y, Tian X, Liu W, Zong B, Ye C, Lu Q, Qiu Y, Guo L. Baicalin and probenecid protect against Glaesserella parasuis challenge in a piglet model. Vet Res 2024; 55:96. [PMID: 39075542 PMCID: PMC11285411 DOI: 10.1186/s13567-024-01352-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
Glaesserella parasuis (G. parasuis) induces vascular damage and systemic inflammation. However, the mechanism by which it causes vascular damage is currently unclear. Baicalin has important anti-inflammatory, antibacterial and immunomodulatory functions. In this study, we explored the ability of baicalin and probenecid to protect against G. parasuis challenge in a piglet model. Sixty piglets were randomly divided into a control group; an infection group; a probenecid group; and 25 mg/kg, 50 mg/kg and 100 mg/kg baicalin groups. The probenecid group and the 25 mg/kg, 50 mg/kg and 100 mg/kg baicalin groups were injected intramuscularly with 20 mg/kg body weight (BW) probenecid and 25 mg/kg BW, 50 mg/kg BW and 100 mg/kg BW baicalin, respectively. All piglets except those from the control group were injected intraperitoneally with 1 × 108 CFU of G. parasuis. The control group was injected intraperitoneally with TSB. The results showed baicalin and probenecid protected piglets against G. parasuis challenge, improved body weight and decreased temperature changes in piglets. Baicalin and probenecid attenuated IL-1β, IL-10, IL-18, TNF-α and IFN-γ mRNA levels in the blood for 48 h, inhibited the production of the nucleosides ATP, ADP, AMP and UMP from 24 to 72 h, reduced Panx-1/P2Y6/P2X7 expression, weakened NF-kB, AP-1, NLRP3/Caspase-1 and ROCK/MLCK/MLC signalling activation, and upregulated VE-cadherin expression in the blood vessels of piglets challenged with G. parasuis. Baicalin and probenecid alleviated pathological tissue damage in piglets induced by G. parasuis. Our results might provide a promising strategy to control and treat G. parasuis infection in the clinical setting.
Collapse
Affiliation(s)
- Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Siyu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Jingyang Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Qiaoli Dong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Yunjian Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Ronghui Luo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Yamin Sun
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Xinyue Tian
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Wei Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Bingbing Zong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Qirong Lu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China.
| | - Ling Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China.
| |
Collapse
|
11
|
Huang Y, Shi Y, Wang M, Liu B, Chang X, Xiao X, Yu H, Cui X, Bai Y. Pannexin1 Channel-Mediated Inflammation in Acute Ischemic Stroke. Aging Dis 2024; 15:1296-1307. [PMID: 37196132 PMCID: PMC11081155 DOI: 10.14336/ad.2023.0303] [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: 12/29/2022] [Accepted: 03/03/2023] [Indexed: 05/19/2023] Open
Abstract
Emerging evidence suggests that inflammation mediated by the pannexin1 channel contributes significantly to acute ischemic stroke. It is believed that the pannexin1 channel is key in initiating central system inflammation during the early stages of acute ischemic stroke. Moreover, the pannexin1 channel is involved in the inflammatory cascade to maintain the inflammation levels. Specifically, the interaction of pannexin1 channels with ATP-sensitive P2X7 purinoceptors or promotion of potassium efflux mediates the activation of the NLRP3 inflammasome, triggering the release of pro-inflammatory factors such as IL-1 and IL-18, exacerbating and sustaining inflammation of brain. Also, increased release of ATP induced by cerebrovascular injury activates pannexin1 in vascular endothelial cells. This signal directs peripheral leukocytes to migrate into ischemic brain tissue, leading to an expansion of the inflammatory zone. Intervention strategies targeting pannexin1 channels may greatly alleviate inflammation after acute ischemic stroke to improve this patient population's clinical outcomes. In this review, we sought to summarize relevant studies on inflammation mediated by the pannexin1 channel in acute ischemic stroke and discussed the possibility of using brain organoid-on-a-chip technology to screen miRNAs that exclusively target the pannexin1 channel to provide new therapeutic measures for targeted regulation of pannexin1 channel to reduce inflammation in acute ischemic stroke.
Collapse
Affiliation(s)
- Yubing Huang
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Yutong Shi
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Mengmeng Wang
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Medical College, Institute of Microanalysis, Dalian University, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Bingyi Liu
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Xueqin Chang
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Xia Xiao
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Huihui Yu
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Xiaodie Cui
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Ying Bai
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| |
Collapse
|
12
|
Fu R, Zhao L, Guo Y, Qin X, Xu W, Cheng X, Zhang Y, Xu S. AIM2 inflammasome: A potential therapeutic target in ischemic stroke. Clin Immunol 2024; 259:109881. [PMID: 38142900 DOI: 10.1016/j.clim.2023.109881] [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: 09/28/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Ischemic stroke (IS) is a significant global public health issue with a high incidence, disability, and mortality rate. A robust inflammatory cascade with complex and wide-ranging mechanisms occurs following ischemic brain injury. Inflammasomes are multiprotein complexes in the cytoplasm that modulate the inflammatory response by releasing pro-inflammatory cytokines and inducing cellular pyroptosis. Among these inflammasomes, the Absent in Melanoma 2 (AIM2) inflammasome shows the ability to detect a wide range of pathogen DNAs, thereby triggering an inflammatory response. Recent studies have indicated that the aberrant expression of AIM2 inflammasome in various cells is closely associated with the pathological processes of ischemic brain injury. This paper summarizes the expression and regulatory role of AIM2 in CNS and peripheral immune cells and discusses current therapeutic approaches targeting AIM2 inflammasome. These findings aim to serve as a reference for future research in this field.
Collapse
Affiliation(s)
- Rong Fu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Linna Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Yuying Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Xiaoli Qin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenzhe Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xueqi Cheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
13
|
Hernandez CA, Eugenin EA. The role of Pannexin-1 channels, ATP, and purinergic receptors in the pathogenesis of HIV and SARS-CoV-2. Curr Opin Pharmacol 2023; 73:102404. [PMID: 37734241 PMCID: PMC10838406 DOI: 10.1016/j.coph.2023.102404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/25/2023] [Indexed: 09/23/2023]
Abstract
Infectious agents such as human immune deficiency virus-1 (HIV) and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) use host proteins to infect, replicate, and induce inflammation within the host. A critical component of these diseases is the axis between pannexin-1 channels, extracellular ATP, and purinergic receptors. Here, we describe the potential therapeutic role of Pannexin-1/purinergic approaches to prevent or reduce the devastating consequences of these pathogens.
Collapse
Affiliation(s)
- Cristian A Hernandez
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Eliseo A Eugenin
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA.
| |
Collapse
|
14
|
Rusiecka OM, Molica F, Nielsen MS, Tollance A, Morel S, Frieden M, Chanson M, Boengler K, Kwak BR. Mitochondrial pannexin1 controls cardiac sensitivity to ischaemia/reperfusion injury. Cardiovasc Res 2023; 119:2342-2354. [PMID: 37556386 PMCID: PMC10597630 DOI: 10.1093/cvr/cvad120] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 04/27/2023] [Accepted: 05/18/2023] [Indexed: 08/11/2023] Open
Abstract
AIMS No effective therapy is available in clinics to protect the heart from ischaemia/reperfusion (I/R) injury. Endothelial cells are activated after I/R, which may drive the inflammatory response by releasing ATP through pannexin1 (Panx1) channels. Here, we investigated the role of Panx1 in cardiac I/R. METHODS AND RESULTS Panx1 was found in cardiac endothelial cells, neutrophils, and cardiomyocytes. After in vivo I/R, serum Troponin-I, and infarct size were less pronounced in Panx1-/- mice, but leukocyte infiltration in the infarct area was similar between Panx1-/- and wild-type mice. Serum Troponin-I and infarct size were not different between mice with neutrophil-specific deletion of Panx1 and Panx1fl/fl mice, suggesting that cardioprotection by Panx1 deletion rather involved cardiomyocytes than the inflammatory response. Physiological cardiac function in wild-type and Panx1-/- hearts was similar. The time to onset of contracture and time to maximal contracture were delayed in Panx1-/- hearts, suggesting reduced sensitivity of these hearts to ischaemic injury. Moreover, Panx1-/- hearts showed better recovery of left ventricle developed pressure, cardiac contractility, and relaxation after I/R. Ischaemic preconditioning failed to confer further protection in Panx1-/- hearts. Panx1 was found in subsarcolemmal mitochondria (SSM). SSM in WT or Panx1-/- hearts showed no differences in morphology. The function of the mitochondrial permeability transition pore and production of reactive oxygen species in SSM was not affected, but mitochondrial respiration was reduced in Panx1-/- SSM. Finally, Panx1-/- cardiomyocytes had a decreased mitochondrial membrane potential and an increased mitochondrial ATP content. CONCLUSION Panx1-/- mice display decreased sensitivity to cardiac I/R injury, resulting in smaller infarcts and improved recovery of left ventricular function. This cardioprotective effect of Panx1 deletion seems to involve cardiac mitochondria rather than a reduced inflammatory response. Thus, Panx1 may represent a new target for controlling cardiac reperfusion damage.
Collapse
Affiliation(s)
- Olga M Rusiecka
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Filippo Molica
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Morten S Nielsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Axel Tollance
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sandrine Morel
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Maud Frieden
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marc Chanson
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Kerstin Boengler
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Brenda R Kwak
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| |
Collapse
|
15
|
Tien TY, Wu YJ, Su CH, Hsieh CL, Wang BJ, Lee YN, Su Y, Yeh HI. Pannexin 1 Modulates Angiogenic Activities of Human Endothelial Colony-Forming Cells Through IGF-1 Mechanism and Is a Marker of Senescence. Arterioscler Thromb Vasc Biol 2023; 43:1935-1951. [PMID: 37589139 DOI: 10.1161/atvbaha.123.319529] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND We examined the role of Panxs (pannexins) in human endothelial progenitor cell (EPC) senescence. METHODS Young and replication-induced senescent endothelial colony-forming cells (ECFCs) derived from human circulating EPCs were used to examine cellular activities and senescence-associated indicators after transfection of short interference RNA specific to Panx1 or lentivirus-mediated Panx1 overexpression. Hind limb ischemia mice were used as in vivo angiogenesis model. Protein and phospho-kinase arrays were used to determine underlying mechanisms. RESULTS Panx1 was the predominant Panx isoform in human ECFCs and upregulated in both replication-induced senescent ECFCs and circulating EPCs from aged mice and humans. Cellular activities of the young ECFCs were enhanced by Panx1 downregulation but attenuated by its upregulation. In addition, reduction of Panx1 in the senescent ECFCs could rejuvenate cellular activities with reduced senescence-associated indicators, including senescence-associated β-galactosidase activity, p16INK4a (cyclin-dependent kinase inhibitor 2A), p21 (cyclin-dependent kinase inhibitor 1), acetyl-p53 (tumor protein P53), and phospho-histone H2A.X (histone family member X). In mouse ischemic hind limbs injected senescent ECFCs, blood perfusion ratio, salvaged limb outcome, and capillary density were all improved by Panx1 knockdown. IGF-1 (insulin-like growth factor 1) was significantly increased in the supernatant from senescent ECFCs after Panx1 knockdown. The enhanced activities and paracrine effects of Panx1 knockdown senescent ECFCs were completely inhibited by anti-IGF-1 antibodies. FAK (focal adhesion kinase), ERK (extracellular signal-regulated kinase), and STAT3 (signal transducer and activator of transcription 3) were activated in senescent ECFCs with Panx1 knockdown, in which the intracellular calcium level was reduced, and the activation was inhibited by supplemented calcium. The increased IGF-1 in Panx1-knockdown ECFCs was abrogated, respectively, by inhibitors of FAK (PF562271), ERK (U0126), and STAT3 (NSC74859) and supplemented calcium. CONCLUSIONS Panx1 expression is upregulated in human ECFCs/EPCs with replication-induced senescence and during aging. Angiogenic potential of senescent ECFCs is improved by Panx1 reduction through increased IGF-1 production via activation of the FAK-ERK axis following calcium influx reduction. Our findings provide new strategies to evaluate EPC activities and rejuvenate senescent EPCs for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Ting-Yi Tien
- Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan (T.-Y.T., Y.S.)
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yih-Jer Wu
- Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-I.Y.)
| | - Cheng-Huang Su
- Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-I.Y.)
| | - Chin-Ling Hsieh
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Bo-Jeng Wang
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yi-Nan Lee
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yeu Su
- Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan (T.-Y.T., Y.S.)
| | - Hung-I Yeh
- Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-I.Y.)
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
Su L, Zhang J, Wang J, Wang X, Cao E, Yang C, Sun Q, Sivakumar R, Peng Z. Pannexin 1 targets mitophagy to mediate renal ischemia/reperfusion injury. Commun Biol 2023; 6:889. [PMID: 37644178 PMCID: PMC10465551 DOI: 10.1038/s42003-023-05226-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 08/07/2023] [Indexed: 08/31/2023] Open
Abstract
Renal ischemia/reperfusion (I/R) injury contributes to the development of acute kidney injury (AKI). Kidney is the second organ rich in mitochondrial content next to the heart. Mitochondrial damage substantially contributes for AKI development. Mitophagy eliminates damaged mitochondria from the cells to maintain a healthy mitochondrial population, which plays an important role in AKI. Pannexin 1 (PANX1) channel transmembrane proteins are known to drive inflammation and release of adenosine triphosphate (ATP) during I/R injury. However, the specific role of PANX1 on mitophagy regulation in renal I/R injury remains elusive. In this study, we find that serum level of PANX1 is elevated in patients who developed AKI after cardiac surgery, and the level of PANX1 is positively correlated with serum creatinine and urea nitrogen levels. Using the mouse model of renal I/R injury in vivo and cell-based hypoxia/reoxygenation (H/R) model in vitro, we prove that genetic deletion of PANX1 mitigate the kidney tubular cell death, oxidative stress and mitochondrial damage after I/R injury through enhanced mitophagy. Mechanistically, PANX1 disrupts mitophagy by influencing ATP-P2Y-mTOR signal pathway. These observations provide evidence that PANX1 could be a potential biomarker for AKI and a therapeutic target to alleviate AKI caused by I/R injury.
Collapse
Affiliation(s)
- Lianjiu Su
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China.
- Department of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Jiahao Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Jing Wang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Xiaozhan Wang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Edward Cao
- Department of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Chen Yang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Qihao Sun
- Department of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ramadoss Sivakumar
- Department of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China.
- Center of Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15206, USA.
| |
Collapse
|
20
|
Kahan R, Cray PL, Abraham N, Gao Q, Hartwig MG, Pollara JJ, Barbas AS. Sterile inflammation in liver transplantation. Front Med (Lausanne) 2023; 10:1223224. [PMID: 37636574 PMCID: PMC10449546 DOI: 10.3389/fmed.2023.1223224] [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: 05/15/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Sterile inflammation is the immune response to damage-associated molecular patterns (DAMPs) released during cell death in the absence of foreign pathogens. In the setting of solid organ transplantation, ischemia-reperfusion injury results in mitochondria-mediated production of reactive oxygen and nitrogen species that are a major cause of uncontrolled cell death and release of various DAMPs from the graft tissue. When properly regulated, the immune response initiated by DAMP-sensing serves as means of damage control and is necessary for initiation of recovery pathways and re-establishment of homeostasis. In contrast, a dysregulated or overt sterile inflammatory response can inadvertently lead to further injury through recruitment of immune cells, innate immune cell activation, and sensitization of the adaptive immune system. In liver transplantation, sterile inflammation may manifest as early graft dysfunction, acute graft failure, or increased risk of immunosuppression-resistant rejection. Understanding the mechanisms of the development of sterile inflammation in the setting of liver transplantation is crucial for finding reliable biomarkers that predict graft function, and for development of therapeutic approaches to improve long-term transplant outcomes. Here, we discuss the recent advances that have been made to elucidate the early signs of sterile inflammation and extent of damage from it. We also discuss new therapeutics that may be effective in quelling the detrimental effects of sterile inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andrew S. Barbas
- Duke Ex-Vivo Organ Lab (DEVOL)—Division of Abdominal Transplant Surgery, Duke University, Durham, NC, United States
| |
Collapse
|
21
|
Koval M, Schug WJ, Isakson BE. Pharmacology of pannexin channels. Curr Opin Pharmacol 2023; 69:102359. [PMID: 36858833 PMCID: PMC10023479 DOI: 10.1016/j.coph.2023.102359] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/18/2023] [Accepted: 01/29/2023] [Indexed: 03/02/2023]
Abstract
Pannexin channels play fundamental roles in regulating inflammation and have been implicated in many diseases including hypertension, stroke, and neuropathic pain. Thus, the ability to pharmacologically block these channels is a vital component of several therapeutic approaches. Pharmacologic interrogation of model systems also provides a means to discover new roles for pannexins in cell physiology. Here, we review the state of the art for agents that can be used to block pannexin channels, with a focus on chemical pharmaceuticals and peptide mimetics that act on pannexin 1. Guidance on interpreting results obtained with pannexin pharmacologics in experimental systems is discussed, as well as strengths and caveats of different agents, including specificity and feasibility of clinical application.
Collapse
Affiliation(s)
- Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Wyatt J Schug
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
22
|
Onódi Z, Koch S, Rubinstein J, Ferdinandy P, Varga ZV. Drug repurposing for cardiovascular diseases: New targets and indications for probenecid. Br J Pharmacol 2023; 180:685-700. [PMID: 36484549 DOI: 10.1111/bph.16001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/12/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
The available pharmacological options in the management of cardiovascular diseases such as ischaemic heart disease and subsequent heart failure are effective in slowing the progression of this condition. However, the long-term prognosis is still poor, raising the demand for new therapeutic strategies. Drug repurposing is a time- and cost-effective drug development strategy that offers approved and abandoned drugs a new chance for new indications. Recently, drugs used for the management of gout-related inflammation such as canakinumab or colchicine have been considered for drug repurposing in cardiovascular indications. The old uricosuric drug, probenecid, has been identified as a novel therapeutic option in the management of specific cardiac diseases as well. Probenecid can modulate myocardial contractility and vascular tone and exerts anti-inflammatory properties. The mechanisms behind these beneficial effects might be related inhibition of inflammasomes, and to modulation purinergic-pannexin-1 signalling and TRPV2 channels, which are recently identified molecular targets of probenecid. In this review, we provide an overview on repurposing probenecid for ischaemic heart disease and subsequent heart failure by summarizing the related experimental and clinical data and propose its potential repurposing to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Sheryl Koch
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| |
Collapse
|
23
|
Mendes CE, Palombit K, Alves Pereira TT, Riceti Magalhães HI, Ferreira Caetano MA, Castelucci P. Effects of probenecid and brilliant blue G on rat enteric glial cells following intestinal ischemia and reperfusion. Acta Histochem 2023; 125:151985. [PMID: 36495673 DOI: 10.1016/j.acthis.2022.151985] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022]
Abstract
The P2X7 receptor participates in several intracellular events and acts with the pannexin-1 channel. This study examined the effects of probenecid (PB) and brilliant blue G (BBG), which are antagonists of the pannexin-1 channel and P2X7 receptor, respectively, on rat ileum enteric glial cells after on ischemia and reperfusion. The ileal vessels were occluded for 45 min with nontraumatic vascular tweezers, and reperfusion was performed for periods of 24 h and 14 and 28 days. After ischemia (IR groups), the animals were treated with BBG (BG group) or PB (PB group). The double-labeling results demonstrated the following: the P2X7 receptor was present in enteric glial cells (S100β) and enteric neurons positive for HuC/D; enteric glial cells exhibited different phenotypes; some enteric glial cells were immunoreactive to only S100β or GFAP; and the pannexin-1 channel was present in enteric glial cells (GFAP). Density (in cells/cm2) analyses showed that the IR group exhibited a decrease in the number of cells immunoreactive for the P2X7 receptor, pannexin-1, and HuC/D and that treatment with BBG or PB resulted in the recovery of the numbers of these cells. The number of glial cells (S100β and GFAP) was higher in the IR group, and the treatments decreased the number of these cells to the normal value. However, the PB group did not exhibit recovery of S100β-positive glia. The cell profile area (μm2) of S100β-positive enteric glial cells decreased to the normal value after BBG treatment, whereas no recovery was observed in the PB group. The ileum contractile activity was decreased in the IR group and returned to baseline in the BG and PB groups. BBG and PB can effectively induce the recovery of neurons and glia cells and are thus potential therapeutic agents in the treatment of gastrointestinal tract diseases.
Collapse
Affiliation(s)
| | - Kelly Palombit
- Department of Morphology, University Federal of Piaui, Brazil
| | | | | | | | | |
Collapse
|
24
|
Metz LM, Feige T, de Biasi L, Ehrenberg A, Mulorz J, Toska LM, Reusswig F, Quast C, Gerdes N, Kelm M, Schelzig H, Elvers M. Platelet pannexin-1 channels modulate neutrophil activation and migration but not the progression of abdominal aortic aneurysm. Front Mol Biosci 2023; 10:1111108. [PMID: 36950521 PMCID: PMC10025481 DOI: 10.3389/fmolb.2023.1111108] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/23/2023] [Indexed: 03/08/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a common disease and highly lethal if untreated. The progressive dilatation of the abdominal aorta is accompanied by degradation and remodeling of the vessel wall due to chronic inflammation. Pannexins represent anion-selective channels and play a crucial role in non-vesicular ATP release to amplify paracrine signaling in cells. Thus, pannexins are involved in many (patho-) physiological processes. Recently, Panx1 channels were identified to be significantly involved in abdominal aortic aneurysm formation through endothelial derived Panx1 regulated inflammation and aortic remodeling. In platelets, Panx1 becomes activated following activation of glycoprotein (GP) VI. Since platelets play a role in cardiovascular diseases including abdominal aortic aneurysm, we analyzed the contribution of platelet Panx1 in the progression of abdominal aortic aneurysm. We detected enhanced Panx1 plasma levels in abdominal aortic aneurysm patients. In experimental abdominal aortic aneurysm using the pancreatic porcine elastase (PPE) mouse model, a major contribution of platelet Panx1 channels in platelet activation, pro-coagulant activity of platelets and platelet-mediated inflammation has been detected. In detail, platelets are important for the migration of neutrophils into the aortic wall induced by direct cell interaction and by activation of endothelial cells. Decreased platelet activation and inflammation did not affect ECM remodeling or wall thickness in platelet-specific Panx1 knock-out mice following PPE surgery. Thus, aortic diameter expansion at different time points after elastase infusion of the aortic wall was unaltered in platelet-specific Panx1 deficient mice suggesting that the modulation of inflammation alone does not affect abdominal aortic aneurysm formation and progression. In conclusion, our data strongly supports the role of platelets in inflammatory responses in abdominal aortic aneurysm via Panx1 channels and adds important knowledge about the significance of platelets in abdominal aortic aneurysm pathology important for the establishment of an anti-platelet therapy for abdominal aortic aneurysm patients.
Collapse
Affiliation(s)
- Lisa Maria Metz
- Department of Vascular- and Endovascular Surgery, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Tobias Feige
- Department of Vascular- and Endovascular Surgery, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Larissa de Biasi
- Department of Vascular- and Endovascular Surgery, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Agnes Ehrenberg
- Department of Vascular- and Endovascular Surgery, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Joscha Mulorz
- Department of Vascular- and Endovascular Surgery, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Laura Mara Toska
- Department of Vascular- and Endovascular Surgery, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Friedrich Reusswig
- Department of Vascular- and Endovascular Surgery, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Christine Quast
- Department of Cardiology, Pulmonology and Vascular Medicine, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Norbert Gerdes
- Department of Cardiology, Pulmonology and Vascular Medicine, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Hubert Schelzig
- Department of Vascular- and Endovascular Surgery, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Margitta Elvers
- Department of Vascular- and Endovascular Surgery, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
- *Correspondence: Margitta Elvers,
| |
Collapse
|
25
|
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
|
26
|
Vinken M. Toxic talk: pannexin1 channel communication as an emerging mechanism of toxicity. Toxicology 2022; 478:153295. [PMID: 35998787 DOI: 10.1016/j.tox.2022.153295] [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: 07/11/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 10/15/2022]
Abstract
Pannexin1 channels facilitate the extracellular release of a number of messengers, including adenosine triphosphate. Although fulfilling some physiological functions, pannexin1 channel communication has to date been primarily studied in the context of inflammation and cell death. In the past decade, a variety of chemical substances have been reported to induce pannexin1 channel opening, including metals, chelating agents, particulate matter, nanoparticles and drugs. While the pathophysiological aspects of pannexin1 channel communication have been reviewed on many previous occasions, the present paper intends to provide a short perspective in order to motivate research that will advance mechanistic understanding of the roles of pannexin1 signaling in chemical toxicity.
Collapse
Affiliation(s)
- Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
27
|
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
|
28
|
Zheng Y, Tang W, Zeng H, Peng Y, Yu X, Yan F, Cao S. Probenecid-Blocked Pannexin-1 Channel Protects Against Early Brain Injury via Inhibiting Neuronal AIM2 Inflammasome Activation After Subarachnoid Hemorrhage. Front Neurol 2022; 13:854671. [PMID: 35401398 PMCID: PMC8983901 DOI: 10.3389/fneur.2022.854671] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Aim Previous studies have proved that inhibiting inflammasome activation provides neuroprotection against early brain injury (EBI) after subarachnoid hemorrhage (SAH), which is mainly focused on the microglial inflammatory response, but the potential role of neuronal inflammasome activation in EBI has not been clearly identified. This study examined whether the pannexin-1 channel inhibitor probenecid could reduce EBI after SAH by inhibiting neuronal AIM2 inflammasome activation. Methods There are in vivo and in vitro parts in this study. First, adult male SD rats were subjected to the endovascular perforation mode of SAH. The time course of pannexin-1 and AIM2 expressions were determined after SAH in 72 h. Brain water content, neurological function, AIM2 inflammasome activation, and inflammatory response were evaluated at 24 h after SAH in sham, SAH, and SAH + probenecid groups. In the in vitro part, HT22 cell treated with hemin was applied to mimic SAH. The expression of AIM2 inflammasome was detected by immunofluorescence staining. Neuronal death and mitochondrial dysfunction were determined by the LDH assay kit and JC-1 staining. Results The pannexin-1 and AIM2 protein levels were upregulated after SAH. Pannexin-1 channel inhibitor probenecid attenuated brain edema and improved neurological dysfunction by reducing AIM2 inflammasome activation and reactive oxygen species (ROS) generation after SAH in rats. Treating HT22 cells with hemin for 12 h resulted in AIM2 and caspase-1 upregulation and increased mitochondrial dysfunction and neuronal cell death. Probenecid significantly attenuated the hemin-induced AIM2 inflammasome activation and neuronal death. Conclusions AIM2 inflammasome is activated in neurons after SAH. Pharmacological inhibition of the pannexin-1 channel by probenecid attenuated SAH-induced AIM2 inflammasome activation and EBI in vivo and hemin-induced AIM2 inflammasome activation and neuronal death in vitro.
Collapse
Affiliation(s)
- Yonghe Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wenwen Tang
- Zhejiang University School of Medicine, Hangzhou, China
| | - Hanhai Zeng
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yucong Peng
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaobo Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shenglong Cao
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|