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
|
Illanes-González J, Flores-Muñoz C, Vitureira N, Ardiles ÁO. Pannexin 1 channels: A bridge between synaptic plasticity and learning and memory processes. Neurosci Biobehav Rev 2025; 174:106173. [PMID: 40274202 DOI: 10.1016/j.neubiorev.2025.106173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/13/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
The Pannexin 1 channel is a membrane protein widely expressed in various vertebrate cell types, including microglia, astrocytes, and neurons within the central nervous system. Growing research has demonstrated the significant involvement of Panx1 in synaptic physiology, such as its contribution to long-term synaptic plasticity, with a particular focus on the hippocampus, an essential structure for learning and memory. Investigations studying the role of Panx1 in synaptic plasticity have utilized knockout animal models and channel inhibition techniques, revealing that the absence or blockade of Panx1 channels in this region promotes synaptic potentiation, dendritic arborization, and spine formation. Despite substantial progress, the precise mechanism by which Panx1 regulates synaptic plasticity remains to be determined. Nevertheless, evidence suggests that Panx1 may exert its influence by releasing signaling molecules, such as adenosine triphosphate (ATP), or through the clearance of endocannabinoids (eCBs). This review aims to comprehensively explore the current literature on the role of Panx1 in synapses. By examining relevant articles, we seek to enhance our understanding of Panx1's contribution to synaptic fundamental processes and the potential implications for cognitive function.
Collapse
Affiliation(s)
- Javiera Illanes-González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; Centro para la Investigación Traslacional en Neurofarmacología, CItNe, Universidad de Valparaíso, Valparaíso, Chile
| | - Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; Centro para la Investigación Traslacional en Neurofarmacología, CItNe, Universidad de Valparaíso, Valparaíso, Chile
| | - Nathalia Vitureira
- Unidad Académica de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Álvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; Centro para la Investigación Traslacional en Neurofarmacología, CItNe, Universidad de Valparaíso, Valparaíso, Chile; Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
3
|
Meng Z, Huang H, Guo J, Wang D, Tao X, Dai Q, Bai Y, Ma C, Huang L, Fu Y, Lu C, Wang H, Wang Q, Li X, Ren H. Promote Sepsis Recovery through the Inhibition of Immunothrombosis via a Combination of Probenecid Nanocrystals and Cefotaxime Sodium. ACS APPLIED MATERIALS & INTERFACES 2025; 17:21013-21032. [PMID: 40152149 DOI: 10.1021/acsami.5c05609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Sepsis is a life-threatening organ dysfunction syndrome caused by a dysregulated host immune response to pathogenic infection. Due to its high mortality rate, it has been a major global public health problem. Recent studies have shown that the formation of immunothrombosis plays as a "double-edged sword" in the pathogenesis of sepsis, and how to properly regulate immunothrombosis to avoid organ damage and end the high-inflammation state as early as possible are the key steps for sepsis therapy. Considering the complexity of sepsis therapy, the development of an effective combined therapeutic strategy is the goal of this study. First, the insoluble Panexin1 (Panx1) channel inhibitor probenecid (Prob) was prepared as nanocrystals and administered via intramuscular injection. At the same time, septic mice were intravenously injected with cefotaxime sodium through the tail vein for combination therapy. After treatment, the number of infection foci and the level of serum inflammatory factors in septic mice were significantly reduced, and also neutrophil NETosis was significantly inhibited; thus, the survival rate of septic mice was dramatically increased. Pathological analysis revealed that the combination treatment was safe and effective and could significantly reduce the formation of immunothrombosis in septic mice.
Collapse
Affiliation(s)
- Zhengjie Meng
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Haixiao Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Jiaqi Guo
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Dong Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Xinyue Tao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Qihao Dai
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Yunhao Bai
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Chenyu Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Luming Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Yangkai Fu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Chenyu Lu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Hengjian Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Qiyue Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Xueming Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Hao Ren
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| |
Collapse
|
4
|
Sayedyahossein S, Huang K, Zhang C, Karimi M, Bahmani M, O’Donnell BL, Wakefield B, Li Z, Johnston D, Leighton SE, Huver MS, Dagnino L, Sacks DB, Penuela S. Pannexin 1 crosstalk with the Hippo pathway in malignant melanoma. FEBS J 2025; 292:1633-1653. [PMID: 39786847 PMCID: PMC11971028 DOI: 10.1111/febs.17396] [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: 02/22/2024] [Revised: 08/19/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
In this study, we explored the intricate relationship between Pannexin 1 (PANX1) and the Hippo signaling pathway effector, Yes-associated protein (YAP). Analysis of The Cancer Genome Atlas (TCGA) data revealed a significant positive correlation between PANX1 mRNA and core Hippo components, Yes-associated protein 1 [YAP], Transcriptional coactivator with PDZ-binding motif [TAZ], and Hippo scaffold, Ras GTPase-activating-like protein IQGAP1 [IQGAP1], in invasive cutaneous melanoma and breast carcinoma. Furthermore, we demonstrated that PANX1 expression is upregulated in invasive melanoma cell lines and is associated with increased YAP protein levels. Notably, our investigations uncovered a previously unrecognized interaction between endogenous PANX1 and the Hippo scaffold protein IQGAP1 in melanoma cells. Moreover, our findings revealed that IQGAP1 exhibits differential expression in melanoma cells and plays a regulatory role in cellular morphology. Functional studies involving PANX1 knockdown provided compelling evidence that PANX1 modulates YAP protein levels and its cotranscriptional activity in melanoma and breast carcinoma cells. Importantly, our study highlights the potential therapeutic significance of targeting PANX1. Pharmacological inhibition of PANX1 using selective FDA-approved inhibitors or PANX1 knockdown reduced YAP levels in melanoma cells. Furthermore, our Clariom™ S analysis unveiled key genes implicated in cell proliferation, such as neuroglin1 (NRG1), β-galactoside binding protein and galectin-3 (LGALS3), that are affected in PANX1-deficient cells. In summary, our investigation delves into the intricate interplay between PANX1 and YAP in the context of invasive melanoma, offering valuable insights into potential therapeutic strategies for effective treatment.
Collapse
Affiliation(s)
- Samar Sayedyahossein
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Kenneth Huang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Christopher Zhang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Mehdi Karimi
- Department of Mathematics, Illinois State University, Normal, Illinois, USA, 61790
| | | | - Brooke L. O’Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Brent Wakefield
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Zhigang Li
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
| | - Danielle Johnston
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Stephanie E. Leighton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Matthew S. Huver
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - David B. Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| |
Collapse
|
5
|
Li K, Jan YN. Experimental tools and emerging principles of organellar mechanotransduction. Trends Cell Biol 2025:S0962-8924(24)00279-4. [PMID: 39828483 DOI: 10.1016/j.tcb.2024.12.011] [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: 10/15/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
Mechanotransduction is the process by which cells detect mechanical forces and convert them into biochemical or electrical signals. This process occurs across various cellular compartments, including the plasma membrane, cytoskeleton, and intracellular organelles. While research has focused mainly on force sensing at the plasma membrane, the mechanisms and significance of intracellular mechanotransduction are just beginning to be understood. This review summarizes current techniques for studying organellar mechanobiology, and highlights advances in our understanding of the mechanosensitive events occurring in organelles such as the endoplasmic reticulum (ER), Golgi apparatus, and endolysosomes. Additionally, some open questions and promising directions are identified for future research.
Collapse
Affiliation(s)
- Kai Li
- Institute for Medical Physiology, Chinese Institutes for Medical Research, Beijing, 100069, China; School of Basic Medicine, Capital Medical University, Beijing, 100069, China
| | - Yuh Nung Jan
- Department of Physiology, University of California at San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
6
|
Guan X, Li H, Zhang L, Zhi H. Mechanisms of mitochondrial damage-associated molecular patterns associated with inflammatory response in cardiovascular diseases. Inflamm Res 2025; 74:18. [PMID: 39806203 DOI: 10.1007/s00011-025-01993-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: 10/20/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Cardiovascular diseases (CVDs) continue to be a substantial global healthcare burden despite considerable progress in therapies. The inflammatory response during the progression of CVD has attracted considerable attention. Mitochondria serve as the principal energy source for the heart. In cardiovascular illnesses, mitochondrial homeostasis is disrupted, accompanied by structural and functional impairments. During mitochondrial stress or injury, mitochondrial damage-associated molecular patterns (mtDAMPs), such as mitochondrial DNA, cardiolipin, N-formyl peptide, and adenosine triphosphate, are released to activate pattern recognition receptors and trigger immunological responses. Inflammatory responses mediated by mtDAMPs substantially contribute to the pathophysiology of cardiovascular illnesses. In this review, we discuss the molecular mechanisms by which different mtDAMPs control the inflammatory response, address the pathological consequences of mtDAMPs in inducing or exacerbating the inflammatory response in CVDs, and summarize potential therapeutic targets in relevant experimental studies. Preventing or reducing mtDAMP release may play a role in CVD progression by alleviating the inflammatory response.
Collapse
Affiliation(s)
- Xiuju Guan
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Haitao Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China
| | - Lijuan Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China.
| | - Hongwei Zhi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China.
| |
Collapse
|
7
|
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
|
8
|
Nielsen AC, Anderson CL, Ens C, Boyce AKJ, Thompson RJ. Non-ionotropic NMDAR signalling activates Panx1 to induce P2X4R-dependent long-term depression in the hippocampus. J Physiol 2024. [PMID: 39709529 DOI: 10.1113/jp285193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 11/28/2024] [Indexed: 12/23/2024] Open
Abstract
In recent years, evidence supporting non-ionotropic signalling by the NMDA receptor (niNMDAR) has emerged, including roles in long-term depression (LTD). Here, we investigated whether niNMDAR-pannexin-1 (Panx1) contributes to LTD at the CA3-CA1 hippocampal synapse. Using whole-cell, patch clamp electrophysiology in rat hippocampal slices, we show that a low-frequency stimulation (3 Hz) of the Schaffer collaterals produces LTD that is blocked by continuous but not transient application of the NMDAR competitive antagonist, MK-801. After transient MK-801, LTD involved pannexin-1 and sarcoma (Src) kinase. We show that pannexin-1 is not permeable to Ca2+, but probably releases ATP to induce LTD via P2X4 purinergic receptors because LTD after transient MK-801 application was prevented by 5-BDBD. Thus, we conclude that niNMDAR activation of Panx1 can link glutamatergic and purinergic pathways to produce LTD following low frequency synaptic stimulation when NMDARs are transiently inhibited. KEY POINTS: Differential effect of short-term D-APV and MK-801 application on long-term depression (LTD) suggests that the NMDA receptor (niNMDAR) contributes to later phases of synaptic depression. niNMDAR LTD involved sarcoma (Src) kinase and pannexin-1 (Panx1), which is a pathway previously identified to be active during excitotoxicity. Panx1 was not calcium permeable but may contribute to late phase LTD via ATP release. Panx1 blockers prevent LTD, and this was rescued with exogenous ATP application. Inhibition of LTD with 5-BDBD suggests the downstream involvement of postsynaptic P2X4 receptors.
Collapse
Affiliation(s)
- Allison C Nielsen
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Connor L Anderson
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carina Ens
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Andrew K J Boyce
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Roger J Thompson
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
9
|
Henze E, Ehrlich JJ, Robertson JL, Gelsleichter E, Kawate T. The C-terminal activating domain promotes pannexin 1 channel opening. Proc Natl Acad Sci U S A 2024; 121:e2411898121. [PMID: 39671183 DOI: 10.1073/pnas.2411898121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
Pannexin 1 (Panx1) constitutes a large pore channel responsible for the release of adenosine triphosphate (ATP) from apoptotic cells. Strong evidence indicates that caspase-mediated cleavage of the C-terminus promotes the opening of the Panx1 channel by unplugging the pore. However, this simple pore-plugging mechanism alone cannot account for the observation that a Panx1 construct ending before the caspase cleavage site remains closed. Here, we show that a helical region located immediately before the caspase cleavage site, referred to as the "C-terminal activating domain (CAD)", plays a pivotal role in facilitating Panx1 activation. Electrophysiology and mutagenesis studies uncovered that two conserved leucine residues within the CAD play a pivotal role. Cryoelectron microscopy (Cryo-EM) analysis of the construct ending before reaching the CAD demonstrated that the N terminus extends into an intracellular pocket. In contrast, the construct including the CAD revealed that this domain occupies the intracellular pocket, causing the N terminus to flip upward within the pore. Analysis of electrostatic free energy landscape in the closed conformation indicated that the intracellular side of the ion permeation pore may be occupied by anions like ATP, creating an electrostatic barrier for anions attempting to permeate the pore. When the N terminus flips up, it diminishes the positively charged surface, thereby reducing the drive to accumulate anions inside the pore. This dynamic change in the electrostatic landscape likely contributes to the selection of permeant ions. Collectively, these experiments put forth a mechanism in which C-terminal cleavage liberates the CAD, causing the repositioning of the N terminus to promote Panx1 channel opening.
Collapse
Affiliation(s)
- Erik Henze
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | | | - Janice L Robertson
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Eric Gelsleichter
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| |
Collapse
|
10
|
O'Donnell BL, Johnston D, Bhatt A, Kardan Z, Stefan D, Bysice A, Sayedyahossein S, Dagnino L, Cecchini M, Loganathan SK, Roth K, Penuela S. Pannexin 1 and pannexin 3 differentially regulate the cancer cell properties of cutaneous squamous cell carcinoma. J Physiol 2024. [PMID: 39560179 DOI: 10.1113/jp286172] [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: 03/12/2024] [Accepted: 10/23/2024] [Indexed: 11/20/2024] Open
Abstract
Pannexin (PANX) channels are present in skin and facilitate the movement of signalling molecules during cellular communication. PANX1 and PANX3 function in skin homeostasis and keratinocyte differentiation but were previously reduced in a small cohort of human cutaneous squamous cell carcinoma (cSCC) tumours compared to normal epidermis. In our study we used SCC-13 cells, limited publicly available RNA-seq data and a larger cohort of cSCC patient-matched samples to analyse PANX1 and PANX3 expression and determine the association between their dysregulation and the malignant properties of cSCC. In a bioinformatics analysis, PANX1 transcripts were increased in cSCC and head and neck SCC tumours compared to normal tissues, but PANX3 mRNA showed no differences. However, in our own cohort PANX3 transcripts were decreased in cSCC compared to patient-matched aged skin, whereas PANX1 protein was upregulated in cSCC. PANX1 localized to all regions within the cSCC tumour microenvironment, and increased levels were associated with larger tumour dimensions. To investigate PANX1 function in SCC-13 cells, we deleted PANX1 via CRISPR/Cas9 and treated with PANX1 inhibitors, which markedly reduced cell growth and migration. To assess PANX3 function in cutaneous carcinogenesis, we employed the 7,12-dimethylbenz(a)anthracene/12-otetradecanoylphorbol-13-acetate (DMBA/TPA) model using our global Panx3 knockout (KO) mice, where 60% of wild-type and 100% of KO mice formed precancerous papillomas. Average papilloma volumes at endpoint were significantly increased in KO mice and showed moderate evidence of increases in KO mice over time. Collectively, these findings suggest PANX1 and PANX3 dysregulation may have potential tumour-promoting and tumour-suppressive effects for keratinocyte transformation, respectively. KEY POINTS: Pannexin 1 (PANX1) and pannexin 3 (PANX3) are channel-forming proteins which are critical in the normal maintenance and function of keratinocytes in the skin but may become altered in cutaneous squamous cell carcinoma (cSCC) tumours. In this study we used a combination of culture models, mouse models and patient-derived tissues. We found PANX1 levels are increased in cSCC tumours and present in all tumour regions, functioning to promote cSCC cell growth and migration. Conversely, PANX3 levels are decreased in cSCC tumours, and this protein reduces the incidence and growth of precancerous lesions. Taken together our data indicate that in cSCC these pannexin family members seem to have opposite effects, in either promoting or restricting cancer cell properties. These results help us to better understand the mechanisms of malignant transformation of keratinocytes and offer a new potential therapeutic target for the treatment of advanced cSCC.
Collapse
Affiliation(s)
- Brooke L O'Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Danielle Johnston
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Ayushi Bhatt
- Faculty of Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Zahra Kardan
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Dan Stefan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Andrew Bysice
- Department of Otolaryngology - Head and Neck Surgery, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Samar Sayedyahossein
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Division of Experimental Oncology, Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Matthew Cecchini
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sampath Kumar Loganathan
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
- Departments of Otolaryngology - Head and Neck Surgery, Biochemistry and Experimental Medicine, Rosalind and Moris Goodman Cancer Research Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Kathryn Roth
- Department of Otolaryngology - Head and Neck Surgery, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Division of Experimental Oncology, Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
11
|
Mutafova-Yambolieva VN. Mechanosensitive release of ATP in the urinary bladder mucosa. Purinergic Signal 2024:10.1007/s11302-024-10063-6. [PMID: 39541058 DOI: 10.1007/s11302-024-10063-6] [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: 08/19/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
The urinary bladder mucosa (urothelium and suburothelium/lamina propria) functions as a barrier between the content of the urine and the underlying bladder tissue. The bladder mucosa is also a mechanosensitive tissue that releases signaling molecules that affect functions of cells in the bladder wall interconnecting the mucosa with the detrusor muscle and the CNS. Adenosine 5'-triphosphate (ATP) is a primary mechanotransduction signal that is released from cells in the bladder mucosa in response to bladder wall distention and activates cell membrane-localized P2X and P2Y purine receptors on urothelial cells, sensory and efferent neurons, interstitial cells, and detrusor smooth muscle cells. The amounts of ATP at active receptor sites depend significantly on the amounts of extracellularly released ATP. Spontaneous and distention-induced release of ATP appear to be under differential control. This review is focused on mechanisms underlying urothelial release of ATP in response to mechanical stimulation. First, we present a brief overview of studies that report mechanosensitive ATP release in bladder cells or tissues. Then, we discuss experimental evidence for mechanosensitive release of urothelial ATP by vesicular and non-vesicular mechanisms and roles of the stretch-activated channels PIEZO channels, transient receptor potential vanilloid type 4, and pannexin 1. This is followed by brief discussion of possible involvement of calcium homeostasis modulator 1, acid-sensing channels, and connexins in the release of urothelial ATP. We conclude with brief discussion of limitations of current research and of needs for further studies to increase our understanding of mechanotransduction in the bladder wall and of purinergic regulation of bladder function.
Collapse
|
12
|
Pruitt L, Abbott RK. Hypoxia-adenosinergic regulation of B cell responses. Front Immunol 2024; 15:1478506. [PMID: 39559353 PMCID: PMC11570280 DOI: 10.3389/fimmu.2024.1478506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
Hypoxic microenvironments induce widespread metabolic changes that have been shown to be critical in regulating innate and adaptive immune responses. Hypoxia-induced changes include the generation of extracellular adenosine followed by subsequent signaling through adenosine receptors on immune cells. This evolutionarily conserved "hypoxia-adenosinergic" pathway of hypoxia → extracellular adenosine → adenosine receptor signaling has been shown to be critical in limiting and redirecting T cell responses including in tumor microenvironments and the gut mucosa. However, the question of whether hypoxic microenvironments are involved in the development of B cell responses has remained unexplored until recently. The discovery that germinal centers (GC), the anatomic site in which B cells undergo secondary diversification and affinity maturation, develop a hypoxic microenvironment has sparked new interest in how this evolutionarily conserved pathway affects antibody responses. In this review we will summarize what is known about hypoxia-adenosinergic microenvironments in lymphocyte development and ongoing immune responses. Specific focus will be placed on new developments regarding the role of the hypoxia-adenosinergic pathway in regulating GC development and humoral immunity.
Collapse
Affiliation(s)
| | - Robert K. Abbott
- Department of Pathology, University of Texas Medical Branch,
Galveston, TX, United States
| |
Collapse
|
13
|
Di Virgilio F, Vultaggio-Poma V, Tarantini M, Giuliani AL. Overview of the role of purinergic signaling and insights into its role in cancer therapy. Pharmacol Ther 2024; 262:108700. [PMID: 39111410 DOI: 10.1016/j.pharmthera.2024.108700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 07/05/2024] [Accepted: 07/31/2024] [Indexed: 08/30/2024]
Abstract
Innovation of cancer therapy has received a dramatic acceleration over the last fifteen years thanks to the introduction of the novel immune checkpoint inhibitors (ICI). On the other hand, the conspicuous scientific knowledge accumulated in purinergic signaling since the early seventies is finally being transferred to the clinic. Several Phase I/II clinical trials are currently underway to investigate the effect of drugs interfering with purinergic signaling as stand-alone or combination therapy in cancer. This is supporting the novel concept of "purinergic immune checkpoint" (PIC) in cancer therapy. In the present review we will address a) the basic pharmacology and cell biology of the purinergic system; b) principles of its pathophysiology in human diseases; c) implications for cell death, cell proliferation and cancer; d) novel molecular tools to investigate nucleotide homeostasis in the extracellular environment; e) recent developments in the pharmacology of P1, P2 receptors and related ecto-enzymes; f) P1 and P2 ligands as novel diagnostic tools; g) current issues in PIC-based anti-cancer therapy. This review will provide an appraisal of the current status of purinergic signaling in cancer and will help identify future avenues of development.
Collapse
Affiliation(s)
| | | | - Mario Tarantini
- Department of Medical Sciences, University of Ferrara, Italy
| | | |
Collapse
|
14
|
Casillas Martinez A, Wicki-Stordeur LE, Ariano AV, Swayne LA. Dual role for pannexin 1 at synapses: regulating functional and morphological plasticity. J Physiol 2024. [PMID: 39264228 DOI: 10.1113/jp285228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
Pannexin 1 (PANX1) is an ion and metabolite membrane channel and scaffold protein enriched in synaptic compartments of neurons in the central nervous system. In addition to a well-established link between PANX1 and synaptic plasticity, we recently identified a role for PANX1 in the regulation of dendritic spine stability. Notably, PANX1 and its interacting proteins are linked to neurological conditions involving dendritic spine loss. Understanding the dual role of PANX1 in synaptic function and morphology may help to shed light on these links. We explore potential mechanisms, including PANX1's interactions with postsynaptic receptors and cytoskeleton regulating proteins. Finally, we contextualize PANX1's dual role within neurological diseases involving dendritic spine and synapse dysfunction.
Collapse
Affiliation(s)
| | - Leigh E Wicki-Stordeur
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Annika V Ariano
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
15
|
Sayedyahossein S, Huang K, Zhang C, Karimi M, Bahmani M, O’Donnell BL, Wakefield B, Li Z, Johnston D, Leighton SE, Huver MS, Dagnino L, Sacks DB, Penuela S. Pannexin 1 crosstalk with the Hippo pathway in malignant melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.611059. [PMID: 39372769 PMCID: PMC11451602 DOI: 10.1101/2024.09.03.611059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
In this study, we explored the intricate relationship between Pannexin 1 (PANX1) and the Hippo signaling pathway effector, Yes-associated protein (YAP). Analysis of The Cancer Genome Atlas (TCGA) data revealed a significant positive correlation between PANX1 mRNA and core Hippo components, YAP, TAZ, and Hippo scaffold, IQGAP1, in invasive cutaneous melanoma and breast carcinoma. Furthermore, we demonstrated that PANX1 expression is upregulated in invasive melanoma cell lines and is associated with increased YAP protein levels. Notably, our investigations uncovered a previously unrecognized interaction between endogenous PANX1 and the Hippo scaffold protein IQGAP1 in melanoma cells. Moreover, our findings revealed that IQGAP1 exhibits differential expression in melanoma cells and plays a regulatory role in cellular morphology. Functional studies involving PANX1 knockdown provided compelling evidence that PANX1 modulates YAP protein levels and its co-transcriptional activity in both melanoma and breast carcinoma cells. Importantly, our study showcases the potential therapeutic relevance of targeting PANX1, as pharmacological inhibition of PANX1 using selective FDA-approved inhibitors or PANX1 knockdown reduced YAP abundance in melanoma cells. Furthermore, our Clariom™ S analysis unveiled key genes implicated in cell proliferation, such as neuroglin1 (NRG1), β-galactoside binding protein, galectin-3 (LGALS3), that are affected in PANX1-deficient cells. In summary, our investigation delves into the intricate interplay between PANX1 and YAP in the context of invasive melanoma, offering valuable insights into potential therapeutic strategies for effective treatment.
Collapse
Affiliation(s)
- Samar Sayedyahossein
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Kenneth Huang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Christopher Zhang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Mehdi Karimi
- Department of Mathematics, Illinois State University, Normal, Illinois, USA, 61790
| | | | - Brooke L. O’Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Brent Wakefield
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Zhigang Li
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
| | - Danielle Johnston
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Stephanie E. Leighton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Matthew S. Huver
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - David B. Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| |
Collapse
|
16
|
Buckley C, Lee MD, Zhang X, Wilson C, McCarron JG. Signalling switches maintain intercellular communication in the vascular endothelium. Br J Pharmacol 2024; 181:2810-2832. [PMID: 38651236 DOI: 10.1111/bph.16366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND AND PURPOSE The single layer of cells lining all blood vessels, the endothelium, is a sophisticated signal co-ordination centre that controls a wide range of vascular functions including the regulation of blood pressure and blood flow. To co-ordinate activities, communication among cells is required for tissue level responses to emerge. While a significant form of communication occurs by the propagation of signals between cells, the mechanism of propagation in the intact endothelium is unresolved. EXPERIMENTAL APPROACH Precision signal generation and targeted cellular manipulation was used in conjunction with high spatiotemporal mesoscale Ca2+ imaging in the endothelium of intact blood vessels. KEY RESULTS Multiple mechanisms maintain communication so that Ca2+ wave propagation occurs irrespective of the status of connectivity among cells. Between adjoining cells, regenerative IP3-induced IP3 production transmits Ca2+ signals and explains the propagated vasodilation that underlies the increased blood flow accompanying tissue activity. The inositide is itself sufficient to evoke regenerative phospholipase C-dependent Ca2+ waves across coupled cells. None of gap junctions, Ca2+ diffusion or the release of extracellular messengers is required to support this type of intercellular Ca2+ signalling. In contrast, when discontinuities exist between cells, ATP released as a diffusible extracellular messenger transmits Ca2+ signals across the discontinuity and drives propagated vasodilation. CONCLUSION AND IMPLICATIONS These results show that signalling switches underlie endothelial cell-to-cell signal transmission and reveal how communication is maintained in the face of endothelial damage. The findings provide a new framework for understanding wave propagation and cell signalling in the endothelium.
Collapse
Affiliation(s)
- Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
17
|
Feng YH, Tang RJ, Zhang YY, Lin J, Liu YJ, Li YK, Li CJ, Zhou C, Liu F, Shen JF. Contribution of inwardly rectifying potassium channel 4.1 in orofacial neuropathic pain: Regulation of pannexin 3 via the reactive oxygen species-activated P38 MAPK signal pathway. Eur J Neurosci 2024; 60:4569-4585. [PMID: 38992988 DOI: 10.1111/ejn.16458] [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: 08/01/2023] [Revised: 05/17/2024] [Accepted: 06/19/2024] [Indexed: 07/13/2024]
Abstract
The involvement of inwardly rectifying potassium channel 4.1 (Kir4.1) in neuropathic pain has been established. However, there is limited understanding of the downstream mechanism through which Kir4.1 contributes to orofacial neuropathic pain. The objective of this study was to examine the regulation of Kir4.1 on the expression of pannexin 3 (Panx3) in the trigeminal ganglion (TG) and the underlying mechanism in the context of orofacial neuropathic pain caused by chronic constriction injury of the infraorbital nerve (CCI-ION). The study observed a significant increase in Panx3 expression in the TG of mice with CCI-ION. Inhibition of Panx3 in the TG of CCI-ION mice resulted in alleviation of orofacial mechanical allodynia. Furthermore, conditional knockdown (CKD) of Kir4.1 in the TG of both male and female mice led to mechanical allodynia and upregulation of Panx3 expression. Conversely, overexpression of Kir4.1 decreased Panx3 levels in the TG and relieved mechanical allodynia in CCI-ION mice. In addition, silencing Kir4.1 in satellite glial cells (SGCs) decreased Panx3 expression and increased the phosphorylation of P38 MAPK. Moreover, silencing Kir4.1 in SGCs increased the levels of reactive oxygen species (ROS). The elevated phosphorylation of P38 MAPK resulting from Kir4.1 silencing was inhibited by using a superoxide scavenger known as the tempol. Silencing Panx3 in the TG in vivo attenuated the mechanical allodynia caused by Kir4.1 CKD. In conclusion, these findings suggest that the reduction of Kir4.1 promotes the expression of Panx3 by activating the ROS-P38 MAPK signalling pathway, thus contributing to the development of orofacial neuropathic pain.
Collapse
Affiliation(s)
- Yu-Heng Feng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ren-Jie Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiu Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Jing Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi-Ke Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chun-Jie Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
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
|
19
|
Obot P, Cibelli A, Pan J, Velíšek L, Velíšková J, Scemes E. Pannexin1 Mediates Early-Life Seizure-Induced Social Behavior Deficits. ASN Neuro 2024; 16:2371164. [PMID: 39024558 PMCID: PMC11262470 DOI: 10.1080/17590914.2024.2371164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/11/2024] [Indexed: 07/20/2024] Open
Abstract
There is a high co-morbidity between childhood epilepsy and autism spectrum disorder (ASD), with age of seizure onset being a critical determinant of behavioral outcomes. The interplay between these comorbidities has been investigated in animal models with results showing that the induction of seizures at early post-natal ages leads to learning and memory deficits and to autistic-like behavior in adulthood. Modifications of the excitation/inhibition (glutamate/GABA, ATP/adenosine) balance that follows early-life seizures (ELS) are thought to be the physiological events that underlie neuropsychiatric and neurodevelopmental disorders. Although alterations in purinergic/adenosinergic signaling have been implicated in seizures and ASD, it is unknown whether the ATP release channels, Pannexin1 (Panx1), contribute to ELS-induced behavior changes. To tackle this question, we used the ELS-kainic acid model in transgenic mice with global and cell type specific deletion of Panx1 to evaluate whether these channels were involved in behavioral deficits that occur later in life. Our studies show that ELS results in Panx1 dependent social behavior deficits and also in poor performance in a spatial memory test that does not involve Panx1. These findings provide support for a link between ELS and adult behavioral deficits. Moreover, we identify neuronal and not astrocyte Panx1 as a potential target to specifically limit astrogliosis and social behavioral deficits resultant from early-life seizures.
Collapse
Affiliation(s)
- Price Obot
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| | - Antonio Cibelli
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jian Pan
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| | - Libor Velíšek
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
- Department of Neurology, New York Medical College, Valhalla, New York, USA
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Jana Velíšková
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
- Department of Neurology, New York Medical College, Valhalla, New York, USA
- Department of Obstetrics and Gynecology, New York Medical College, Valhalla, New York, USA
| | - Eliana Scemes
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
20
|
Hu CQ, Hou T, Xiang R, Li X, Li J, Wang TT, Liu WJ, Hou S, Wang D, Zhao QH, Yu XX, Xu M, Liu XK, Chi YJ, Yang JC. PANX1-mediated ATP release confers FAM3A's suppression effects on hepatic gluconeogenesis and lipogenesis. Mil Med Res 2024; 11:41. [PMID: 38937853 PMCID: PMC11210080 DOI: 10.1186/s40779-024-00543-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Extracellular adenosine triphosphate (ATP) is an important signal molecule. In previous studies, intensive research had revealed the crucial roles of family with sequence similarity 3 member A (FAM3A) in controlling hepatic glucolipid metabolism, islet β cell function, adipocyte differentiation, blood pressure, and other biological and pathophysiological processes. Although mitochondrial protein FAM3A plays crucial roles in the regulation of glucolipid metabolism via stimulating ATP release to activate P2 receptor pathways, its mechanism in promoting ATP release in hepatocytes remains unrevealed. METHODS db/db, high-fat diet (HFD)-fed, and global pannexin 1 (PANX1) knockout mice, as well as liver sections of individuals, were used in this study. Adenoviruses and adeno-associated viruses were utilized for in vivo gene overexpression or inhibition. To evaluate the metabolic status in mice, oral glucose tolerance test (OGTT), pyruvate tolerance test (PTT), insulin tolerance test (ITT), and magnetic resonance imaging (MRI) were conducted. Protein-protein interactions were determined by coimmunoprecipitation with mass spectrometry (MS) assays. RESULTS In livers of individuals and mice with steatosis, the expression of ATP-permeable channel PANX1 was increased (P < 0.01). Hepatic PANX1 overexpression ameliorated the dysregulated glucolipid metabolism in obese mice. Mice with hepatic PANX1 knockdown or global PANX1 knockout exhibited disturbed glucolipid metabolism. Restoration of hepatic PANX1 rescued the metabolic disorders of PANX1-deficient mice (P < 0.05). Mechanistically, ATP release is mediated by the PANX1-activated protein kinase B-forkhead box protein O1 (Akt-FOXO1) pathway to inhibit gluconeogenesis via P2Y receptors in hepatocytes. PANX1-mediated ATP release also activated calmodulin (CaM) (P < 0.01), which interacted with c-Jun N-terminal kinase (JNK) to inhibit its activity, thereby deactivating the transcription factor activator protein-1 (AP1) and repressing fatty acid synthase (FAS) expression and lipid synthesis (P < 0.05). FAM3A stimulated the expression of PANX1 via heat shock factor 1 (HSF1) in hepatocytes (P < 0.05). Notably, FAM3A overexpression failed to promote ATP release, inhibit the expression of gluconeogenic and lipogenic genes, and suppress gluconeogenesis and lipid deposition in PANX1-deficient hepatocytes and livers. CONCLUSIONS PANX1-mediated release of ATP plays a crucial role in maintaining hepatic glucolipid homeostasis, and it confers FAM3A's suppressive effects on hepatic gluconeogenesis and lipogenesis.
Collapse
Affiliation(s)
- Cheng-Qing Hu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
- Department of Obstetrics and Gynecology, Peking University Third Hospital/National Clinical Research Center for Obstetrics and Gynecology, Beijing, 100191, China
| | - Tao Hou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Rui Xiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Xin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Jing Li
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Tian-Tian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Wen-Jun Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Song Hou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Di Wang
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing, 100044, China
| | - Qing-He Zhao
- Department of Gastroenterology, Peking University People's Hospital, Beijing, 100044, China
| | - Xiao-Xing Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Ming Xu
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital/Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Beijing, 100191, China
| | - Xing-Kai Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Centre, the First Hospital of Jilin University, Changchun, 130061, China.
| | - Yu-Jing Chi
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing, 100044, China.
- Department of Gastroenterology, Peking University People's Hospital, Beijing, 100044, China.
| | - Ji-Chun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China.
- Department of Cardiology, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
21
|
Thakku Sivakumar D, Jain K, Alfehaid N, Wang Y, Teng X, Fischer W, Engel T. The Purinergic P2X7 Receptor as a Target for Adjunctive Treatment for Drug-Refractory Epilepsy. Int J Mol Sci 2024; 25:6894. [PMID: 39000004 PMCID: PMC11241490 DOI: 10.3390/ijms25136894] [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: 04/17/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 07/14/2024] Open
Abstract
Epilepsy is one of the most common neurological diseases worldwide. Anti-seizure medications (ASMs) with anticonvulsants remain the mainstay of epilepsy treatment. Currently used ASMs are, however, ineffective to suppress seizures in about one third of all patients. Moreover, ASMs show no significant impact on the pathogenic mechanisms involved in epilepsy development or disease progression and may cause serious side-effects, highlighting the need for the identification of new drug targets for a more causal therapy. Compelling evidence has demonstrated a role for purinergic signalling, including the nucleotide adenosine 5'-triphosphate (ATP) during the generation of seizures and epilepsy. Consequently, drugs targeting specific ATP-gated purinergic receptors have been suggested as promising treatment options for epilepsy including the cationic P2X7 receptor (P27XR). P2X7R protein levels have been shown to be increased in the brain of experimental models of epilepsy and in the resected brain tissue of patients with epilepsy. Animal studies have provided evidence that P2X7R blocking can reduce the severity of acute seizures and the epileptic phenotype. The current review will provide a brief summary of recent key findings on P2X7R signalling during seizures and epilepsy focusing on the potential clinical use of treatments based on the P2X7R as an adjunctive therapeutic strategy for drug-refractory seizures and epilepsy.
Collapse
Affiliation(s)
- Divyeshz Thakku Sivakumar
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Krishi Jain
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Noura Alfehaid
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Yitao Wang
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
- International College of Pharmaceutical Innovation, Soochow University, Suzhou 215123, China
| | - Xinchen Teng
- International College of Pharmaceutical Innovation, Soochow University, Suzhou 215123, China
| | | | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| |
Collapse
|
22
|
Henze E, Ehrlich JJ, Robertson JL, Kawate T. The C-terminal activating domain promotes Panx1 channel opening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598903. [PMID: 38915727 PMCID: PMC11195165 DOI: 10.1101/2024.06.13.598903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Pannexin 1 (Panx1) constitutes a large pore channel responsible for the release of ATP from apoptotic cells. Strong evidence indicates that caspase-mediated cleavage of the C-terminus promotes the opening of the Panx1 channel by unplugging the pore. However, this simple pore-plugging mechanism alone cannot account for the observation that a Panx1 construct ending before the caspase cleavage site remains closed. Here, we show that a helical region located immediately before the caspase cleavage site, referred to as the "C-terminal activating domain (CAD)," plays a pivotal role in facilitating Panx1 activation. Electrophysiology and mutagenesis studies uncovered that two conserved leucine residues within the CAD plays a pivotal role. Cryo-EM analysis of the construct ending before reaching the CAD demonstrated that the N-terminus extends into an intracellular pocket. In contrast, the construct including the CAD revealed that this domain occupies the intracellular pocket, causing the N-terminus to flip upward within the pore. Analysis of electrostatic free energy landscape in the closed conformation indicated that the intracellular side of the ion permeation pore may be occupied by anions like ATP, creating an electrostatic barrier for anions attempting to permeate the pore. When the N-terminus flips up, it diminishes the positively charged surface, thereby reducing the drive to accumulate anions inside the pore. This dynamic change in the electrostatic landscape likely contributes to the selection of permeant ions. Collectively, these experiments put forth a novel mechanism in which C-terminal cleavage liberates the CAD, causing the repositioning of the N-terminus to promote Panx1 channel opening.
Collapse
Affiliation(s)
- Erik Henze
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | - Janice L. Robertson
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
23
|
Vitureira N, Rafael A, Abudara V. P2X7 receptors and pannexin1 hemichannels shape presynaptic transmission. Purinergic Signal 2024; 20:223-236. [PMID: 37713157 PMCID: PMC11189373 DOI: 10.1007/s11302-023-09965-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023] Open
Abstract
Over the last decades, since the discovery of ATP as a transmitter, accumulating evidence has been reported about the role of this nucleotide and purinergic receptors, in particular P2X7 receptors, in the modulation of synaptic strength and plasticity. Purinergic signaling has emerged as a crucial player in orchestrating the molecular interaction between the components of the tripartite synapse, and much progress has been made in how this neuron-glia interaction impacts neuronal physiology under basal and pathological conditions. On the other hand, pannexin1 hemichannels, which are functionally linked to P2X7 receptors, have appeared more recently as important modulators of excitatory synaptic function and plasticity under diverse contexts. In this review, we will discuss the contribution of ATP, P2X7 receptors, and pannexin hemichannels to the modulation of presynaptic strength and its impact on motor function, sensory processing, synaptic plasticity, and neuroglial communication, with special focus on the P2X7 receptor/pannexin hemichannel interplay. We also address major hypotheses about the role of this interaction in physiological and pathological circumstances.
Collapse
Affiliation(s)
- Nathalia Vitureira
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - Alberto Rafael
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Verónica Abudara
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
24
|
Lamb IR, Novielli‐Kuntz NM, Murrant CL. The role of pannexin/purinergic signaling in intervascular communication from capillaries during skeletal muscle contraction in male Golden hamsters. Physiol Rep 2024; 12:e16113. [PMID: 38898485 PMCID: PMC11186745 DOI: 10.14814/phy2.16113] [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: 02/26/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
We sought to determine the physiological relevance of pannexin/purinergic-dependent signaling in mediating conducted vasodilation elicited by capillary stimulation through skeletal muscle contraction. Using hamster cremaster muscle and intravital microscopy we stimulated capillaries through local muscle contraction while observing the associated upstream arteriole. Capillaries were stimulated with muscle contraction at low and high contraction (6 and 60CPM) and stimulus frequencies (4 and 40 Hz) in the absence and presence of pannexin blocker mefloquine (MEF; 10-5 M), purinergic receptor antagonist suramin (SUR 10-5 M) and gap-junction uncoupler halothane (HALO, 0.07%) applied between the capillary stimulation site and the upstream arteriolar observation site. Conducted vasodilations elicited at 6CPM were inhibited by HALO while vasodilations at 60CPM were inhibited by MEF and SUR. The conducted response elicited at 4 Hz was inhibited by MEF while the vasodilation at 40 Hz was unaffected by any blocker. Therefore, upstream vasodilations resulting from capillary stimulation via muscle contraction are dependent upon a pannexin/purinergic-dependent pathway that appears to be stimulation parameter-dependent. Our data highlight a physiological importance of the pannexin/purinergic pathway in facilitating communication between capillaries and upstream arteriolar microvasculature and, consequently, indicating that this pathway may play a crucial role in regulating blood flow in response to skeletal muscle contraction.
Collapse
Affiliation(s)
- Iain R. Lamb
- Department of Human Health and Nutritional SciencesUniversity of GuelphGuelphOntarioCanada
| | | | - Coral L. Murrant
- Department of Human Health and Nutritional SciencesUniversity of GuelphGuelphOntarioCanada
| |
Collapse
|
25
|
Guo Z, Yue N, Chen M, Li J, Lv R, Wang J, Liu T, Huang J, Gao S, Li Y, Yuan B, Wang J, Kang L, Ji B, Xin W. Purinergic Receptor Antagonists Inhibit Hemolysis Induced by Clostridium perfringens Alpha Toxin. Pathogens 2024; 13:454. [PMID: 38921752 PMCID: PMC11206506 DOI: 10.3390/pathogens13060454] [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: 03/10/2024] [Revised: 04/28/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
Clostridium perfringens alpha toxin (CPA), which causes yellow lamb disease in sheep and gas gangrene and food poisoning in humans, is produced by all types of C. perfringens and is the major virulence determinant of C. perfringens type A. CPA induces hemolysis in many species, including humans, murines, sheep and rabbits, through its enzymatic activity, which dissolves the cell membrane. Recent studies have shown that some pore-forming toxins cause hemolysis, which is achieved by the activation of purinergic receptors (P2). However, the relationship between P2 receptors and non-pore-forming toxin hemolysis has not been investigated. In the present study, we examined the function of P2 receptors in CPA toxin hemolysis and found that CPA-induced hemolysis was dependent on P2 receptor activation, and this was also true for Staphylococcus aureus β-Hemolysin, another non-pore-forming toxin. Furthermore, we use selective P2 receptor antagonists to demonstrate that P2X1 and P2X7 play important roles in the hemolysis of human and murine erythrocytes. In addition, we found that redox metabolism was mainly involved in CPA-induced hemolysis using metabolomic analysis. We further demonstrate that CPA activates P2 receptors and then activates NADPH oxidase through the PI3K/Akt and MEK1/ERK1 pathways, followed by the production of active oxygen to induce hemolysis. These findings contribute to our understanding of the pathological effects of CPA, clarify the relationship between P2 activation and non-pore-forming toxin-induced hemolysis, and provide new insights into CPA-induced hemolysis.
Collapse
Affiliation(s)
- Zishuo Guo
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
- Emergency Department, 96605 Army Hospital of the People’s Liberation Army, Jilin 134000, China
| | - Nan Yue
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Ming Chen
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Jiaxin Li
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Ruomei Lv
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Jing Wang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Tingting Liu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Jing Huang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Shan Gao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Yanwei Li
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Bing Yuan
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Jinglin Wang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Lin Kang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| | - Bin Ji
- Department of Disease Control, The Affiliated Wuxi Center for Disease Control and Prevention, Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi 214101, China
| | - Wenwen Xin
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (Z.G.); (N.Y.); (M.C.); (J.L.); (R.L.); (J.W.); (T.L.); (J.H.); (S.G.); (Y.L.); (B.Y.); (J.W.)
| |
Collapse
|
26
|
Misiti F, Diotaiuti P, Lombardo GE, Tellone E. Sphingosine-1-phosphate Decreases Erythrocyte Dysfunction Induced by β-Amyloid. Int J Mol Sci 2024; 25:5184. [PMID: 38791223 PMCID: PMC11121638 DOI: 10.3390/ijms25105184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/28/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Amyloid beta peptides (Aβ) have been identified as the main pathogenic agents in Alzheimer's disease (AD). Soluble Aβ oligomers, rather than monomer or insoluble amyloid fibrils, show red blood cell (RBC) membrane-binding capacity and trigger several morphological and functional alterations in RBCs that can result in impaired oxygen transport and delivery. Since bioactive lipids have been recently proposed as potent protective agents against Aβ toxicity, we investigated the role of sphingosine-1-phosphate (S1P) in signaling pathways involved in the mechanism underlying ATP release in Ab-treated RBCs. In RBCs following different treatments, the ATP, 2,3 DPG and cAMP levels and caspase 3 activity were determined by spectrophotometric and immunoassay. S1P rescued the inhibition of ATP release from RBCs triggered by Ab, through a mechanism involving caspase-3 and restoring 2,3 DPG and cAMP levels within the cell. These findings reveal the molecular basis of S1P protection against Aβ in RBCs and suggest new therapeutic avenues in AD.
Collapse
Affiliation(s)
- Francesco Misiti
- Human Sciences, Social and Health Department, University of Cassino and Lazio Meridionale, V. S. Angelo, Loc. Folcara, 03043 Cassino, Italy;
| | - Pierluigi Diotaiuti
- Human Sciences, Social and Health Department, University of Cassino and Lazio Meridionale, V. S. Angelo, Loc. Folcara, 03043 Cassino, Italy;
| | - Giovanni Enrico Lombardo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (G.E.L.); (E.T.)
| | - Ester Tellone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (G.E.L.); (E.T.)
| |
Collapse
|
27
|
Lucero CM, Navarro L, Barros-Osorio C, Cáceres-Conejeros P, Orellana JA, Gómez GI. Activation of Pannexin-1 channels causes cell dysfunction and damage in mesangial cells derived from angiotensin II-exposed mice. Front Cell Dev Biol 2024; 12:1387234. [PMID: 38660621 PMCID: PMC11041381 DOI: 10.3389/fcell.2024.1387234] [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: 02/17/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Chronic kidney disease (CKD) is a prevalent health concern associated with various pathological conditions, including hypertensive nephropathy. Mesangial cells are crucial in maintaining glomerular function, yet their involvement in CKD pathogenesis remains poorly understood. Recent evidence indicates that overactivation of Pannexin-1 (Panx1) channels could contribute to the pathogenesis and progression of various diseases. Although Panx1 is expressed in the kidney, its contribution to the dysfunction of renal cells during pathological conditions remains to be elucidated. This study aimed to investigate the impact of Panx1 channels on mesangial cell function in the context of hypertensive nephropathy. Using an Ang II-infused mouse model and primary mesangial cell cultures, we demonstrated that in vivo exposure to Ang II sensitizes cultured mesangial cells to show increased alterations when they are subjected to subsequent in vitro exposure to Ang II. Particularly, mesangial cell cultures treated with Ang II showed elevated activity of Panx1 channels and increased release of ATP. The latter was associated with enhanced basal intracellular Ca2+ ([Ca2+]i) and increased ATP-mediated [Ca2+]i responses. These effects were accompanied by increased lipid peroxidation and reduced cell viability. Crucially, all the adverse impacts evoked by Ang II were prevented by the blockade of Panx1 channels, underscoring their critical role in mediating cellular dysfunction in mesangial cells. By elucidating the mechanisms by which Ang II negatively impacts mesangial cell function, this study provides valuable insights into the pathogenesis of renal damage in hypertensive nephropathy.
Collapse
Affiliation(s)
- Claudia M. Lucero
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Laura Navarro
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Cristián Barros-Osorio
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Patricio Cáceres-Conejeros
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Juan A. Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gonzalo I. Gómez
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
28
|
Hussain N, Apotikar A, Pidathala S, Mukherjee S, Burada AP, Sikdar SK, Vinothkumar KR, Penmatsa A. Cryo-EM structures of pannexin 1 and 3 reveal differences among pannexin isoforms. Nat Commun 2024; 15:2942. [PMID: 38580658 PMCID: PMC10997603 DOI: 10.1038/s41467-024-47142-6] [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: 10/01/2022] [Accepted: 03/19/2024] [Indexed: 04/07/2024] Open
Abstract
Pannexins are single-membrane large-pore channels that release ions and ATP upon activation. Three isoforms of pannexins 1, 2, and 3, perform diverse cellular roles and differ in their pore lining residues. In this study, we report the cryo-EM structure of pannexin 3 at 3.9 Å and analyze its structural differences with pannexin isoforms 1 and 2. The pannexin 3 vestibule has two distinct chambers and a wider pore radius in comparison to pannexins 1 and 2. We further report two cryo-EM structures of pannexin 1, with pore substitutions W74R/R75D that mimic the pore lining residues of pannexin 2 and a germline mutant of pannexin 1, R217H at resolutions of 3.2 Å and 3.9 Å, respectively. Substitution of cationic residues in the vestibule of pannexin 1 results in reduced ATP interaction propensities to the channel. The germline mutant R217H in transmembrane helix 3 (TM3), leads to a partially constricted pore, reduced ATP interaction and weakened voltage sensitivity. The study compares the three pannexin isoform structures, the effects of substitutions of pore and vestibule-lining residues and allosteric effects of a pathological substitution on channel structure and function thereby enhancing our understanding of this vital group of ATP-release channels.
Collapse
Affiliation(s)
- Nazia Hussain
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Ashish Apotikar
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Shabareesh Pidathala
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sourajit Mukherjee
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
- Department of Chemistry, The University of Chicago, Chicago, USA
| | - Ananth Prasad Burada
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Sujit Kumar Sikdar
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Kutti R Vinothkumar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, 560065, India
| | - Aravind Penmatsa
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
29
|
Volonte D, Benson CJ, Daugherty SL, Beckel JM, Trebak M, Galbiati F. Purinergic signaling promotes premature senescence. J Biol Chem 2024; 300:107145. [PMID: 38460941 PMCID: PMC11002311 DOI: 10.1016/j.jbc.2024.107145] [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: 01/24/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/11/2024] Open
Abstract
Extracellular ATP activates P2 purinergic receptors. Whether purinergic signaling is functionally coupled to cellular senescence is largely unknown. We find that oxidative stress induced release of ATP and caused senescence in human lung fibroblasts. Inhibition of P2 receptors limited oxidative stress-induced senescence, while stimulation with exogenous ATP promoted premature senescence. Pharmacological inhibition of P2Y11 receptor (P2Y11R) inhibited premature senescence induced by either oxidative stress or ATP, while stimulation with a P2Y11R agonist was sufficient to induce cellular senescence. Our data show that both extracellular ATP and a P2Y11R agonist induced calcium (Ca++) release from the endoplasmic reticulum (ER) and that either inhibition of phospholipase C or intracellular Ca++ chelation impaired ATP-induced senescence. We also find that Ca++ that was released from the ER, following ATP-mediated activation of phospholipase C, entered mitochondria in a manner dependent on P2Y11R activation. Once in mitochondria, excessive Ca++ promoted the production of reactive oxygen species in a P2Y11R-dependent fashion, which drove development of premature senescence of lung fibroblasts. Finally, we show that conditioned medium derived from senescent lung fibroblasts, which were induced to senesce through the activation of ATP/P2Y11R-mediated signaling, promoted the proliferation of triple-negative breast cancer cells and their tumorigenic potential by secreting amphiregulin. Our study identifies the existence of a novel purinergic signaling pathway that links extracellular ATP to the development of a protumorigenic premature senescent phenotype in lung fibroblasts that is dependent on P2Y11R activation and ER-to-mitochondria calcium signaling.
Collapse
Affiliation(s)
- Daniela Volonte
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Cory J Benson
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Stephanie L Daugherty
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jonathan M Beckel
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mohamed Trebak
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ferruccio Galbiati
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
30
|
McAllister BB, Stokes-Heck S, Harding EK, van den Hoogen NJ, Trang T. Targeting Pannexin-1 Channels: Addressing the 'Gap' in Chronic Pain. CNS Drugs 2024; 38:77-91. [PMID: 38353876 DOI: 10.1007/s40263-024-01061-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/07/2024] [Indexed: 02/22/2024]
Abstract
Chronic pain complicates many diseases and is notoriously difficult to treat. In search of new therapeutic targets, pannexin-1 (Panx1) channels have sparked intense interest as a key mechanism involved in a variety of chronic pain conditions. Panx1 channels are transmembrane proteins that release ions and small molecules, such as adenosine triphosphate (ATP). They are expressed along important nodes of the pain pathway, modulating activity of diverse cell types implicated in the development and progression of chronic pain caused by injury or pathology. This review highlights advances that have unlocked the core structure and machinery controlling Panx1 function with a focus on understanding and treating chronic pain.
Collapse
Affiliation(s)
- Brendan B McAllister
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Sierra Stokes-Heck
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Erika K Harding
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Nynke J van den Hoogen
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Tuan Trang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
31
|
Fukuyama K, Motomura E, Okada M. Age-Dependent Activation of Pannexin1 Function Contributes to the Development of Epileptogenesis in Autosomal Dominant Sleep-related Hypermotor Epilepsy Model Rats. Int J Mol Sci 2024; 25:1619. [PMID: 38338895 PMCID: PMC10855882 DOI: 10.3390/ijms25031619] [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: 01/12/2024] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
To explore the processes of epileptogenesis/ictogenesis, this study determined the age-dependent development of the functional abnormalities in astroglial transmission associated with pannexin1-hemichannel using a genetic rat model of autosomal dominant sleep-related hypermotor epilepsy (ADSHE) named 'S286L-TG'. Pannexin1 expression in the plasma membrane of primary cultured cortical astrocytes and the orbitofrontal cortex (OFC), which is an ADSHE focus region, were determined using capillary immunoblotting. Astroglial D-serine releases induced by artificial high-frequency oscillation (HFO)-evoked stimulation, the removal of extracellular Ca2+, and the P2X7 receptor agonist (BzATP) were determined using ultra-high performance liquid chromatography (UHPLC). The expressions of pannexin1 in the plasma membrane fraction of the OFC in S286L-TG at four weeks old were almost equivalent when compared to the wild type. The pannexin1 expression in the OFC of the wild type non-statistically decreased age-dependently, whereas that in S286L-TG significantly increased age-dependently, resulting in relatively increasing pannexin1 expression from the 7- (at the onset of interictal discharge) and 10-week-old (after the ADSHE seizure onset) S286L-TG compared to the wild type. However, no functional abnormalities of astroglial pannexin1 expression or D-serine release through the pannexin1-hemichannels from the cultured astrocytes of S286L-TG could be detected. Acutely HFO-evoked stimulation, such as physiological ripple burst (200 Hz) and epileptogenic fast ripple burst (500 Hz), frequency-dependently increased both pannexin1 expression in the astroglial plasma membrane and astroglial D-serine release. Neither the selective inhibitors of pannexin1-hemichannel (10PANX) nor connexin43-hemichannel (Gap19) affected astroglial D-serine release during the resting stage, whereas HFO-evoked D-serine release was suppressed by both inhibitors. The inhibitory effect of 10PANX on the ripple burst-evoked D-serine release was more predominant than that of Gap19, whereas fast ripple burst-evoked D-serine release was predominantly suppressed by Gap19 rather than 10PANX. Astroglial D-serine release induced by acute exposure to BzATP was suppressed by 10PANX but not by Gap19. These results suggest that physiological ripple burst during the sleep spindle plays important roles in the organization of some components of cognition in healthy individuals, but conversely, it contributes to the initial development of epileptogenesis/ictogenesis in individuals who have ADSHE vulnerability via activation of the astroglial excitatory transmission associated with pannexin1-hemichannels.
Collapse
Affiliation(s)
| | | | - Motohiro Okada
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan; (K.F.); (E.M.)
| |
Collapse
|
32
|
Cibelli A, Dohare P, Spray DC, Scemes E. Differential activation of mouse and human Panx1 channel variants. PLoS One 2023; 18:e0295710. [PMID: 38100403 PMCID: PMC10723736 DOI: 10.1371/journal.pone.0295710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
Pannexins are ubiquitously expressed in human and mouse tissues. Pannexin 1 (Panx1), the most thoroughly characterized member of this family, forms plasmalemmal membrane channels permeable to relatively large molecules, such as ATP. Although human and mouse Panx1 amino acid sequences are conserved in the presently known regulatory sites involved in trafficking and modulation of the channel, differences are reported in the N- and C-termini of the protein, and the mechanisms of channel activation by different stimuli remain controversial. Here we used a neuroblastoma cell line to study the activation properties of endogenous mPanx1 and exogenously expressed hPanx1. Dye uptake and electrophysiological recordings revealed that in contrast to mouse Panx1, the human ortholog is insensitive to stimulation with high extracellular [K+] but responds similarly to activation of the purinergic P2X7 receptor. The two most frequent Panx1 polymorphisms found in the human population, Q5H (rs1138800) and E390D (rs74549886), exogenously expressed in Panx1-null N2a cells revealed that regarding P2X7 receptor mediated Panx1 activation, the Q5H mutant is a gain of function whereas the E390D mutant is a loss of function variant. Collectively, we demonstrate differences in the activation between human and mouse Panx1 orthologs and suggest that these differences may have translational implications for studies where Panx1 has been shown to have significant impact.
Collapse
Affiliation(s)
- Antonio Cibelli
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Preeti Dohare
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - David C. Spray
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Eliana Scemes
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States of America
| |
Collapse
|
33
|
O'Donnell BL, Penuela S. Skin in the game: pannexin channels in healthy and cancerous skin. Biochem J 2023; 480:1929-1949. [PMID: 38038973 DOI: 10.1042/bcj20230176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
The skin is a highly organized tissue composed of multiple layers and cell types that require coordinated cell to cell communication to maintain tissue homeostasis. In skin cancer, this organized structure and communication is disrupted, prompting the malignant transformation of healthy cells into melanoma, basal cell carcinoma or squamous cell carcinoma tumours. One such family of channel proteins critical for cellular communication is pannexins (PANX1, PANX2, PANX3), all of which are present in the skin. These heptameric single-membrane channels act as conduits for small molecules and ions like ATP and Ca2+ but have also been shown to have channel-independent functions through their interacting partners or action in signalling pathways. Pannexins have diverse roles in the skin such as in skin development, aging, barrier function, keratinocyte differentiation, inflammation, and wound healing, which were discovered through work with pannexin knockout mice, organotypic epidermis models, primary cells, and immortalized cell lines. In the context of cutaneous cancer, PANX1 is present at high levels in melanoma tumours and functions in melanoma carcinogenesis, and both PANX1 and PANX3 expression is altered in non-melanoma skin cancer. PANX2 has thus far not been implicated in any skin cancer. This review will discuss pannexin isoforms, structure, trafficking, post-translational modifications, interactome, and channel activity. We will also outline the expression, localization, and function of pannexin channels within the diverse cell types of the epidermis, dermis, hypodermis, and adnexal structures of the skin, and how these properties are exploited or abrogated in instances of skin cancer.
Collapse
Affiliation(s)
- Brooke L O'Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
- Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| |
Collapse
|
34
|
Chai XX, Liu J, Yu TY, Zhang G, Sun WJ, Zhou Y, Ren L, Cao HL, Yin DC, Zhang CY. Recent progress of mechanosensitive mechanism on breast cancer. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 185:1-16. [PMID: 37793504 DOI: 10.1016/j.pbiomolbio.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/10/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
The mechanical environment is important for tumorigenesis and progression. Tumor cells can sense mechanical signals by mechanosensitive receptors, and these mechanical signals can be converted to biochemical signals to regulate cell behaviors, such as cell differentiation, proliferation, migration, apoptosis, and drug resistance. Here, we summarized the effects of the mechanical microenvironment on breast cancer cell activity, and mechanotransduction mechanism from cellular microenvironment to cell membrane, and finally to the nucleus, and also relative mechanosensitive proteins, ion channels, and signaling pathways were elaborated, therefore the mechanical signal could be transduced to biochemical or molecular signal. Meanwhile, the mechanical models commonly used for biomechanics study in vitro and some quantitative descriptions were listed. It provided an essential theoretical basis for the occurrence and development of mechanosensitive breast cancer, and also some potential drug targets were proposed to treat such disease.
Collapse
Affiliation(s)
- Xiao-Xia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Jie Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Tong-Yao Yu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Wen-Jun Sun
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Yan Zhou
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Li Ren
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China; Key Laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, Ningbo, 315103, Zhejiang, PR China
| | - Hui-Ling Cao
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, School of Pharmacy, Xi'an Medical University, Xi'an, 710021, Shaanxi, PR China.
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China.
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China.
| |
Collapse
|
35
|
Shao M, Yu H, Santhakumar V, Yu J. Antiepileptogenic and neuroprotective effect of mefloquine after experimental status epilepticus. Epilepsy Res 2023; 198:107257. [PMID: 37989006 DOI: 10.1016/j.eplepsyres.2023.107257] [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: 08/04/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/23/2023]
Abstract
Acquired temporal lobe epilepsy (TLE) characterized by spontaneous recurrent seizures (SRS) and hippocampal inhibitory neuron dysfunction is often refractory to current therapies. Gap junctional or electrical coupling between inhibitory neurons has been proposed to facilitate network synchrony and intercellular molecular exchange suggesting a role in both seizures and neurodegeneration. While gap junction blockers can limit acute seizures, whether blocking neuronal gap junctions can modify development of chronic epilepsy has not been examined. This study examined whether mefloquine, a selective blocker of Connexin 36 gap junctions which are well characterized in inhibitory neurons, can limit epileptogenesis and related cellular and behavioral pathology in a model of acquired TLE. A single, systemic dose of mefloquine administered early after pilocarpine-induced status epilepticus (SE) in rat reduced both development of SRS and behavioral co-morbidities. Immunostaining for interneuron subtypes identified that mefloquine treatment likely reduced delayed inhibitory neuronal loss after SE. Uniquely, parvalbumin expressing neurons in the hippocampal dentate gyrus appeared relatively resistant to early cell loss after SE. Functionally, whole cell patch clamp recordings revealed that mefloquine treatment preserved inhibitory synaptic drive to projection neurons one week and one month after SE. These results demonstrate that mefloquine, a drug already approved for malaria prophylaxis, is potentially antiepileptogenic and can protect against progressive interneuron loss and behavioral co-morbidities of epilepsy.
Collapse
Affiliation(s)
- Mingting Shao
- Department of Neurosurgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Hang Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Vijayalakshmi Santhakumar
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA
| | - Jiandong Yu
- Department of Neurosurgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, China.
| |
Collapse
|
36
|
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
|
37
|
Spurlock M, An W, Reshetnikova G, Wen R, Wang H, Braha M, Solis G, Kurtenbach S, Galindez OJ, de Rivero Vaccari JP, Chou TH, Porciatti V, Shestopalov VI. The Inflammasome-Dependent Dysfunction and Death of Retinal Ganglion Cells after Repetitive Intraocular Pressure Spikes. Cells 2023; 12:2626. [PMID: 37998361 PMCID: PMC10670000 DOI: 10.3390/cells12222626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023] Open
Abstract
The dysfunction and selective loss of retinal ganglion cells (RGCs) is a known cause of vision loss in glaucoma and other neuropathies, where ocular hypertension (OHT) is the major risk factor. We investigated the impact of transient non-ischemic OHT spikes (spOHT) on RGC function and viability in vivo to identify cellular pathways linking low-grade repetitive mechanical stress to RGC pathology. We found that repetitive spOHT had an unexpectedly high impact on intraocular homeostasis and RGC viability, while exposure to steady OHT (stOHT) of a similar intensity and duration failed to induce pathology. The repetitive spOHT induced the rapid activation of the inflammasome, marked by the upregulation of NLRP1, NLRP3, AIM2, caspases -1, -3/7, -8, and Gasdermin D (GSDMD), and the release of interleukin-1β (IL-1β) and other cytokines into the vitreous. Similar effects were also detected after 5 weeks of exposure to chronic OHT in an induced glaucoma model. The onset of these immune responses in both spOHT and glaucoma models preceded a 50% deficit in pattern electroretinogram (PERG) amplitude and a significant loss of RGCs 7 days post-injury. The inactivation of inflammasome complexes in Nlrp1-/-, Casp1-/-, and GsdmD-/- knockout animals significantly suppressed the spOHT-induced inflammatory response and protected RGCs. Our results demonstrate that mechanical stress produced by acute repetitive spOHT or chronic OHT is mechanistically linked to inflammasome activation, which leads to RGC dysfunction and death.
Collapse
Affiliation(s)
- Markus Spurlock
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
- Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Weijun An
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
| | - Galina Reshetnikova
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
| | - Rong Wen
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
| | - Hua Wang
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
| | - Michelle Braha
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
| | - Gabriela Solis
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
| | - Stefan Kurtenbach
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
| | - Orlando J. Galindez
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Juan Pablo de Rivero Vaccari
- Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Tsung-Han Chou
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
| | - Vittorio Porciatti
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
| | - Valery I. Shestopalov
- Bascom Palmer Eye Institute Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (W.A.); (G.R.); (R.W.); (H.W.); (M.B.); (G.S.); (S.K.); (V.P.)
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
38
|
Alarcon-Martinez L, Shiga Y, Villafranca-Baughman D, Cueva Vargas JL, Vidal Paredes IA, Quintero H, Fortune B, Danesh-Meyer H, Di Polo A. Neurovascular dysfunction in glaucoma. Prog Retin Eye Res 2023; 97:101217. [PMID: 37778617 DOI: 10.1016/j.preteyeres.2023.101217] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Retinal ganglion cells, the neurons that die in glaucoma, are endowed with a high metabolism requiring optimal provision of oxygen and nutrients to sustain their activity. The timely regulation of blood flow is, therefore, essential to supply firing neurons in active areas with the oxygen and glucose they need for energy. Many glaucoma patients suffer from vascular deficits including reduced blood flow, impaired autoregulation, neurovascular coupling dysfunction, and blood-retina/brain-barrier breakdown. These processes are tightly regulated by a community of cells known as the neurovascular unit comprising neurons, endothelial cells, pericytes, Müller cells, astrocytes, and microglia. In this review, the neurovascular unit takes center stage as we examine the ability of its members to regulate neurovascular interactions and how their function might be altered during glaucomatous stress. Pericytes receive special attention based on recent data demonstrating their key role in the regulation of neurovascular coupling in physiological and pathological conditions. Of particular interest is the discovery and characterization of tunneling nanotubes, thin actin-based conduits that connect distal pericytes, which play essential roles in the complex spatial and temporal distribution of blood within the retinal capillary network. We discuss cellular and molecular mechanisms of neurovascular interactions and their pathophysiological implications, while highlighting opportunities to develop strategies for vascular protection and regeneration to improve functional outcomes in glaucoma.
Collapse
Affiliation(s)
- Luis Alarcon-Martinez
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada; Centre for Eye Research Australia, University of Melbourne, Melbourne, Australia
| | - Yukihiro Shiga
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Jorge L Cueva Vargas
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Isaac A Vidal Paredes
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Heberto Quintero
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Healthy, Portland, OR, USA
| | - Helen Danesh-Meyer
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada.
| |
Collapse
|
39
|
Wurl JA, Mac Nair CE, Dietz JA, Shestopalov VI, Nickells RW. Contralateral Astrocyte Response to Acute Optic Nerve Damage Is Mitigated by PANX1 Channel Activity. Int J Mol Sci 2023; 24:15641. [PMID: 37958624 PMCID: PMC10647301 DOI: 10.3390/ijms242115641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Glial reactivity is considered a hallmark of damage-induced innate immune responses in the central nervous system. In the visual system, unilateral optic nerve damage elicits dramatic glial reactivity in the retina directly affected by the lesion and a similar, albeit more modest, effect in the contralateral eye. Evaluation of astrocyte changes in a mouse model of optic nerve crush indicates that astrocyte reactivity, as a function of retinal coverage and cellular hypertrophy, occurs within both the experimental and contralateral retinas, although the hypertrophic response of the astrocytes in the contralateral eyes is delayed for at least 24 h. Evaluation of astrocytic reactivity as a function of Gfap expression indicates a similar, muted but significant, response in contralateral eyes. This constrained glial response is completely negated by conditional knock out of Panx1 in both astrocytes and Müller cells. Further studies are required to identify if this is an autocrine or a paracrine suppression of astroglial reactivity.
Collapse
Affiliation(s)
- Jasmine A. Wurl
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.A.W.); (C.E.M.N.); (J.A.D.)
| | - Caitlin E. Mac Nair
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.A.W.); (C.E.M.N.); (J.A.D.)
| | - Joel A. Dietz
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.A.W.); (C.E.M.N.); (J.A.D.)
| | - Valery I. Shestopalov
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL 33136, USA;
| | - Robert W. Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.A.W.); (C.E.M.N.); (J.A.D.)
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
40
|
Li X, Hu J, Yin P, Liu L, Chen Y. Mechanotransduction in the urothelium: ATP signalling and mechanoreceptors. Heliyon 2023; 9:e19427. [PMID: 37674847 PMCID: PMC10477517 DOI: 10.1016/j.heliyon.2023.e19427] [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: 04/23/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
The urothelium, which covers the inner surface of the bladder, is continuously exposed to a complex physical environment where it is stimulated by, and responds to, a wide range of mechanical cues. Mechanically activated ion channels endow the urothelium with functioning in the conversion of mechanical stimuli into biochemical events that influence the surface of the urothelium itself as well as suburothelial tissues, including afferent nerve fibres, interstitial cells of Cajal and detrusor smooth muscle cells, to ensure normal urinary function during the cycle of filling and voiding. However, under prolonged and abnormal loading conditions, the urothelial sensory system can become maladaptive, leading to the development of bladder dysfunction. In this review, we summarize developments in the understanding of urothelial mechanotransduction from two perspectives: first, with regard to the functions of urothelial mechanotransduction, particularly stretch-mediated ATP signalling and the regulation of urothelial surface area; and secondly, with regard to the mechanoreceptors present in the urothelium, primarily transient receptor potential channels and mechanosensitive Piezo channels, and the potential pathophysiological role of these channels in the bladder. A more thorough understanding of urothelial mechanotransduction function may inspire the development of new therapeutic strategies for lower urinary tract diseases.
Collapse
Affiliation(s)
| | | | - Ping Yin
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Lumin Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yuelai Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| |
Collapse
|
41
|
Güiza J, Solís F, Valenzuela B, Arancibia D, Zamorano P, González J, Saavedra J, Neely A, Salgado M, Martínez AD, Sáez JC, Vega JL. Unnexin is a protein subunit of a large-pore channel expressed by unicellular organisms. Proc Natl Acad Sci U S A 2023; 120:e2307898120. [PMID: 37487087 PMCID: PMC10400985 DOI: 10.1073/pnas.2307898120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/23/2023] [Indexed: 07/26/2023] Open
Abstract
Cells of vertebrate and invertebrate organisms express proteins specialized in membrane channel-based cell-cell communication that are absent in unicellular organisms. We recently described the prediction of some members of the large-pore channel family in kinetoplastids, consisting of proteins called unnexins, which share several structural features with innexin and pannexin proteins. Here, we demonstrated that the unnexin1 protein (Unx1) is delivered to the cell membrane, displaying a topology consisting of four transmembrane domains with C and N termini on the cytoplasmic side and form large-pore channels that are permeable to small molecules. Low extracellular Ca2+/Mg2+ levels or extracellular alkalinization, but not mechanical stretching, increases channel activity. The Unx1 channel mediates the influx of Ca2+ and does not form intercellular dye coupling between HeLa Unx1 transfected cells. Unx1 channel function was further evidenced by its ability to mediate ionic currents when expressed in Xenopus oocytes. Downregulation of Unx1 mRNA with morpholine contains Trypanosoma cruzi invasion. Phylogenetic analysis revealed the presence of Unx1 homologs in other protozoan parasites, suggesting a conserved function for these channel parasites in other protists. Our data demonstrate that Unx1 forms large-pore membrane channels, which may serve as a diffusional pathway for ions and small molecules that are likely to be metabolic substrates or waste products, and signaling autocrine and paracrine molecules that could be involved in cell invasion. As morpholinos-induced downregulation of Unx1 reduces the infectivity of trypomastigotes, the Unx1 channels might be an attractive target for developing trypanocide drugs.
Collapse
Affiliation(s)
- Juan Güiza
- Laboratory of Gap Junction Proteins and Parasitic Diseases (GaPaL), Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Francisco Solís
- Laboratorio de Microorganismos Extremófilos, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Bernardita Valenzuela
- Laboratorio de Microorganismos Extremófilos, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Duxan Arancibia
- Laboratorio de Microorganismos Extremófilos, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Pedro Zamorano
- Laboratorio de Microorganismos Extremófilos, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Jorge González
- Departamento de Tecnología Médica, Unidad de Parasitología Molecular, Facultad Ciencias de la Salud, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Jonathan Saavedra
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2381850, Chile
| | - Alan Neely
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2381850, Chile
| | - Magdiel Salgado
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2381850, Chile
| | - Agustín D. Martínez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2381850, Chile
| | - Juan C. Sáez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2381850, Chile
| | - José L. Vega
- Laboratory of Gap Junction Proteins and Parasitic Diseases (GaPaL), Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
- Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta1240000, Chile
| |
Collapse
|
42
|
Truong NV, Phan TTT, Hsu TS, Phu Duc P, Lin LY, Wu WG. Action mechanism of snake venom l-amino acid oxidase and its double-edged sword effect on cancer treatment: Role of pannexin 1-mediated interleukin-6 expression. Redox Biol 2023; 64:102791. [PMID: 37385076 PMCID: PMC10331595 DOI: 10.1016/j.redox.2023.102791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/16/2023] [Indexed: 07/01/2023] Open
Abstract
Snake venom l-amino acid oxidases (svLAAOs) have been recognized as promising candidates for anticancer therapeutics. However, multiple aspects of their catalytic mechanism and the overall responses of cancer cells to these redox enzymes remain ambiguous. Here, we present an analysis of the phylogenetic relationships and active site-related residues among svLAAOs and reveal that the previously proposed critical catalytic residue His 223 is highly conserved in the viperid but not the elapid svLAAO clade. To gain further insight into the action mechanism of the elapid svLAAOs, we purify and characterize the structural, biochemical, and anticancer therapeutic potentials of the Thailand elapid snake Naja kaouthia LAAO (NK-LAAO). We find that NK-LAAO, with Ser 223, exhibits high catalytic activity toward hydrophobic l-amino acid substrates. Moreover, NK-LAAO induces substantial oxidative stress-mediated cytotoxicity with the magnitude relying on both the levels of extracellular hydrogen peroxide (H2O2) and intracellular reactive oxygen species (ROS) generated during the enzymatic redox reactions, but not being influenced by the N-linked glycans on its surface. Unexpectedly, we discover a tolerant mechanism deployed by cancer cells to dampen the anticancer activities of NK-LAAO. NK-LAAO treatment amplifies interleukin (IL)-6 expression via the pannexin 1 (Panx1)-directed intracellular calcium (iCa2+) signaling pathway to confer adaptive and aggressive phenotypes on cancer cells. Accordingly, IL-6 silencing renders cancer cells vulnerable to NK-LAAO-induced oxidative stress together with abrogating NK-LAAO-stimulated metastatic acquisition. Collectively, our study urges caution when using svLAAOs in cancer treatment and identifies the Panx1/iCa2+/IL-6 axis as a therapeutic target for improving the effectiveness of svLAAOs-based anticancer therapies.
Collapse
Affiliation(s)
- Nam V Truong
- Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Trinh T T Phan
- Institute of Molecular and Cellular Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Phan Phu Duc
- Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Lih-Yuan Lin
- Institute of Molecular and Cellular Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC.
| | - Wen-Guey Wu
- Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC.
| |
Collapse
|
43
|
Gou Z, Zhang H, Misbah C. Heterogeneous ATP patterns in microvascular networks. J R Soc Interface 2023; 20:20230186. [PMID: 37464803 PMCID: PMC10354495 DOI: 10.1098/rsif.2023.0186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/20/2023] Open
Abstract
ATP is not only an energy carrier but also serves as an important signalling molecule in many physiological processes. Abnormal ATP level in blood vessel is known to be related to several pathologies, such as inflammation, hypoxia and atherosclerosis. Using advanced numerical methods, we analysed ATP released by red blood cells (RBCs) and its degradation by endothelial cells (ECs) in a cat mesentery-inspired vascular network, accounting for RBC mutual interaction and interactions with vascular walls. Our analysis revealed a heterogeneous ATP distribution in the network, with higher concentrations in the cell-free layer, concentration peaks around bifurcations and heterogeneity among vessels of the same level. These patterns arise from the spatio-temporal organization of RBCs induced by the network geometry. It is further shown that an alteration of hematocrit and flow strength significantly affects ATP level as well as heterogeneity in the network. These findings constitute a first building block to elucidate the intricate nature of ATP patterns in vascular networks and the far reaching consequences for other biochemical signalling, such as calcium, by ECs.
Collapse
Affiliation(s)
- Zhe Gou
- CNRS, LIPhy, Université Grenoble Alpes, 38000 Grenoble, France
| | - Hengdi Zhang
- CNRS, LIPhy, Université Grenoble Alpes, 38000 Grenoble, France
- Shenzhen Sibionics Co. Ltd, Shenzhen, People’s Republic of China
| | - Chaouqi Misbah
- CNRS, LIPhy, Université Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
44
|
Cauli B, Dusart I, Li D. Lactate as a determinant of neuronal excitability, neuroenergetics and beyond. Neurobiol Dis 2023:106207. [PMID: 37331530 DOI: 10.1016/j.nbd.2023.106207] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023] Open
Abstract
Over the last decades, lactate has emerged as important energy substrate for the brain fueling of neurons. A growing body of evidence now indicates that it is also a signaling molecule modulating neuronal excitability and activity as well as brain functions. In this review, we will briefly summarize how different cell types produce and release lactate. We will further describe different signaling mechanisms allowing lactate to fine-tune neuronal excitability and activity, and will finally discuss how these mechanisms could cooperate to modulate neuroenergetics and higher order brain functions both in physiological and pathological conditions.
Collapse
Affiliation(s)
- Bruno Cauli
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS), 9 quai Saint Bernard, 75005 Paris, France.
| | - Isabelle Dusart
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS), 9 quai Saint Bernard, 75005 Paris, France
| | - Dongdong Li
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS), 9 quai Saint Bernard, 75005 Paris, France
| |
Collapse
|
45
|
Aresta Branco MSL, Gutierrez Cruz A, Peri LE, Mutafova-Yambolieva VN. The Pannexin 1 Channel and the P2X7 Receptor Are in Complex Interplay to Regulate the Release of Soluble Ectonucleotidases in the Murine Bladder Lamina Propria. Int J Mol Sci 2023; 24:9964. [PMID: 37373111 PMCID: PMC10298213 DOI: 10.3390/ijms24129964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The bladder urothelium releases ATP into the lamina propria (LP) during filling, which can activate P2X receptors on afferent neurons and trigger the micturition reflex. Effective ATP concentrations are largely dependent on metabolism by membrane-bound and soluble ectonucleotidases (s-ENTDs), and the latter are released in the LP in a mechanosensitive manner. Pannexin 1 (PANX1) channel and P2X7 receptor (P2X7R) participate in urothelial ATP release and are physically and functionally coupled, hence we investigated whether they modulate s-ENTDs release. Using ultrasensitive HPLC-FLD, we evaluated the degradation of 1,N6-etheno-ATP (eATP, substrate) to eADP, eAMP, and e-adenosine (e-ADO) in extraluminal solutions that were in contact with the LP of mouse detrusor-free bladders during filling prior to substrate addition, as an indirect measure of s-ENDTS release. Deletion of Panx1 increased the distention-induced, but not the spontaneous, release of s-ENTDs, whereas activation of P2X7R by BzATP or high concentration of ATP in WT bladders increased both. In Panx1-/- bladders or WT bladders treated with the PANX1 inhibitory peptide 10Panx, however, BzATP had no effect on s-ENTDS release, suggesting that P2X7R activity depends on PANX1 channel opening. We concluded, therefore, that P2X7R and PANX1 are in complex interaction to regulate s-ENTDs release and maintain suitable ATP concentrations in the LP. Thus, while stretch-activated PANX1 hinders s-ENTDS release possibly to preserve effective ATP concentration at the end of bladder filling, P2X7R activation, presumably in cystitis, would facilitate s-ENTDs-mediated ATP degradation to counteract excessive bladder excitability.
Collapse
Affiliation(s)
| | | | | | - Violeta N. Mutafova-Yambolieva
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada Reno, Reno, NV 89557, USA; (M.S.L.A.B.); (A.G.C.); (L.E.P.)
| |
Collapse
|
46
|
Oliveira-Mendes BBR, Alameh M, Ollivier B, Montnach J, Bidère N, Souazé F, Escriou N, Charpentier F, Baró I, De Waard M, Loussouarn G. SARS-CoV-2 E and 3a Proteins Are Inducers of Pannexin Currents. Cells 2023; 12:1474. [PMID: 37296595 PMCID: PMC10252541 DOI: 10.3390/cells12111474] [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: 03/13/2023] [Revised: 05/10/2023] [Accepted: 05/18/2023] [Indexed: 06/12/2023] Open
Abstract
Controversial reports have suggested that SARS-CoV E and 3a proteins are plasma membrane viroporins. Here, we aimed at better characterizing the cellular responses induced by these proteins. First, we show that expression of SARS-CoV-2 E or 3a protein in CHO cells gives rise to cells with newly acquired round shapes that detach from the Petri dish. This suggests that cell death is induced upon expression of E or 3a protein. We confirmed this by using flow cytometry. In adhering cells expressing E or 3a protein, the whole-cell currents were not different from those of the control, suggesting that E and 3a proteins are not plasma membrane viroporins. In contrast, recording the currents on detached cells uncovered outwardly rectifying currents much larger than those observed in the control. We illustrate for the first time that carbenoxolone and probenecid block these outwardly rectifying currents; thus, these currents are most probably conducted by pannexin channels that are activated by cell morphology changes and also potentially by cell death. The truncation of C-terminal PDZ binding motifs reduces the proportion of dying cells but does not prevent these outwardly rectifying currents. This suggests distinct pathways for the induction of these cellular events by the two proteins. We conclude that SARS-CoV-2 E and 3a proteins are not viroporins expressed at the plasma membrane.
Collapse
Affiliation(s)
| | - Malak Alameh
- L’institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (B.B.R.O.-M.); (M.A.)
- Labex Ion Channels, Science and Therapeutics, F-06560 Valbonne, France
| | - Béatrice Ollivier
- L’institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (B.B.R.O.-M.); (M.A.)
| | - Jérôme Montnach
- L’institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (B.B.R.O.-M.); (M.A.)
| | - Nicolas Bidère
- Team SOAP, CRCI2NA, INSERM, CNRS, Nantes Université, Université d’Angers, F-44000 Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France
| | | | - Nicolas Escriou
- Institut Pasteur, Université Paris Cité, Département de Santé Globale, F-75015 Paris, France
| | - Flavien Charpentier
- L’institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (B.B.R.O.-M.); (M.A.)
| | - Isabelle Baró
- L’institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (B.B.R.O.-M.); (M.A.)
| | - Michel De Waard
- L’institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (B.B.R.O.-M.); (M.A.)
- Labex Ion Channels, Science and Therapeutics, F-06560 Valbonne, France
| | - Gildas Loussouarn
- L’institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (B.B.R.O.-M.); (M.A.)
| |
Collapse
|
47
|
García-Rodríguez C, Mujica P, Illanes-González J, López A, Vargas C, Sáez JC, González-Jamett A, Ardiles ÁO. Probenecid, an Old Drug with Potential New Uses for Central Nervous System Disorders and Neuroinflammation. Biomedicines 2023; 11:1516. [PMID: 37371611 DOI: 10.3390/biomedicines11061516] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
Probenecid is an old uricosuric agent used in clinics to treat gout and reduce the renal excretion of antibiotics. In recent years, probenecid has gained attention due to its ability to interact with membrane proteins such as TRPV2 channels, organic anion transporters, and pannexin 1 hemichannels, which suggests new potential therapeutic utilities in medicine. Some current functions of probenecid include their use as an adjuvant to increase the bioavailability of several drugs in the Central Nervous System (CNS). Numerous studies also suggest that this drug has important neuroprotective, antiepileptic, and anti-inflammatory properties, as evidenced by their effect against neurological and neurodegenerative diseases. In these studies, the use of probenecid as a Panx1 hemichannel blocker to reduce neuroinflammation is highlighted since neuroinflammation is a major trigger for diverse CNS alterations. Although the clinical use of probenecid has declined over the years, advances in its use in preclinical research indicate that it may be useful to improve conventional therapies in the psychiatric field where the drugs used have a low bioavailability, either because of a deficient passage through the blood-brain barrier or a high efflux from the CNS or also a high urinary clearance. This review summarizes the history, pharmacological properties, and recent research uses of probenecid and discusses its future projections as a potential pharmacological strategy to intervene in neurodegeneration as an outcome of neuroinflammation.
Collapse
Affiliation(s)
- Claudia García-Rodríguez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Paula Mujica
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Javiera Illanes-González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Araceli López
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Camilo Vargas
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Juan C Sáez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Arlek González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Álvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| |
Collapse
|
48
|
Astigiano C, Piacente F, Laugieri ME, Benzi A, Di Buduo CA, Miguel CP, Soncini D, Cea M, Antonelli A, Magnani M, Balduini A, De Flora A, Bruzzone S. Sirtuin 6 Regulates the Activation of the ATP/Purinergic Axis in Endothelial Cells. Int J Mol Sci 2023; 24:ijms24076759. [PMID: 37047732 PMCID: PMC10095398 DOI: 10.3390/ijms24076759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
Sirtuin 6 (SIRT6) is a member of the mammalian NAD+-dependent deac(et)ylase sirtuin family. SIRT6’s anti-inflammatory roles are emerging increasingly often in different diseases and cell types, including endothelial cells. In this study, the role of SIRT6 in pro-inflammatory conditions was investigated by engineering human umbilical vein endothelial cells to overexpress SIRT6 (SIRT6+ HUVECs). Our results showed that SIRT6 overexpression affected the levels of adhesion molecules and sustained megakaryocyte proliferation and proplatelet formation. Interestingly, the pro-inflammatory activation of the ATP/purinergic axis was reduced in SIRT6+ HUVECs. Specifically, the TNFα-induced release of ATP in the extracellular space and the increase in pannexin-1 hemichannel expression, which mediates ATP efflux, were hampered in SIRT6+ cells. Instead, NAD+ release and Connexin43 expression were not modified by SIRT6 levels. Moreover, the Ca2+ influx in response to ATP and the expression of the purinergic receptor P2X7 were decreased in SIRT6+ HUVECs. Contrary to extracellular ATP, extracellular NAD+ did not evoke pro-inflammatory responses in HUVECs. Instead, NAD+ administration reduced endothelial cell proliferation and motility and counteracted the TNFα-induced angiogenesis. Altogether, our data reinforce the view of SIRT6 activation as an anti-inflammatory approach in vascular endothelium.
Collapse
Affiliation(s)
- Cecilia Astigiano
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
| | - Francesco Piacente
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
| | - Maria Elena Laugieri
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
| | - Andrea Benzi
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
| | - Christian A. Di Buduo
- Department of Molecular Medicine, University of Pavia, Via C. Forlanini 6, 27100 Pavia, Italy
| | - Carolina P. Miguel
- Department of Molecular Medicine, University of Pavia, Via C. Forlanini 6, 27100 Pavia, Italy
| | - Debora Soncini
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
| | - Michele Cea
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo R. Benzi, 16132 Genova, Italy
| | - Antonella Antonelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Via C. Forlanini 6, 27100 Pavia, Italy
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Antonio De Flora
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo R. Benzi, 16132 Genova, Italy
| |
Collapse
|
49
|
Dale N, Butler J, Dospinescu VM, Nijjar S. Channel-mediated ATP release in the nervous system. Neuropharmacology 2023; 227:109435. [PMID: 36690324 DOI: 10.1016/j.neuropharm.2023.109435] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023]
Abstract
ATP is well established as a transmitter and modulator in the peripheral and central nervous system. While conventional exocytotic release of ATP at synapses occurs, this transmitter is unusual in also being released into the extracellular space via large-pored plasma membrane channels. This review considers the channels that are known to be permeable to ATP and some of the functions of channel-mediated ATP release. While the possibility of ATP release via channels mediating volume transmission has been known for some time, localised ATP release via channels at specialised synapses made by taste cells to the afferent nerve has recently been documented in taste buds. This raises the prospect that "channel synapses" may occur in other contexts. However, volume transmission and channel synapses are not necessarily mutually exclusive. We suggest that certain glial cells in the brain stem and hypothalamus, which possess long processes and are known to release ATP, may be candidates for both modes of ATP release -channel-mediated volume transmission in the region of their somata and more localised transmission possibly via either conventional or channel synapses from their processes at distal targets. Finally, we consider the different characteristics of vesicular and channel synapses and suggest that channel synapses may be advantageous in requiring less energy than their conventional vesicular counterparts. This article is part of the Special Issue on "Purinergic Signaling: 50 years".
Collapse
Affiliation(s)
- Nicholas Dale
- School of Life Sciences, University of Warwick, Coventry, CV4, AL, UK.
| | - Jack Butler
- School of Life Sciences, University of Warwick, Coventry, CV4, AL, UK
| | | | - Sarbjit Nijjar
- School of Life Sciences, University of Warwick, Coventry, CV4, AL, UK
| |
Collapse
|
50
|
Yang D, Chen M, Yang S, Deng F, Guo X. Connexin hemichannels and pannexin channels in toxicity: Recent advances and mechanistic insights. Toxicology 2023; 488:153488. [PMID: 36918108 DOI: 10.1016/j.tox.2023.153488] [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: 01/18/2023] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 03/14/2023]
Abstract
Connexin hemichannels and pannexin channels are two types of transmembrane channels that allow autocrine/paracrine signalling through the exchange of ions and molecules between the intra- and extracellular compartments. However, owing to the poor selectivity of permeable ions and metabolites, the massive opening of these plasma membrane channels can lead to an excessive influx of toxic substances and an outflux of essential metabolites, such as adenosine triphosphate, glutathione, glutamate and ions, resulting in unbalanced cell homeostasis and impaired cell function. It is becoming increasingly clear that these channels can be activated in response to external stimuli and are involved in toxicity, yet their concrete mechanistic roles in the toxic effects induced by stress and various environmental changes remain poorly defined. This review provides an updated understanding of connexin hemichannels and pannexin channels in response to multiple extrinsic stressors and how these activated channels and their permeable messengers participate in toxicological pathways and processes, including inflammation, oxidative damage, intracellular calcium imbalance, bystander DNA damage and excitotoxicity.
Collapse
Affiliation(s)
- Di Yang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Mengyuan Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Sijia Yang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Furong Deng
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Xinbiao Guo
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China.
| |
Collapse
|