1
|
Wang J, Levi NJ, Diaz-Solares M, Mim C, Dahl G, Barro-Soria R. A metastasis-associated pannexin-1 mutant (Panx1 1-89) forms a minimalist ATP release channel. FEBS J 2025. [PMID: 40087867 DOI: 10.1111/febs.70060] [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: 06/26/2024] [Revised: 01/20/2025] [Accepted: 02/03/2025] [Indexed: 03/17/2025]
Abstract
A truncated form of the ATP release channel pannexin 1 (Panx1), Panx11-89, is enriched in metastatic breast cancer cells and has been proposed to mediate metastatic cell survival by increasing ATP release through mechanosensitive Panx1 channels. However, whether Panx11-89 on its own [without the presence of wild-type Panx1 (wtPanx1)] mediates ATP release has not been tested. Here, we show that Panx11-89 by itself can form a constitutively active membrane channel, capable of releasing ATP even in the absence of wtPanx1. Our biophysical characterization reveals that most basic structure-function features of the channel pore are conserved in the truncated Panx11-89 polypeptide. Thus, augmenting extracellular potassium ion concentrations enhances Panx11-89-mediated conductance. Moreover, despite the severe truncation, Panx11-89 retains sensitivity to most wtPanx1 channel inhibitors. Therefore, Panx1 blockers may be of therapeutic value to combat metastatic cell survival. Our study both provides a mechanism for ATP release from cancer cells and suggests that Panx11-89 might aid in the structure-function analysis of Panx1 channels.
Collapse
Affiliation(s)
- Junjie Wang
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, FL, USA
| | - Noah J Levi
- Department of Medicine, University of Miami School of Medicine, Miami, FL, USA
| | | | - Carsten Mim
- Department of Biomedical Engineering and Health Systems, Royal Institute of Technology, Huddinge, Sweden
| | - Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, FL, USA
| | - Rene Barro-Soria
- Department of Medicine, University of Miami School of Medicine, Miami, FL, USA
| |
Collapse
|
2
|
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
|
3
|
Weilinger NL, Yang K, Choi HB, Groten CJ, Wendt S, Murugan M, Wicki-Stordeur LE, Bernier LP, Velayudhan PS, Zheng J, LeDue JM, Rungta RL, Tyson JR, Snutch TP, Wu LJ, MacVicar BA. Pannexin-1 opening in neuronal edema causes cell death but also leads to protection via increased microglia contacts. Cell Rep 2023; 42:113128. [PMID: 37742194 PMCID: PMC10824275 DOI: 10.1016/j.celrep.2023.113128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/26/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
Neuronal swelling during cytotoxic edema is triggered by Na+ and Cl- entry and is Ca2+ independent. However, the causes of neuronal death during swelling are unknown. Here, we investigate the role of large-conductance Pannexin-1 (Panx1) channels in neuronal death during cytotoxic edema. Panx1 channel inhibitors reduce and delay neuronal death in swelling triggered by voltage-gated Na+ entry with veratridine. Neuronal swelling causes downstream production of reactive oxygen species (ROS) that opens Panx1 channels. We confirm that ROS activates Panx1 currents with whole-cell electrophysiology and find scavenging ROS is neuroprotective. Panx1 opening and subsequent ATP release attract microglial processes to contact swelling neurons. Depleting microglia using the CSF1 receptor antagonist PLX3397 or blocking P2Y12 receptors exacerbates neuronal death, suggesting that the Panx1-ATP-dependent microglia contacts are neuroprotective. We conclude that cytotoxic edema triggers oxidative stress in neurons that opens Panx1 to trigger death but also initiates neuroprotective feedback mediated by microglia contacts.
Collapse
Affiliation(s)
- Nicholas L Weilinger
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Kai Yang
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hyun B Choi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Christopher J Groten
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Stefan Wendt
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | | - Leigh E Wicki-Stordeur
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Louis-Philippe Bernier
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Prashanth S Velayudhan
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jeffrey M LeDue
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ravi L Rungta
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Stomatology and Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - John R Tyson
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Terrance P Snutch
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Brian A MacVicar
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
4
|
Suarez-Berumen K, Collins-Hooper H, Gromova A, Meech R, Sacco A, Dash PR, Mitchell R, Shestopalov VI, Woolley TE, Vaiyapuri S, Patel K, Makarenkova HP. Pannexin 1 Regulates Skeletal Muscle Regeneration by Promoting Bleb-Based Myoblast Migration and Fusion Through a Novel Lipid Based Signaling Mechanism. Front Cell Dev Biol 2021; 9:736813. [PMID: 34676213 PMCID: PMC8523994 DOI: 10.3389/fcell.2021.736813] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Adult skeletal muscle has robust regenerative capabilities due to the presence of a resident stem cell population called satellite cells. Muscle injury leads to these normally quiescent cells becoming molecularly and metabolically activated and embarking on a program of proliferation, migration, differentiation, and fusion culminating in the repair of damaged tissue. These processes are highly coordinated by paracrine signaling events that drive cytoskeletal rearrangement and cell-cell communication. Pannexins are a family of transmembrane channel proteins that mediate paracrine signaling by ATP release. It is known that Pannexin1 (Panx1) is expressed in skeletal muscle, however, the role of Panx1 during skeletal muscle development and regeneration remains poorly understood. Here we show that Panx1 is expressed on the surface of myoblasts and its expression is rapidly increased upon induction of differentiation and that Panx1-/- mice exhibit impaired muscle regeneration after injury. Panx1-/- myoblasts activate the myogenic differentiation program normally, but display marked deficits in migration and fusion. Mechanistically, we show that Panx1 activates P2 class purinergic receptors, which in turn mediate a lipid signaling cascade in myoblasts. This signaling induces bleb-driven amoeboid movement that in turn supports myoblast migration and fusion. Finally, we show that Panx1 is involved in the regulation of cell-matrix interaction through the induction of ADAMTS (Disintegrin-like and Metalloprotease domain with Thrombospondin-type 5) proteins that help remodel the extracellular matrix. These studies reveal a novel role for lipid-based signaling pathways activated by Panx1 in the coordination of myoblast activities essential for skeletal muscle regeneration.
Collapse
Affiliation(s)
- Katia Suarez-Berumen
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States.,West Anaheim Medical Center, Anaheim, CA, United States
| | | | - Anastasia Gromova
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States.,Development, Aging and Regeneration Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Robyn Meech
- Department of Clinical Pharmacology, Flinders University, Adelaide, SA, Australia
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Phil R Dash
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Robert Mitchell
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Valery I Shestopalov
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, United States.,Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
| | - Thomas E Woolley
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Helen P Makarenkova
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
5
|
Mim C, Perkins G, Dahl G. Structure versus function: Are new conformations of pannexin 1 yet to be resolved? J Gen Physiol 2021; 153:e202012754. [PMID: 33835130 PMCID: PMC8042604 DOI: 10.1085/jgp.202012754] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pannexin 1 (Panx1) plays a decisive role in multiple physiological and pathological settings, including oxygen delivery to tissues, mucociliary clearance in airways, sepsis, neuropathic pain, and epilepsy. It is widely accepted that Panx1 exerts its role in the context of purinergic signaling by providing a transmembrane pathway for ATP. However, under certain conditions, Panx1 can also act as a highly selective membrane channel for chloride ions without ATP permeability. A recent flurry of publications has provided structural information about the Panx1 channel. However, while these structures are consistent with a chloride selective channel, none show a conformation with strong support for the ATP release function of Panx1. In this Viewpoint, we critically assess the existing evidence for the function and structure of the Panx1 channel and conclude that the structure corresponding to the ATP permeation pathway is yet to be determined. We also list a set of additional topics needing attention and propose ways to attain the large-pore, ATP-permeable conformation of the Panx1 channel.
Collapse
Affiliation(s)
- Carsten Mim
- Department of Biomedical Engineering and Health Systems Royal Institute of Technology, Huddinge, Sweden
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California, San Diego School of Medicine, La Jolla, CA
| | - Gerhard Dahl
- Department of Physiology, University of Miami School of Medicine, Miami, FL
| |
Collapse
|
6
|
Harcha PA, Garcés P, Arredondo C, Fernández G, Sáez JC, van Zundert B. Mast Cell and Astrocyte Hemichannels and Their Role in Alzheimer's Disease, ALS, and Harmful Stress Conditions. Int J Mol Sci 2021; 22:ijms22041924. [PMID: 33672031 PMCID: PMC7919494 DOI: 10.3390/ijms22041924] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/02/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Considered relevant during allergy responses, numerous observations have also identified mast cells (MCs) as critical effectors during the progression and modulation of several neuroinflammatory conditions, including Alzheimer’s disease (AD) and amyotrophic lateral sclerosis (ALS). MC granules contain a plethora of constituents, including growth factors, cytokines, chemokines, and mitogen factors. The release of these bioactive substances from MCs occurs through distinct pathways that are initiated by the activation of specific plasma membrane receptors/channels. Here, we focus on hemichannels (HCs) formed by connexins (Cxs) and pannexins (Panxs) proteins, and we described their contribution to MC degranulation in AD, ALS, and harmful stress conditions. Cx/Panx HCs are also expressed by astrocytes and are likely involved in the release of critical toxic amounts of soluble factors—such as glutamate, adenosine triphosphate (ATP), complement component 3 derivate C3a, tumor necrosis factor (TNFα), apoliprotein E (ApoE), and certain miRNAs—known to play a role in the pathogenesis of AD, ALS, and other neurodegenerative disorders. We propose that blocking HCs on MCs and glial cells offers a promising novel strategy for ameliorating the progression of neurodegenerative diseases by reducing the release of cytokines and other pro-inflammatory compounds.
Collapse
Affiliation(s)
- Paloma A. Harcha
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Valparaíso 2381850, Chile
- Correspondence: (P.A.H.); (J.C.S.); (B.v.Z.)
| | - Polett Garcés
- Institute of Biomedical Sciences (ICB), Faculty of Medicine & Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370186, Chile; (P.G.); (C.A.); (G.F.)
- CARE Biomedical Research Center, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8330005, Chile
| | - Cristian Arredondo
- Institute of Biomedical Sciences (ICB), Faculty of Medicine & Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370186, Chile; (P.G.); (C.A.); (G.F.)
- CARE Biomedical Research Center, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8330005, Chile
| | - Germán Fernández
- Institute of Biomedical Sciences (ICB), Faculty of Medicine & Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370186, Chile; (P.G.); (C.A.); (G.F.)
- CARE Biomedical Research Center, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8330005, Chile
| | - Juan C. Sáez
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Valparaíso 2381850, Chile
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Correspondence: (P.A.H.); (J.C.S.); (B.v.Z.)
| | - Brigitte van Zundert
- Institute of Biomedical Sciences (ICB), Faculty of Medicine & Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370186, Chile; (P.G.); (C.A.); (G.F.)
- CARE Biomedical Research Center, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8330005, Chile
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Correspondence: (P.A.H.); (J.C.S.); (B.v.Z.)
| |
Collapse
|
7
|
Navis KE, Fan CY, Trang T, Thompson RJ, Derksen DJ. Pannexin 1 Channels as a Therapeutic Target: Structure, Inhibition, and Outlook. ACS Chem Neurosci 2020; 11:2163-2172. [PMID: 32639715 DOI: 10.1021/acschemneuro.0c00333] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Pannexin 1 (Panx1) channels are transmembrane proteins that release adenosine triphosphate and play an important role in intercellular communication. They are widely expressed in somatic and nervous system tissues, and their activity has been associated with many pathologies such as stroke, epilepsy, inflammation, and chronic pain. While there are a variety of small molecules known to inhibit Panx1, currently little is known about the mechanism of channel inhibition, and there is a dearth of sufficiently potent and selective drugs targeting Panx1. Herein we provide a review of the current literature on Panx1 structural biology and known pharmacological agents that will help provide a basis for rational development of Panx1 chemical modulators.
Collapse
Affiliation(s)
- Kathleen E. Navis
- Department of Chemistry, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Churmy Y. Fan
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Tuan Trang
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Roger J. Thompson
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Darren J. Derksen
- Department of Chemistry, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| |
Collapse
|
8
|
Abudara V, Retamal MA, Del Rio R, Orellana JA. Synaptic Functions of Hemichannels and Pannexons: A Double-Edged Sword. Front Mol Neurosci 2018; 11:435. [PMID: 30564096 PMCID: PMC6288452 DOI: 10.3389/fnmol.2018.00435] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/08/2018] [Indexed: 01/18/2023] Open
Abstract
The classical view of synapses as the functional contact between presynaptic and postsynaptic neurons has been challenged in recent years by the emerging regulatory role of glial cells. Astrocytes, traditionally considered merely supportive elements are now recognized as active modulators of synaptic transmission and plasticity at the now so-called "tripartite synapse." In addition, an increasing body of evidence indicates that beyond immune functions microglia also participate in various processes aimed to shape synaptic plasticity. Release of neuroactive compounds of glial origin, -process known as gliotransmission-, constitute a widespread mechanism through which glial cells can either potentiate or reduce the synaptic strength. The prevailing vision states that gliotransmission depends on an intracellular Ca2+/exocytotic-mediated release; notwithstanding, growing evidence is pointing at hemichannels (connexons) and pannexin channels (pannexons) as alternative non-vesicular routes for gliotransmitters efflux. In concurrence with this novel concept, both hemichannels and pannexons are known to mediate the transfer of ions and signaling molecules -such as ATP and glutamate- between the cytoplasm and the extracellular milieu. Importantly, recent reports show that glial hemichannels and pannexons are capable to perceive synaptic activity and to respond to it through changes in their functional state. In this article, we will review the current information supporting the "double edge sword" role of hemichannels and pannexons in the function of central and peripheral synapses. At one end, available data support the idea that these channels are chief components of a feedback control mechanism through which gliotransmitters adjust the synaptic gain in either resting or stimulated conditions. At the other end, we will discuss how the excitotoxic release of gliotransmitters and [Ca2+]i overload linked to the opening of hemichannels/pannexons might impact cell function and survival in the nervous system.
Collapse
Affiliation(s)
- Verónica Abudara
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.,Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Envejecimiento y Regeneración, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, Punta Arenas, 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.,Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes, Santiago, Chile
| |
Collapse
|
9
|
Makarenkova HP, Shah SB, Shestopalov VI. The two faces of pannexins: new roles in inflammation and repair. J Inflamm Res 2018; 11:273-288. [PMID: 29950881 PMCID: PMC6016592 DOI: 10.2147/jir.s128401] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Pannexins belong to a family of ATP-release channels expressed in almost all cell types. An increasing body of literature on pannexins suggests that these channels play dual and sometimes contradictory roles, contributing to normal cell function, as well as to the pathological progression of disease. In this review, we summarize our understanding of pannexin "protective" and "harmful" functions in inflammation, regeneration and mechanical signaling. We also suggest a possible basis for pannexin's dual roles, related to extracellular ATP and K+ levels and the activation of various types of P2 receptors that are associated with pannexin. Finally, we speculate upon therapeutic strategies related to pannexin using eyes, lacrimal glands, and peripheral nerves as examples of interesting therapeutic targets.
Collapse
Affiliation(s)
| | - Sameer B Shah
- Departments of Orthopaedic Surgery and Bioengineering, University of California.,Research Division, Veterans Affairs San Diego Healthcare System, San Diego, CA
| | - Valery I Shestopalov
- Bascom Eye Institute, Department of Ophthalmology, University of Miami, Miami, FL, USA.,Vavilov Institute for General Genetics, Russian Academy of Sciences.,Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
10
|
Dvoriantchikova G, Pronin A, Kurtenbach S, Toychiev A, Chou TH, Yee CW, Prindeville B, Tayou J, Porciatti V, Sagdullaev BT, Slepak VZ, Shestopalov VI. Pannexin 1 sustains the electrophysiological responsiveness of retinal ganglion cells. Sci Rep 2018; 8:5797. [PMID: 29643381 PMCID: PMC5895610 DOI: 10.1038/s41598-018-23894-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/20/2018] [Indexed: 12/16/2022] Open
Abstract
Pannexin 1 (Panx1) forms ATP-permeable membrane channels that play a key role in purinergic signaling in the nervous system in both normal and pathological conditions. In the retina, particularly high levels of Panx1 are found in retinal ganglion cells (RGCs), but the normal physiological function in these cells remains unclear. In this study, we used patch clamp recordings in the intact inner retina to show that evoked currents characteristic of Panx1 channel activity were detected only in RGCs, particularly in the OFF-type cells. The analysis of pattern electroretinogram (PERG) recordings indicated that Panx1 contributes to the electrical output of the retina. Consistently, PERG amplitudes were significantly impaired in the eyes with targeted ablation of the Panx1 gene in RGCs. Under ocular hypertension and ischemic conditions, however, high Panx1 activity permeated cell membranes and facilitated the selective loss of RGCs or stably transfected Neuro2A cells. Our results show that high expression of the Panx1 channel in RGCs is essential for visual function in the inner retina but makes these cells highly sensitive to mechanical and ischemic stresses. These findings are relevant to the pathophysiology of retinal disorders induced by increased intraocular pressure, such as glaucoma.
Collapse
Affiliation(s)
- Galina Dvoriantchikova
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, 900 NW 10 Ave., Miami, FL, 33136, USA
| | - Alexey Pronin
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave., Miami, FL, 33136, USA
| | - Sarah Kurtenbach
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, 900 NW 10 Ave., Miami, FL, 33136, USA
| | - Abduqodir Toychiev
- Department of Ophthalmology, Weill Cornell Medical College, 156 William St., New York, NY, 10038, USA
| | - Tsung-Han Chou
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, 900 NW 10 Ave., Miami, FL, 33136, USA
| | - Christopher W Yee
- Winifred Masterson Burke Medical Research Institute, New York, 785 Mamaroneck Ave., White Plains, NY, 10605, USA
| | - Breanne Prindeville
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, 900 NW 10 Ave., Miami, FL, 33136, USA
| | - Junior Tayou
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave., Miami, FL, 33136, USA
| | - Vittorio Porciatti
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, 900 NW 10 Ave., Miami, FL, 33136, USA
| | - Botir T Sagdullaev
- Department of Ophthalmology, Weill Cornell Medical College, 156 William St., New York, NY, 10038, USA
- Winifred Masterson Burke Medical Research Institute, New York, 785 Mamaroneck Ave., White Plains, NY, 10605, USA
| | - Vladlen Z Slepak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave., Miami, FL, 33136, USA
| | - Valery I Shestopalov
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, 900 NW 10 Ave., Miami, FL, 33136, USA.
- Department of Cell Biology, University of Miami Miller School of Medicine, 1600 NW 10th Ave., Miami, FL, 33136, USA.
- Vavilov Institute for General Genetics, Gubkina Str. 3, Russian Academy of Sciences, Moscow, Russia.
- Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
11
|
Abstract
Adenosine triphosphate (ATP) has been well established as an important extracellular ligand of autocrine signaling, intercellular communication, and neurotransmission with numerous physiological and pathophysiological roles. In addition to the classical exocytosis, non-vesicular mechanisms of cellular ATP release have been demonstrated in many cell types. Although large and negatively charged ATP molecules cannot diffuse across the lipid bilayer of the plasma membrane, conductive ATP release from the cytosol into the extracellular space is possible through ATP-permeable channels. Such channels must possess two minimum qualifications for ATP permeation: anion permeability and a large ion-conducting pore. Currently, five groups of channels are acknowledged as ATP-release channels: connexin hemichannels, pannexin 1, calcium homeostasis modulator 1 (CALHM1), volume-regulated anion channels (VRACs, also known as volume-sensitive outwardly rectifying (VSOR) anion channels), and maxi-anion channels (MACs). Recently, major breakthroughs have been made in the field by molecular identification of CALHM1 as the action potential-dependent ATP-release channel in taste bud cells, LRRC8s as components of VRACs, and SLCO2A1 as a core subunit of MACs. Here, the function and physiological roles of these five groups of ATP-release channels are summarized, along with a discussion on the future implications of understanding these channels.
Collapse
|
12
|
Cabahug-Zuckerman P, Stout RF, Majeska RJ, Thi MM, Spray DC, Weinbaum S, Schaffler MB. Potential role for a specialized β 3 integrin-based structure on osteocyte processes in bone mechanosensation. J Orthop Res 2018; 36:642-652. [PMID: 29087614 PMCID: PMC5839970 DOI: 10.1002/jor.23792] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/21/2017] [Indexed: 02/04/2023]
Abstract
Osteocyte processes are an order of magnitude more sensitive to mechanical loading than their cell bodies. The mechanisms underlying this remarkable mechanosensitivity are not clear, but may be related to the infrequent αV β3 integrin sites where the osteocyte cell processes attach to canalicular walls. These sites develop dramatically elevated strains during load-induced fluid flow in the lacunar-canalicular system and were recently shown to be primary sites for osteocyte-like MLO-Y4 cell mechanotransduction. These αV β3 integrin sites lack typical integrin transduction mechanisms. Rather, stimulation at these sites alters Ca2+ signaling, ATP release and membrane potential. In the current studies, we tested the hypothesis that in authentic osteocytes in situ, key membrane proteins implicated in osteocyte mechanotransduction are preferentially localized at or near to β3 integrin-foci. We analyzed these spatial relationships in mouse bone osteocytes using immunohistochemistry combined with Structured Illumination Super Resolution Microscopy, a method that permits structural resolution at near electron microscopy levels in tissue sections. We discovered that the purinergic channel pannexin1, the ATP-gated purinergic receptor P2 × 7R and the low voltage transiently opened T-type calcium channel CaV3.2-1 all reside in close proximity to β3 integrin attachment foci on osteocyte processes, suggesting a specialized mechanotransduction complex at these sites. We further confirmed this observation on isolated osteocytes in culture using STochasitc Optical Resonance Microscopy. These findings identify a possible structural basis for the unique mechanosensation and transduction capabilities of the osteocyte process. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:642-652, 2018.
Collapse
Affiliation(s)
| | - Randy F. Stout
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine
- Department of Neuroscience, Albert Einstein College of Medicine
| | | | - Mia M. Thi
- Department of Orthopaedic Surgery, Albert Einstein College of Medicine
| | - David C. Spray
- Department of Neuroscience, Albert Einstein College of Medicine
| | - Sheldon Weinbaum
- Department of Biomedical Engineering, The City College of New York
| | | |
Collapse
|
13
|
Whyte-Fagundes P, Siu R, Brown C, Zoidl G. Pannexins in vision, hearing, olfaction and taste. Neurosci Lett 2017; 695:32-39. [PMID: 28495272 DOI: 10.1016/j.neulet.2017.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 04/06/2017] [Accepted: 05/05/2017] [Indexed: 12/25/2022]
Abstract
In mammals, the pannexin gene family consists of three members (Panx1, 2, 3), which represent a class of integral membrane channel proteins sharing some structural features with chordate gap junction proteins, the connexins. Since their discovery in the early 21st century, pannexin expression has been detected throughout the vertebrate body including eye, ear, nose and tongue, making the investigation of the roles of this new class of channel protein in health and disease very appealing. The localization in sensory organs, coupled with unique channel properties and associations with major signaling pathways make Panx1, and its relative's, significant contributors for fundamental functions in sensory perception. Until recently, cell-based studies were at the forefront of pannexin research. Lately, the availability of mice with genetic ablation of pannexins opened new avenues for testing pannexin functions and behavioural phenotyping. Although we are only at the beginning of understanding the roles of pannexins in health and disease, this review summarizes recent advances in elucidating the various emerging roles pannexins play in sensory systems, with an emphasis on unresolved conflicts.
Collapse
Affiliation(s)
- Paige Whyte-Fagundes
- Graduate Program In Biology, Faculty of Science, York University, Toronto, ON, Canada
| | - Ryan Siu
- Graduate Program In Biology, Faculty of Science, York University, Toronto, ON, Canada
| | - Cherie Brown
- Graduate Program In Biology, Faculty of Science, York University, Toronto, ON, Canada
| | - Georg Zoidl
- Department of Biology, Faculty of Science, York University, Toronto, ON, Canada; Center for Vision Research, York University, Toronto, ON, Canada.
| |
Collapse
|
14
|
Abstract
Pannexin channels are newly discovered ATP release channels expressed throughout the body. Pannexin 1 (Panx1) channels have become of great interest as they appear to participate in a multitude of signalling cascades, including regulation of vascular function. Although numerous Panx1 pharmacological inhibitors have been discovered, these inhibitors are not specific for Panx1 and have additional effects on other proteins. Therefore, molecular tools, such as RNA interference and knockout animals, are needed to demonstrate the role of pannexins in various cellular functions. This review focuses on the known roles of Panx1 related to purinergic signalling in the vasculature focusing on post-translational modifications and channel gating mechanisms that may participate in the regulated release of ATP.
Collapse
|
15
|
Abstract
Pannexin-1 (Panx1) forms anion-selective channels with a permeability up to 1 kDa and represents a pathway for the release of cytosolic ATP. Several structurally similar connexin (Cx) proteins have been identified in platelets and shown to play roles in haemostasis and thrombosis. More recently, functional Panx1 channels have been demonstrated on the surface of human platelets [Taylor et al. (2014) J. Thromb. Haemost. 12, 987-998]. Since their identification in the year 2000, several mechanisms have been reported to activate Panx1 channels, including mechanical stimulation, oxygen-glucose deprivation, a rise of [Ca2+]i, caspase cleavage and phosphorylation. Within this review, the regulation of Panx1 channels is discussed, with a focus on how they may contribute to platelet function.
Collapse
|
16
|
Dahl G. ATP release through pannexon channels. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0191. [PMID: 26009770 DOI: 10.1098/rstb.2014.0191] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Extracellular adenosine triphosphate (ATP) serves as a signal for diverse physiological functions, including spread of calcium waves between astrocytes, control of vascular oxygen supply and control of ciliary beat in the airways. ATP can be released from cells by various mechanisms. This review focuses on channel-mediated ATP release and its main enabler, Pannexin1 (Panx1). Six subunits of Panx1 form a plasma membrane channel termed 'pannexon'. Depending on the mode of stimulation, the pannexon has large conductance (500 pS) and unselective permeability to molecules less than 1.5 kD or is a small (50 pS), chloride-selective channel. Most physiological and pathological stimuli induce the large channel conformation, whereas the small conformation so far has only been observed with exclusive voltage activation of the channel. The interaction between pannexons and ATP is intimate. The pannexon is not only the conduit for ATP, permitting ATP efflux from cells down its concentration gradient, but the pannexon is also modulated by ATP. The channel can be activated by ATP through both ionotropic P2X as well as metabotropic P2Y purinergic receptors. In the absence of a control mechanism, this positive feedback loop would lead to cell death owing to the linkage of purinergic receptors with apoptotic processes. A control mechanism preventing excessive activation of the purinergic receptors is provided by ATP binding (with low affinity) to the Panx1 protein and gating the channel shut.
Collapse
Affiliation(s)
- Gerhard Dahl
- School of Medicine, University of Miami, 1600 NW 10th Avenue, Miami, FL 33136, USA
| |
Collapse
|
17
|
Brown IA, McClain JL, Watson RE, Patel BA, Gulbransen BD. Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide. Cell Mol Gastroenterol Hepatol 2015; 2:77-91. [PMID: 26771001 PMCID: PMC4707972 DOI: 10.1016/j.jcmgh.2015.08.007] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND AIMS The concept of enteric glia as regulators of intestinal homeostasis is slowly gaining acceptance as a central concept in neurogastroenterology. Yet how glia contribute to intestinal disease is still poorly understood. Purines generated during inflammation drive enteric neuron death by activating neuronal P2X7 purine receptors (P2X7R), triggering ATP release via neuronal pannexin-1 channels that subsequently recruits intracellular calcium ([Ca2+]i) responses in the surrounding enteric glia. We tested the hypothesis that the activation of enteric glia contributes to neuron death during inflammation. METHODS We studied neuroinflammation in vivo using the 2,4-dinitrobenzenesulfonic acid model of colitis and in situ using whole-mount preparations of human and mouse intestine. Transgenic mice with a targeted deletion of glial connexin-43 (Cx43) [GFAP∷CreERT2+/-/Cx43f/f ] were used to specifically disrupt glial signaling pathways. Mice deficient in inducible nitric oxide (NO) synthase (iNOS-/-) were used to study NO production. Protein expression and oxidative stress were measured using immunohistochemistry and in situ Ca2+ and NO imaging were used to monitor glial [Ca2+]i and [NO]i. RESULTS Purinergic activation of enteric glia drove [Ca2+]i responses and enteric neuron death through a Cx43-dependent mechanism. Neurotoxic Cx43 activity, driven by NO production from glial iNOS, was required for neuron death. Glial Cx43 opening liberated ATP and Cx43-dependent ATP release was potentiated by NO. CONCLUSIONS Our results show that the activation of glial cells in the context of neuroinflammation kills enteric neurons. Mediators of inflammation that include ATP and NO activate neurotoxic pathways that converge on glial Cx43 hemichannels. The glial response to inflammatory mediators might contribute to the development of motility disorders.
Collapse
Affiliation(s)
- Isola A.M. Brown
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Pharmacology and Toxicology Program, Michigan State University, East Lansing, Michigan
| | | | - Ralph E. Watson
- Department of Medicine, Michigan State University, East Lansing, Michigan
| | - Bhavik A. Patel
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Brian D. Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Neuroscience Program, Michigan State University, East Lansing, Michigan
- Correspondence Address correspondence to: Brian D. Gulbransen, PhD, Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, Michigan 48824. fax: (517) 355-5125.Department of PhysiologyMichigan State University567 Wilson RoadEast LansingMichigan 48824
| |
Collapse
|
18
|
Effects on channel properties and induction of cell death induced by c-terminal truncations of pannexin1 depend on domain length. J Membr Biol 2015; 248:285-94. [PMID: 25567359 DOI: 10.1007/s00232-014-9767-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/22/2014] [Indexed: 10/24/2022]
Abstract
Pannexin1 (Panx1) is an integral membrane protein and known to form multifunctional hexameric channels. Recently, Panx1 was identified to be responsible for the release of ATP and UTP from apoptotic cells after site-specific proteolysis by caspases 3/7. Cleavage at the carboxy-terminal (CT) position aa 376-379 irreversibly opens human Panx1 channels and leads to the release of the respective nucleotides resulting in recruitment of macrophages and in subsequent activation of the immunologic response. The fact that cleavage of the CT at this particular residues terminates in a permanently open channel raised the issue of functional relevance of the CT of Panx1 for regulating channel properties. To analyze the impact of the CT on channel gating, we generated 14 truncated versions of rat Panx1 cleaved at different positions in the C-terminus. This allowed elaboration of the influence of defined residues on channel formation, voltage-dependent gating, execution of cell mortality, and susceptibility to the Panx1 inhibitor carbenoxolone. We demonstrate that expression of Panx1 proteins, which were truncated to lengths between 370 and 393 residues, induces differential effects after expression in Xenopus laevis oocytes as well as in Neuro2A cells with strongest impact downstream the caspase 3/7 cleavage site.
Collapse
|
19
|
Casas M, Buvinic S, Jaimovich E. ATP signaling in skeletal muscle: from fiber plasticity to regulation of metabolism. Exerc Sport Sci Rev 2014; 42:110-6. [PMID: 24949845 DOI: 10.1249/jes.0000000000000017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Tetanic electrical stimulation releases adenosine triphosphate (ATP) from muscle fibers through pannexin-1 channels in a frequency-dependent manner; extracellular ATP activates signals that ultimately regulate gene expression and is able to increase glucose transport through activation of P2Y receptors, phosphatidylinositol 3-kinase, Akt, and AS160. We hypothesize that this mechanism is an important link between exercise and the regulation of muscle fiber plasticity and metabolism.
Collapse
Affiliation(s)
- Mariana Casas
- 1Center for Molecular Studies of the Cell, Biomedical Sciences Institute, Faculty of Medicine, Universidad de Chile; and 2Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | | | | |
Collapse
|
20
|
Shoji KF, Sáez PJ, Harcha PA, Aguila HL, Sáez JC. Pannexin1 channels act downstream of P2X 7 receptors in ATP-induced murine T-cell death. Channels (Austin) 2014; 8:142-56. [PMID: 24590064 DOI: 10.4161/chan.28122] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Death of murine T cells induced by extracellular ATP is mainly triggered by activation of purinergic P2X 7 receptors (P2X 7Rs). However, a link between P2X 7Rs and pannexin1 (Panx1) channels, which are non-selective, has been recently demonstrated in other cell types. In this work, we characterized the expression and cellular distribution of pannexin family members (Panxs 1, 2 and 3) in isolated T cells. Panx1 was the main pannexin family member clearly detected in both helper (CD4+) and cytotoxic (CD8+) T cells, whereas low levels of Panx2 were found in both T-cell subsets. Using pharmacological and genetic approaches, Panx1 channels were found to mediate most ATP-induced ethidium uptake since this was drastically reduced by Panx1 channel blockers (10Panx1, Probenecid and low carbenoxolone concentration) and absent in T cells derived from Panx1-/- mice. Moreover, electrophysiological measurements in wild-type CD4+ cells treated with ATP unitary current events and pharmacological sensitivity compatible with Panx1 channels were found. In addition, ATP release from T cells treated with 4Br-A23187, a calcium ionophore, was completely blocked with inhibitors of both connexin hemichannels and Panx1 channels. Panx1 channel blockers drastically reduced the ATP-induced T-cell mortality, indicating that Panx1 channels mediate the ATP-induced T-cell death. However, mortality was not reduced in T cells of Panx1-/- mice, in which levels of P2X 7Rs and ATP-induced intracellular free Ca2+ responses were enhanced suggesting that P2X 7Rs take over Panx1 channels lose-function in mediating the onset of cell death induced by extracellular ATP.
Collapse
Affiliation(s)
- Kenji F Shoji
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile
| | - Pablo J Sáez
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile
| | - Paloma A Harcha
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile
| | - Hector L Aguila
- Department of Immunology; University of Connecticut Health Center; Farmington, CT USA
| | - Juan C Sáez
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile; Instituto Milenio; Centro Interdisciplinario de Neurociencias de Valparaíso; Valparaíso, Chile
| |
Collapse
|
21
|
Retamal MA. Connexin and Pannexin hemichannels are regulated by redox potential. Front Physiol 2014; 5:80. [PMID: 24611056 PMCID: PMC3933782 DOI: 10.3389/fphys.2014.00080] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 02/08/2014] [Indexed: 01/28/2023] Open
Abstract
Connexins (Cxs) and Pannexins (Panxs) are two non-related protein families, having both the property to form hemichannels at the plasma membrane. There are 21 genes coding for different Cx based proteins and only 3 for Panx. Under physiological conditions, these hemichannels (Cxs and Panxs) present a low open probability, but when open, they allow the release of signaling molecules to the extracellular space. However, under pathological conditions, these hemichannels increase their open probability, inducing important lysis of metabolites, and ionic imbalance, which in turn induce the massive entry of Ca+2 to the cell. Actually, it is well recognized that Cxs and Panxs based channels play an important role in several diseases and -in many cases- this is associated with an aberrant hemichannel opening. Hemichannel opening and closing are controlled by a plethora of signaling including changes of the voltage plasma membrane, protein-protein interactions, and several posttranslational modifications, including protein cleavage, phosphorylation, glycosylation, hydroxylation and S-nitrosylation, among others. In particular, it has been recently shown that the cellular redox status modulates the opening/closing and permeability of at least Cx43, Cx46, and Panx1 hemichannels. Thus, for example, the gaseous transmitter nitric oxide (NO) can induce the S-nitrosylation of these proteins modulating in turn several of their properties. The reason is that the redox status of a cell is fundamental to set their response to the environment and also plays an important role in several pathologies. In this review, I will discuss how NO and other molecules associated with redox signaling modulate Cxs and Panx hemichannels properties.
Collapse
Affiliation(s)
- Mauricio A Retamal
- Facultad de Medicina Clínica Alemana, Centro de Fisiología Celular e Integrativa, Universidad del Desarrollo Santiago, Chile
| |
Collapse
|
22
|
Makarenkova HP, Shestopalov VI. The role of pannexin hemichannels in inflammation and regeneration. Front Physiol 2014; 5:63. [PMID: 24616702 PMCID: PMC3933922 DOI: 10.3389/fphys.2014.00063] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 02/02/2014] [Indexed: 12/19/2022] Open
Abstract
Tissue injury involves coordinated systemic responses including inflammatory response, targeted cell migration, cell-cell communication, stem cell activation and proliferation, and tissue inflammation and regeneration. The inflammatory response is an important prerequisite for regeneration. Multiple studies suggest that extensive cell-cell communication during tissue regeneration is coordinated by purinergic signaling via extracellular adenosine triphosphate (ATP). Most recent data indicates that ATP release for such communication is mediated by hemichannels formed by connexins and pannexins. The Pannexin family consists of three vertebrate proteins (Panx 1, 2, and 3) that have low sequence homology with other gap junction proteins and were shown to form predominantly non-junctional plasma membrane hemichannels. Pannexin-1 (Panx1) channels function as an integral component of the P2X/P2Y purinergic signaling pathway and is arguably the major contributor to pathophysiological ATP release. Panx1 is expressed in many tissues, with highest levels detected in developing brain, retina and skeletal muscles. Panx1 channel expression and activity is reported to increase significantly following injury/inflammation and during regeneration and differentiation. Recent studies also report that pharmacological blockade of the Panx1 channel or genetic ablation of the Panx1 gene cause significant disruption of progenitor cell migration, proliferation, and tissue regeneration. These findings suggest that pannexins play important roles in activation of both post-injury inflammatory response and the subsequent process of tissue regeneration. Due to wide expression in multiple tissues and involvement in diverse signaling pathways, pannexins and connexins are currently being considered as therapeutic targets for traumatic brain or spinal cord injuries, ischemic stroke and cancer. The precise role of pannexins and connexins in the balance between tissue inflammation and regeneration needs to be further understood.
Collapse
Affiliation(s)
- Helen P Makarenkova
- Department of Cell and Molecular Biology, The Scripps Research Institute La Jolla, CA, USA
| | - Valery I Shestopalov
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine Miami, FL, USA ; Department of Cell Biology and Anatomy, Vavilov Institute for General Genetics Moscow, Russia
| |
Collapse
|
23
|
Abstract
The pannexins (Panxs) are a family of chordate proteins homologous to the invertebrate gap junction forming proteins named innexins. Three distinct Panx paralogs (Panx1, Panx2, and Panx3) are shared among the major vertebrate phyla, but they appear to have suppressed (or even lost) their ability to directly couple adjacent cells. Connecting the intracellular and extracellular compartments is now widely accepted as Panx's primary function, facilitating the passive movement of ions and small molecules along electrochemical gradients. The tissue distribution of the Panxs ranges from pervasive to very restricted, depending on the paralog, and are often cell type-specific and/or developmentally regulated within any given tissue. In recent years, Panxs have been implicated in an assortment of physiological and pathophysiological processes, particularly with respect to ATP signaling and inflammation, and they are now considered to be a major player in extracellular purinergic communication. The following is a comprehensive review of the Panx literature, exploring the historical events leading up to their discovery, outlining our current understanding of their biochemistry, and describing the importance of these proteins in health and disease.
Collapse
Affiliation(s)
- Stephen R Bond
- Genome Technology Branch, Division of Intramural Research, National Human Genome Research Institute, National Institutes of Health Bethesda, MD, USA ; Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia Vancouver, BC, Canada
| | - Christian C Naus
- Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia Vancouver, BC, Canada
| |
Collapse
|
24
|
Shestopalov VI, Slepak VZ. Molecular pathways of pannexin1-mediated neurotoxicity. Front Physiol 2014; 5:23. [PMID: 24575045 PMCID: PMC3920106 DOI: 10.3389/fphys.2014.00023] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/10/2014] [Indexed: 01/09/2023] Open
Abstract
Pannexin1 (Panx1) forms non-selective membrane channels, structurally similar to gap junction hemichannels, and are permeable to ions, nucleotides, and other small molecules below 900 Da. Panx1 activity has been implicated in paracrine signaling and inflammasome regulation. Recent studies in different animal models showed that overactivation of Panx1 correlates with a selective demise of several types of neurons, including retinal ganglion cells, brain pyramidal, and enteric neurons. The list of Panx1 activators includes extracellular ATP, glutamate, high K(+), Zn(2+), fibroblast growth factors (FGFs),pro-inflammatory cytokines, and elevation of intracellular Ca(2+). Most of these molecules are released following mechanical, ischemic, or inflammatory injury of the CNS, and rapidly activate the Panx1 channel. Prolonged opening of Panx1 channel induced by these "danger signals" triggers a cascade of neurotoxic events capable of killing cells. The most vulnerable cell type are neurons that express high levels of Panx1. Experimental evidence suggests that Panx1 channels mediate at least two distinct neurotoxic processes: increased permeability of the plasma membrane and activation of the inflammasome in neurons and glia. Importantly, both pharmacological and genetic inactivation of Panx1 suppresses both these processes, providing a marked protection in several disease and injury models. These findings indicate that external danger signals generated after diverse types of injuries converge to activate Panx1. In this review we discuss molecular mechanisms associated with Panx1 toxicity and the crosstalk between different pathways.
Collapse
Affiliation(s)
- Valery I Shestopalov
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine Miami, FL, USA ; Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine Miami, FL, USA ; Vavilov Institute of General Genetics, Moscow, Russian Federation, University of Miami Miller School of Medicine Miami, FL, USA
| | - Vladlen Z Slepak
- Department of Molecular Pharmacology, University of Miami Miller School of Medicine Miami, FL, USA ; Neuroscience Program, University of Miami Miller School of Medicine Miami, FL, USA
| |
Collapse
|
25
|
Wicki-Stordeur LE, Swayne LA. The emerging Pannexin 1 signalome: a new nexus revealed? Front Cell Neurosci 2014; 7:287. [PMID: 24409119 PMCID: PMC3884145 DOI: 10.3389/fncel.2013.00287] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 12/23/2013] [Indexed: 11/22/2022] Open
Abstract
Pannexins (Panxs) are a family of single-membrane, large-pore ion, and metabolite permeable channels. Of the three Panx proteins, Panx1 has been most extensively studied, and has recently emerged as an exciting, clinically relevant target in many physiological and pathophysiological settings. This channel is widely expressed across various cell and tissue types; however its links to precise signaling pathways are largely unknown. Here we review the current literature surrounding presently identified Panx1–protein interactions, a critical first step to unraveling the Panx1 signalome. First we elucidate the reported associations of Panx1 with other ion channels, receptors, and channel signaling complexes. Further, we highlight recently identified Panx1–cytoskeleton interactions. Finally, we discuss the implications of these protein–protein interactions for Panx1 function in various cell and tissue types, and identify key outstanding questions arising from this work.
Collapse
Affiliation(s)
| | - Leigh A Swayne
- Division of Medical Sciences, University of Victoria Victoria, BC, Canada ; Department of Biology, University of Victoria Victoria, BC, Canada ; Department of Biochemistry and Microbiology, University of Victoria Victoria, BC, Canada ; Department of Cellular and Physiological Sciences and Island Medical Program, University of British Columbia Vancouver, BC, Canada
| |
Collapse
|
26
|
Boyce AKJ, Prager RT, Wicki-Stordeur LE, Swayne LA. Pore positioning: current concepts in Pannexin channel trafficking. Channels (Austin) 2013; 8:110-7. [PMID: 24300303 DOI: 10.4161/chan.27287] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pannexins (Panxs) are a multifaceted family of ion and metabolite channels that play key roles in a number of physiological and pathophysiological settings. These single membrane large-pore channels exhibit a variety of tissue, cell type, and subcellular distributions. The lifecycles of Panxs are complex, yet must be understood to accurately target these proteins for future therapeutic use. Here we review the basics of Panx function and localization, and then analyze the recent advances in knowledge regarding Panx trafficking. We examine several intrinsic features of Panxs including specific post-translational modifications, the divergent C-termini, and oligomerization, all of which contribute to Panx anterograde transport pathways. Further, we examine the potential influence of extrinsic factors, such as protein-protein interactions, on Panx trafficking. Finally, we highlight what is currently known with respect to Panx internalization and retrograde transport, and present new data illustrating Panx1 internalization following an activating stimulus.
Collapse
Affiliation(s)
- Andrew K J Boyce
- Division of Medical Sciences; Island Medical Program; University of Victoria; Victoria, British Columbia, Canada
| | - Ross T Prager
- Division of Medical Sciences; Island Medical Program; University of Victoria; Victoria, British Columbia, Canada
| | - Leigh E Wicki-Stordeur
- Division of Medical Sciences; Island Medical Program; University of Victoria; Victoria, British Columbia, Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences; Island Medical Program; University of Victoria; Victoria, British Columbia, Canada; Department of Biology; University of Victoria; Victoria, British Columbia, Canada; Department of Biochemistry and Microbiology; University of Victoria; Victoria, British Columbia, Canada; Department of Cellular and Physiological Sciences; University of British Columbia; Vancouver, British Columbia, Canada
| |
Collapse
|
27
|
Križaj D, Ryskamp DA, Tian N, Tezel G, Mitchell CH, Slepak VZ, Shestopalov VI. From mechanosensitivity to inflammatory responses: new players in the pathology of glaucoma. Curr Eye Res 2013; 39:105-19. [PMID: 24144321 DOI: 10.3109/02713683.2013.836541] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF THE STUDY Many blinding diseases of the inner retina are associated with degeneration and loss of retinal ganglion cells (RGCs). Recent evidence implicates several new signaling mechanisms as causal agents associated with RGC injury and remodeling of the optic nerve head. Ion channels such as Transient receptor potential vanilloid isoform 4 (TRPV4), pannexin-1 (Panx1) and P2X7 receptor are localized to RGCs and act as potential sensors and effectors of mechanical strain, ischemia and inflammatory responses. Under normal conditions, TRPV4 may function as an osmosensor and a polymodal molecular integrator of diverse mechanical and chemical stimuli, whereas P2X7R and Panx1 respond to stretch- and/or swelling-induced adenosine triphosphate release from neurons and glia. Ca(2+) influx, induced by stimulation of mechanosensitive ion channels in glaucoma, is proposed to influence dendritic and axonal remodeling that may lead to RGC death while (at least initially) sparing other classes of retinal neuron. The secondary phase of the retinal glaucoma response is associated with microglial activation and an inflammatory response involving Toll-like receptors (TLRs), cluster of differentiation 3 (CD3) immune recognition molecules associated with the T-cell antigen receptor, complement molecules and cell type-specific release of neuroactive cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). The retinal response to mechanical stress thus involves a diversity of signaling pathways that sense and transduce mechanical strain and orchestrate both protective and destructive secondary responses. CONCLUSIONS Mechanistic understanding of the interaction between pressure-dependent and independent pathways is only beginning to emerge. This review focuses on the molecular basis of mechanical strain transduction as a primary mechanism that can damage RGCs. The damage occurs through Ca(2+)-dependent cellular remodeling and is associated with parallel activation of secondary ischemic and inflammatory signaling pathways. Molecules that mediate these mechanosensory and immune responses represent plausible targets for protecting ganglion cells in glaucoma, optic neuritis and retinal ischemia.
Collapse
|
28
|
Dahl G, Qiu F, Wang J. The bizarre pharmacology of the ATP release channel pannexin1. Neuropharmacology 2013; 75:583-93. [PMID: 23499662 DOI: 10.1016/j.neuropharm.2013.02.019] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 02/08/2013] [Accepted: 02/26/2013] [Indexed: 12/29/2022]
Abstract
Pannexins were originally thought to represent a second and redundant family of gap junction proteins in addition to the well characterized connexins. However, it is now evident that pannexins function as unapposed membrane channels and the major role of Panx1 is that of an ATP release channel. Despite the contrasting functional roles, connexins, innexins and pannexins share pharmacological properties. Most gap junction blockers also attenuate the function of Panx1, including carbenoxolone, mefloquine and flufenamic acid. However, in contrast to connexin based gap junction channels, Panx1 channel activity can be attenuated by several groups of drugs hitherto considered very specific for other proteins. The drugs affecting Panx1 channels include several transport inhibitors, chloride channel blockers, mitochondrial inhibitors, P2X7 receptor ligands, inflammasome inhibitors and malaria drugs. These observations indicate that Panx1 may play an extended role in a wider spectrum of physiological functions. Alternatively, Panx1 may share structural domains with other proteins, not readily revealed by sequence alignments. This article is part of the Special Issue Section entitled 'Current Pharmacology of Gap Junction Channels and Hemichannels'.
Collapse
Affiliation(s)
- Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami, School of Medicine, PO Box 016430, Miami, FL 33101, USA.
| | - Feng Qiu
- Department of Physiology and Biophysics, University of Miami, School of Medicine, PO Box 016430, Miami, FL 33101, USA
| | - Junjie Wang
- Department of Physiology and Biophysics, University of Miami, School of Medicine, PO Box 016430, Miami, FL 33101, USA
| |
Collapse
|
29
|
Gap junction channels and hemichannels in the CNS: regulation by signaling molecules. Neuropharmacology 2013; 75:567-82. [PMID: 23499663 DOI: 10.1016/j.neuropharm.2013.02.020] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 02/08/2013] [Accepted: 02/26/2013] [Indexed: 11/20/2022]
Abstract
Coordinated interaction among cells is critical to develop the extremely complex and dynamic tasks performed by the central nervous system (CNS). Cell synchronization is in part mediated by connexins and pannexins; two different protein families that form gap junction channels and hemichannels. Whereas gap junction channels connect the cytoplasm of contacting cells and coordinate electric and metabolic activities, hemichannels communicate intra- and extra-cellular compartments and serve as diffusional pathways for ions and small molecules. Cells in the CNS depend on paracrine/autocrine communication via several extracellular signaling molecules, such as, cytokines, growth factors, transmitters and free radical species to sense changes in microenvironment as well as to adapt to them. These signaling molecules modulate crucial processes of the CNS, including, cellular migration and differentiation, synaptic transmission and plasticity, glial activation, cell viability and microvascular blood flow. Gap junction channels and hemichannels are affected by different signaling transduction pathways triggered by these paracrine/autocrine signaling molecules. Most of the modulatory effects induced by these signaling molecules are specific to the cell type and the connexin and pannexin subtype expressed in different brain areas. In this review, we summarized and discussed most of the relevant and recently published information on the effects of signaling molecules on connexin or pannexin based channels and their possible relevance in CNS physiology and pathology. This article is part of the Special Issue Section entitled 'Current Pharmacology of Gap Junction Channels and Hemichannels'.
Collapse
|
30
|
Dahl G, Keane RW. Pannexin: from discovery to bedside in 11±4 years? Brain Res 2012; 1487:150-9. [PMID: 22771709 PMCID: PMC3590907 DOI: 10.1016/j.brainres.2012.04.058] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 03/29/2012] [Accepted: 04/11/2012] [Indexed: 12/22/2022]
Abstract
Pannexin1 (Panx1) originally was discovered as a gap junction related protein. However, rather than forming the cell-to-cell channels of gap junctions, Panx1 forms a mechanosensitive and highly ATP permeable channel in the cell membrane allowing the exchange of molecules between the cytoplasm and the extracellular space. The list of arguments for Panx1 representing the major ATP release channel includes: (1) Panx1 is expressed in (all?) cells releasing ATP in a non-vesicular fashion, such as erythrocytes; (2) in cells with polar release of ATP, Panx1 is expressed at the ATP release site, such as the apical membrane in airway epithelial cells; (3) the pharmacology of Panx1 channels matches that of ATP release; (4) mutation of Panx1 in strategic positions in the protein modifies ATP release; and (5) knockdown or knockout of Panx1 attenuates or abolishes ATP release. Panx1, in association with the purinergic receptor P2X7, is involved in the innate immune response and in apoptotic/pyroptotic cell death. Inflammatory processes are responsible for amplification of the primary lesion in CNS trauma and stroke. Panx1, as an early signal event and as a signal amplifier in these processes, is an obvious target for the prevention of secondary cell death due to inflammasome activity. Since Panx1 inhibitors such as probenecid are already clinically tested in different settings they should be considered for therapy in stroke and CNS trauma.
Collapse
Affiliation(s)
- Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami, School of Medicine, PO Box 016430, Miami, FL 33101, USA.
| | | |
Collapse
|
31
|
Xiao F, Waldrop SL, Khimji AK, Kilic G. Pannexin1 contributes to pathophysiological ATP release in lipoapoptosis induced by saturated free fatty acids in liver cells. Am J Physiol Cell Physiol 2012; 303:C1034-44. [PMID: 22972801 DOI: 10.1152/ajpcell.00175.2012] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hepatocyte lipoapoptosis induced by saturated free fatty acids (FFA) contributes to hepatic inflammation in lipotoxic liver injury, and the cellular mechanisms involved have not been defined. Recent studies have shown that apoptosis in nonhepatic cells stimulates ATP release via activation of pannexin1 (panx1), and extracellular ATP functions as a proinflammatory signal for recruitment and activation of the inflammatory cells. However, it is not known whether lipoapoptosis stimulates ATP release in liver cells. We found that lipoapoptosis induced by saturated FFA stimulated ATP release in liver cells that increased extracellular ATP concentration by more than fivefold above the values observed in healthy cells. This sustained pathophysiological ATP release was not dependent on caspase-3/7 activation. Inhibition of c-Jun NH(2)-terminal kinase (JNK), a key mediator of lipoapoptosis, with SP600125 blocked pathophysiological ATP release in a dose-dependent manner. RT-PCR analysis indicated that panx1 is expressed in hepatocytes and multiple liver cell lines. Notably, inhibition of panx1 expression with short hairpin (sh)RNA inhibited in part pathophysiological ATP release. Moreover, lipoapoptosis stimulated uptake of a membrane impermeable dye YoPro-1 (indicative of panx1 activation), which was inhibited by panx1 shRNA, probenecid, and mefloquine. These results suggest that panx1 contributes to pathophysiological ATP release in lipoapoptosis induced by saturated FFA. Thus panx1 may play an important role in hepatic inflammation by mediating an increase in extracellular ATP concentration in lipotoxic liver injury.
Collapse
Affiliation(s)
- Feng Xiao
- Deptartment of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9151, USA
| | | | | | | |
Collapse
|
32
|
Romanov RA, Bystrova MF, Rogachevskaya OA, Sadovnikov VB, Shestopalov VI, Kolesnikov SS. The ATP permeability of pannexin 1 channels in a heterologous system and in mammalian taste cells is dispensable. J Cell Sci 2012; 125:5514-23. [PMID: 22956545 DOI: 10.1242/jcs.111062] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Afferent output in type II taste cells is mediated by ATP liberated through ion channels. It is widely accepted that pannexin 1 (Panx1) channels are responsible for ATP release in diverse cell types, including taste cells. While biophysical evidence implicates slow deactivation of ion channels following ATP release in taste cells, recombinant Panx1 activates and deactivates rapidly. This inconsistency could indicate that the cellular context specifies Panx1 functioning. We cloned Panx1 from murine taste tissue, and heterologously expressed it in three different cell lines: HEK-293, CHO and neuroblastoma SK-N-SH cells. In all three cell lines, Panx1 transfection yielded outwardly rectifying anion channels that exhibited fast gating and negligible permeability to anions exceeding 250 Da. Despite expression of Panx1, the host cells did not liberate ATP upon stimulation, making it unclear whether Panx1 is involved in taste-related ATP secretion. This issue was addressed using mice with genetic ablation of the Panx1 gene. The ATP-biosensor assay revealed that, in taste cells devoid of Panx1, ATP secretion was robust and apparently unchanged compared with the control. Our data suggest that Panx1 alone forms a channel that has insufficient permeability to ATP. Perhaps, a distinct subunit and/or a regulatory circuit that is absent in taste cells is required to enable a high ATP-permeability mode of a native Panx1-based channel.
Collapse
Affiliation(s)
- Roman A Romanov
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutional Street 3, Pushchino, Moscow Region, 142290, Russia
| | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Stomatin inhibits pannexin-1-mediated whole-cell currents by interacting with its carboxyl terminal. PLoS One 2012; 7:e39489. [PMID: 22768083 PMCID: PMC3387187 DOI: 10.1371/journal.pone.0039489] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 05/21/2012] [Indexed: 12/20/2022] Open
Abstract
The pannexin-1 (Panx1) channel (often referred to as the Panx1 hemichannel) is a large-conductance channel in the plasma membrane of many mammalian cells. While opening of the channel is potentially detrimental to the cell, little is known about how it is regulated under physiological conditions. Here we show that stomatin inhibited Panx1 channel activity. In transfected HEK-293 cells, stomatin reduced Panx1-mediated whole-cell currents without altering either the total or membrane surface Panx1 protein expression. Stomatin coimmunoprecipitated with full-length Panx1 as well as a Panx1 fragment containing the fourth membrane-spanning domain and the cytosolic carboxyl terminal. The inhibitory effect of stomatin on Panx1-mediated whole-cell currents was abolished by truncating Panx1 at a site in the cytosolic carboxyl terminal. In primary culture of mouse astrocytes, inhibition of endogenous stomatin expression by small interfering RNA enhanced Panx1-mediated outward whole-cell currents. These observations suggest that stomatin may play important roles in astrocytes and other cells by interacting with Panx1 carboxyl terminal to limit channel opening.
Collapse
|
35
|
Marchant JS, Lin-Moshier Y, Walseth TF, Patel S. The Molecular Basis for Ca 2+ Signalling by NAADP: Two-Pore Channels in a Complex? ACTA ACUST UNITED AC 2012; 1:63-76. [PMID: 25309835 DOI: 10.1166/msr.2012.1003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
NAADP is a potent Ca2+ mobilizing messenger in a variety of cells but its molecular mechanism of action is incompletely understood. Accumulating evidence indicates that the poorly characterized two-pore channels (TPCs) in animals are NAADP sensitive Ca2+-permeable channels. TPCs localize to the endo-lysosomal system but are functionally coupled to the better characterized endoplasmic reticulum Ca2+ channels to generate physiologically relevant complex Ca2+ signals. Whether TPCs directly bind NAADP is not clear. Here we discuss the idea based on recent studies that TPCs are the pore-forming subunits of a protein complex that includes tightly associated, low molecular weight NAADP-binding proteins.
Collapse
Affiliation(s)
- Jonathan S Marchant
- Department of Pharmacology, University of Minnesota Medical School, MN 55455, USA
| | - Yaping Lin-Moshier
- Department of Pharmacology, University of Minnesota Medical School, MN 55455, USA
| | - Timothy F Walseth
- Department of Pharmacology, University of Minnesota Medical School, MN 55455, USA
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, WC1E 6BT, UK
| |
Collapse
|
36
|
Lazarowski ER. Vesicular and conductive mechanisms of nucleotide release. Purinergic Signal 2012; 8:359-73. [PMID: 22528679 DOI: 10.1007/s11302-012-9304-9] [Citation(s) in RCA: 234] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 01/21/2012] [Indexed: 12/22/2022] Open
Abstract
Extracellular nucleotides and nucleosides promote a vast range of physiological responses, via activation of cell surface purinergic receptors. Virtually all tissues and cell types exhibit regulated release of ATP, which, in many cases, is accompanied by the release of uridine nucleotides. Given the relevance of extracellular nucleotide/nucleoside-evoked responses, understanding how ATP and other nucleotides are released from cells is an important physiological question. By facilitating the entry of cytosolic nucleotides into the secretory pathway, recently identified vesicular nucleotide and nucleotide-sugar transporters contribute to the exocytotic release of ATP and UDP-sugars not only from endocrine/exocrine tissues, but also from cell types in which secretory granules have not been biochemically characterized. In addition, plasma membrane connexin hemichannels, pannexin channels, and less-well molecularly defined ATP conducting anion channels have been shown to contribute to the release of ATP (and UTP) under a variety of conditions.
Collapse
Affiliation(s)
- Eduardo R Lazarowski
- School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7248, USA.
| |
Collapse
|
37
|
The biochemistry and function of pannexin channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:15-22. [PMID: 22305965 DOI: 10.1016/j.bbamem.2012.01.017] [Citation(s) in RCA: 299] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 01/05/2012] [Accepted: 01/19/2012] [Indexed: 11/21/2022]
Abstract
Three family members compose the pannexin family of channel-forming glycoproteins (Panx1, Panx2 and Panx3). Their primary function is defined by their capacity to form single-membrane channels that are regulated by post-translational modifications, channel intermixing, and sub-cellular expression profiles. Panx1 is ubiquitously expressed in many mammalian tissues, while Panx2 and Panx3 appear to be more restricted in their expression. Paracrine functions of Panx1 as an ATP release channel have been extensively studied and this channel plays a key role, among others, in the release of "find-me" signals for apoptotic cell clearance. In addition Panx1 has been linked to propagation of calcium waves, regulation of vascular tone, mucociliary lung clearance, taste-bud function and has been shown to act like a tumor suppressor in gliomas. Panx1 channel opening can also be detrimental, contributing to cell death and seizures under ischemic or epileptic conditions and even facilitating HIV-1 viral infection. Panx2 is involved in differentiation of neurons while Panx3 plays a role in the differentiation of chondrocytes, osteoblasts and the maturation and transport of sperm. Using the available Panx1 knockout mouse models it has now become possible to explore some of its physiological functions. However, given the potential for one pannexin to compensate for another it seems imperative to generate single and double knockout mouse models involving all three pannexins and evaluate their interplay in normal differentiation and development as well as in malignant transformation and disease. This article is part of a Special Issue entitled: The communicating junctions, roles and dysfunctions.
Collapse
|
38
|
Baroja-Mazo A, Barberà-Cremades M, Pelegrín P. The participation of plasma membrane hemichannels to purinergic signaling. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:79-93. [PMID: 22266266 DOI: 10.1016/j.bbamem.2012.01.002] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 12/30/2011] [Accepted: 01/04/2012] [Indexed: 12/17/2022]
Abstract
The field of hemichannels is closely related to the purinergic signaling and both areas have been growing in parallel. Hemichannels open in response to a wide range of stressful conditions, such as ischemia, pressure or swelling. Hemichannels represent an important mechanism for the cellular release of adenosine 5'-triphosphate (ATP), which is an agonist of the P2Y and P2X family of purinergic receptors. Therefore, hemichannels are key molecules in the regulation of purinergic receptor activation, during physiological and pathophysiological conditions. Furthermore, purinergic receptor activation can also lead to the opening of hemichannels and the subsequent amplification of purinergic signaling via a positive signaling feedback loop, giving rise to the concept of ATP-induced ATP release. Purinergic receptor signaling is involved in regulating many physiological and pathophysiological processes. P2Y receptors activate inositol trisphosphate and transiently increase intracellular calcium. This signaling opens both connexin and pannexin channels, therefore contributing to the expansion of calcium waves across astrocytes and epithelial cells. In addition, several of the P2X receptor subtypes, including the P2X2, P2X4 and P2X7 receptors, activate select cellular permeation pathways to large molecules, including the pannexin-1 channels, which are involved in the initiation of inflammatory responses and cell death. Consequently, the interplay between purinergic receptors and hemichannels could represent a novel target with substantial therapeutic implications in areas such as chronic pain, inflammation or atherosclerosis. This article is part of a Special Issue entitled: The communicating junctions, roles and dysfunctions.
Collapse
Affiliation(s)
- Alberto Baroja-Mazo
- University Hospital Virgen de la Arrixaca, Fundación Formación Investigación Sanitaria Región Murcia, Murcia, Spain
| | | | | |
Collapse
|
39
|
LI ANG, LEUNG CHITING, PETERSON-YANTORNO KIM, STAMER WDANIEL, MITCHELL CLAIREH, CIVAN MORTIMERM. Mechanisms of ATP release by human trabecular meshwork cells, the enabling step in purinergic regulation of aqueous humor outflow. J Cell Physiol 2012; 227:172-82. [PMID: 21381023 PMCID: PMC3117029 DOI: 10.1002/jcp.22715] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Our guiding hypothesis is that ecto-enzymatic conversion of extracellular ATP to adenosine activates A(1) adenosine receptors, reducing resistance to aqueous humor outflow and intraocular pressure. The initial step in this purinergic regulation is ATP release from outflow-pathway cells by mechanisms unknown. We measured similar ATP release from human explant-derived primary trabecular meshwork (TM) cells (HTM) and a human TM cell line (TM5). Responses to 21 inhibitors indicated that pannexin-1 (PX1) and connexin (Cx) hemichannels and P2X(7) receptors (P2RX(7) ) were comparably important in modulating ATP release induced by hypotonic swelling, whereas vesicular release was insignificant. Consistent with prior studies of PX1 activity in certain other cells, ATP release was lowered by the reducing agent dithiothreitol. Overexpressing PX1 in HEK293T cells promoted, while partial knockdown (KD) in both HEK293T and TM5 cells inhibited hypotonicity-activated ATP release. Additionally, KD reduced the pharmacologically defined contribution of PX1 and enhanced those of Cx and P2RX(7) . ATP release was also triggered by raising intracellular Ca(2+) activity with ionomycin after a prolonged lag time and was unaffected by the PX1 blocker probenecid, but nearly abolished by P2RX(7) antagonists. We conclude that swelling-stimulated ATP release from human TM cells is physiologically mediated by PX1 and Cx hemichannels and P2X(7) receptors, but not by vesicular release. PX1 appears not to be stimulated by intracellular Ca(2+) in TM cells, but can be modulated by oxidation-reduction state. The P2RX(7) -dependent component of swelling-activated release may be mediated by PX1 hemichannels or reflect apoptotic magnification of ATP release, either through itself and/or hemichannels.
Collapse
Affiliation(s)
- ANG LI
- Department of Physiology, School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - CHI TING LEUNG
- Department of Physiology, School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - KIM PETERSON-YANTORNO
- Department of Physiology, School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - W. DANIEL STAMER
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ
| | - CLAIRE H. MITCHELL
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
| | - MORTIMER M. CIVAN
- Department of Physiology, School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
40
|
Li A, Banerjee J, Leung CT, Peterson-Yantorno K, Stamer WD, Civan MM. Mechanisms of ATP release, the enabling step in purinergic dynamics. Cell Physiol Biochem 2011; 28:1135-44. [PMID: 22179002 DOI: 10.1159/000335865] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2011] [Indexed: 11/19/2022] Open
Abstract
The only effective intervention to slow onset and progression of glaucomatous blindness is to lower intraocular pressure (IOP). Among other modulators, adenosine receptors (ARs) exert complex regulation of IOP. Agonists of A(3)ARs in the ciliary epithelium activate Cl(-) channels, favoring increased formation of aqueous humor and elevated IOP. In contrast, stimulating A(1)ARs in the trabecular outflow pathway enhances release of matrix metalloproteinases (MMPs) from trabecular meshwork (TM) cells, reducing resistance to outflow of aqueous humor to lower IOP. These opposing actions are thought to be initiated by cellular release of ATP and its ectoenzymatic conversion to adenosine. This view is now supported by our identification of six ectoATPases in trabecular meshwork (TM) cells and by our observation that external ATP enhances TM-cell secretion of MMPs through ectoenzymatic formation of adenosine. ATP release is enhanced by cell swelling and stretch. Also, enhanced ATP release and downstream MMP secretion is one mediator of the action of actin depolymerization to reduce outflow resistance. Inflow and outflow cells share pannexin-1 and connexin hemichannel pathways for ATP release. However, vesicular release and P2X(7) release pathways were functionally limited to inflow and outflow cells, respectively, suggesting that blocking exocytosis might selectively inhibit inflow, lowering IOP.
Collapse
Affiliation(s)
- Ang Li
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6085, USA
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Pannexin 1 (Px1, Panx1) and pannexin 2 (Px2, Panx2) form large-pore nonselective channels in the plasma membrane of cells and were suggested to play a role in the pathophysiology of cerebral ischemia. To directly test a potential contribution of pannexins in ischemia-related mechanisms, we performed experiments in Px1(-/-), Px2(-/-), and Px1(-/-)Px2(-/-) knockout mice. IL-1β release, channel function in astrocytes, and cortical spreading depolarization were not altered in Px1(-/-)Px2(-/-) mice, indicating that, in contrast to previous concepts, these processes occur normally in the absence of pannexin channels. However, ischemia-induced dye release from cortical neurons was lower, indicating that channel function in Px1(-/-)Px2(-/-) neurons was impaired. Furthermore, Px1(-/-)Px2(-/-) mice had a better functional outcome and smaller infarcts than wild-type mice when subjected to ischemic stroke. In conclusion, our data demonstrate that Px1 and Px2 underlie channel function in neurons and contribute to ischemic brain damage.
Collapse
|
42
|
Bunse S, Schmidt M, Hoffmann S, Engelhardt K, Zoidl G, Dermietzel R. Single cysteines in the extracellular and transmembrane regions modulate pannexin 1 channel function. J Membr Biol 2011; 244:21-33. [PMID: 21938521 DOI: 10.1007/s00232-011-9393-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 08/26/2011] [Indexed: 12/01/2022]
Abstract
Pannexins form high-conductance ion channels in the membranes of many vertebrate cells. Functionally, they have been associated with multiple functional pathways like the propagation of calcium waves, ATP release, responses to ischemic conditions and apoptosis. In contrast to accumulating details which uncovered their functions, the molecular mechanisms for pannexin channel regulation and activation are hardly understood. To further elucidate regulatory mechanisms, we substituted cysteine residues, expected key elements for channel function, in extracellular and transmembrane regions of Pannexin 1 (Panx1). Most apparently, substitution of the transmembrane cysteine C40 resulted in constitutively open channels with profoundly increased activity. Hence, Xenopus laevis oocytes injected with corresponding cRNA showed strongly impaired viability, anomalous dye uptake and greatly increased whole-cell conductivity. All changes induced by C40 substitution were significantly reduced by the Panx1 channel blocker carbenoxolone, indicating that channel activity of the mutated Panx1 had been affected. In contrast, no changes occurred after substitution of the two other transmembrane cysteines, C215 and C227, in terms of channel conductivity. Finally, substitution of any of the four extracellular cysteines resulted in complete loss of channel function in both X. laevis oocytes and transfected N2A cells. From this, we conclude that cysteine residues of Panx1 reveal differential functional profiles for channel activation and drug sensitivity.
Collapse
Affiliation(s)
- Stefanie Bunse
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, MA 6/159, 44780 Bochum, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Evidence for sustained ATP release from liver cells that is not mediated by vesicular exocytosis. Purinergic Signal 2011; 7:435-46. [PMID: 21630025 DOI: 10.1007/s11302-011-9240-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 05/23/2011] [Indexed: 10/18/2022] Open
Abstract
Extracellular ATP regulates many important cellular functions in the liver by stimulating purinergic receptors. Recent studies have shown that rapid exocytosis of ATP-enriched vesicles contributes to ATP release from liver cells. However, this rapid ATP release is transient, and ceases in ~30 s after the exposure to hypotonic solution. The purpose of these studies was to assess the role of vesicular exocytosis in sustained ATP release. An exposure to hypotonic solution evoked sustained ATP release that persisted for more than 15 min after the exposure. Using FM1-43 (N-(3-triethylammoniumpropyl)-4-(4-(dibutylamino)styryl)pyridinium dibromide) fluorescence to measure exocytosis, we found that hypotonic solution stimulated a transient increase in FM1-43 fluorescence that lasted ~2 min. Notably, the rate of FM1-43 fluorescence and the magnitude of ATP release were not correlated, indicating that vesicular exocytosis may not mediate sustained ATP release from liver cells. Interestingly, mefloquine potently inhibited sustained ATP release, but did not inhibit an increase in FM1-43 fluorescence evoked by hypotonic solution. Consistent with these findings, when exocytosis of ATP-enriched vesicles was specifically stimulated by 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB), mefloquine failed to inhibit ATP release evoked by NPPB. Thus, mefloquine can pharmacologically dissociate sustained ATP release and vesicular exocytosis. These results suggest that a distinct mefloquine-sensitive membrane ATP transport may contribute to sustained ATP release from liver cells. This novel mechanism of membrane ATP transport may play an important role in the regulation of purinergic signaling in liver cells.
Collapse
|
44
|
Gründken C, Hanske J, Wengel S, Reuter W, Abdulazim A, Shestopalov VI, Dermietzel R, Zoidl G, Prochnow N. Unified patch clamp protocol for the characterization of Pannexin 1 channels in isolated cells and acute brain slices. J Neurosci Methods 2011; 199:15-25. [PMID: 21549752 DOI: 10.1016/j.jneumeth.2011.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 04/12/2011] [Accepted: 04/13/2011] [Indexed: 10/18/2022]
Abstract
In the central nervous system, Pannexin 1 (Panx1) channels are implicated in a variety of physiological and pathological conditions. One of the prerequisites to enlighten the role of Panx1 is the development and standardization of reliable methods. Here, we address the applicability of voltage clamp protocols to identify Panx1 channel mediated currents in neurons of acutely dissected brain slices. We improved an established protocol and report on a modified paradigm that robustly evokes Panx1 channel currents. Crucial advances are the use of physiologic ion gradient conditions and a preconditioning step of depolarizing membrane potential ramps of long duration. This new paradigm provides significant impact on membrane current generation at hypo- and depolarized holding potential steps post voltage ramp preconditioning in heterologous expression systems and primary hippocampal CA1 neurons of mouse brain slices in vitro. Finally, we demonstrate that under these conditions the analysis of tail currents elicited by repolarization of the cells from preconditioning holding potential depolarization permits an independent method to isolate Panx1 mediated channel activity. In summary, this study provides a comprehensive methodological improvement in the biophysical analysis of Panx1 channels with a particular focus on investigations under physiological conditions in complex tissues.
Collapse
Affiliation(s)
- Christina Gründken
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Li A, Leung CT, Peterson-Yantorno K, Mitchell CH, Civan MM. Pathways for ATP release by bovine ciliary epithelial cells, the initial step in purinergic regulation of aqueous humor inflow. Am J Physiol Cell Physiol 2010; 299:C1308-17. [PMID: 20926783 DOI: 10.1152/ajpcell.00333.2010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
ATP release by nonpigmented (NPE) and pigmented (PE) ciliary epithelial cells is the enabling step in purinergic regulation of aqueous humor formation, but the release pathways are unknown. We measured ATP release from primary cultures of bovine mixed NPE and PE (bCE) cells and transformed bovine NPE and PE cells, using the luciferin-luciferase reaction. Hypotonicity-triggered bCE ATP release was inhibited by the relatively selective blocker of pannexin-1 (PX1) hemichannels (probenecid, 1 mM, 47 ± 2%), by a connexin inhibitor (heptanol, 1 mM, 49 ± 4%), and by an inhibitor of vesicular release (bafilomycin A1, 25 ± 2%), but not by the P2X(7) receptor (P2RX(7)) antagonist KN-62. Bafilomycin A1 acts by reducing the driving force for uptake of ATP from the cytosol into vesicles. The reducing agent dithiothreitol reduced probenecid-blockable ATP release. Similar results were obtained with NPE and PE cell lines. Pannexins PX1-3, connexins Cx43 and Cx40, and P2RX(7) were identified in native cells and cell lines by RT-PCR. PX1 mRNA expression was confirmed by Northern blots; its quantitative expression was comparable to that of Cx43 by real-time PCR. Heterologous expression of bovine PX1 in HEK293T cells enhanced swelling-activated ATP release, inhibitable by probenecid. We conclude that P2RX(7)-independent PX1 hemichannels, Cx hemichannels, and vesicular release contribute comparably to swelling-triggered ATP release. The relatively large response to dithiothreitol raises the possibility that the oxidation-reduction state is a substantial regulator of PX1-mediated ATP release from bovine ciliary epithelial cells.
Collapse
Affiliation(s)
- Ang Li
- Department of Physiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6085, USA
| | | | | | | | | |
Collapse
|
46
|
Bunse S, Schmidt M, Prochnow N, Zoidl G, Dermietzel R. Intracellular cysteine 346 is essentially involved in regulating Panx1 channel activity. J Biol Chem 2010; 285:38444-52. [PMID: 20829356 DOI: 10.1074/jbc.m110.101014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Pannexins constitute a family of proteins exhibiting predominantly hemichannel activity. Pannexin channels have been suggested to participate in a wide spectrum of biological functions such as propagation of calcium waves, release of IL-1β, and responses to ischemic conditions. At present, the molecular mechanisms regulating pannexin hemichannel activity are essentially unknown. Because cysteines have been shown to constitute key elements in regulating hemichannel properties of the connexin-type we performed site-directed mutagenesis of intracellular cysteine residues of Panx1. Cysteine to serine exchange (Cys → Ser) at the C-terminal position amino acid 346 led to a constitutively leaky hemichannel and subsequently to cell death. Increased channel activity was demonstrated by dye uptake and electrophysiological profiling in injected Xenopus laevis oocytes and transfected N2A cells. Mutations of the remaining intracellular cysteines did not result in major changes of Panx1 channel properties. From these data we conclude that the Cys-346 residue is important for proper functioning of the Panx1 channel.
Collapse
Affiliation(s)
- Stefanie Bunse
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, 44780 Bochum, Germany
| | | | | | | | | |
Collapse
|
47
|
Ambrosi C, Gassmann O, Pranskevich JN, Boassa D, Smock A, Wang J, Dahl G, Steinem C, Sosinsky GE. Pannexin1 and Pannexin2 channels show quaternary similarities to connexons and different oligomerization numbers from each other. J Biol Chem 2010; 285:24420-31. [PMID: 20516070 DOI: 10.1074/jbc.m110.115444] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Pannexins are homologous to innexins, the invertebrate gap junction family. However, mammalian pannexin1 does not form canonical gap junctions, instead forming hexameric oligomers in single plasma membranes and intracellularly. Pannexin1 acts as an ATP release channel, whereas less is known about the function of Pannexin2. We purified cellular membranes isolated from MDCK cells stably expressing rat Pannexin1 or Pannexin2 and identified pannexin channels (pannexons) in single membranes by negative stain and immunogold labeling. Protein gel and Western blot analysis confirmed Pannexin1 (Panx1) or Pannexin2 (Panx2) as the channel-forming proteins. We expressed and purified Panx1 and Panx2 using a baculovirus Sf9 expression system and obtained doughnut-like structures similar to those seen previously in purified connexin hemichannels (connexons) and mammalian membranes. Purified pannexons were comparable in size and overall appearance to Connexin46 and Connexin50 connexons. Pannexons and connexons were further analyzed by single-particle averaging for oligomer and pore diameters. The oligomer diameter increased with increasing monomer molecular mass, and we found that the measured oligomeric pore diameter for Panxs was larger than for Connexin26. Panx1 and Panx2 formed active homomeric channels in Xenopus oocytes and in vitro vesicle assays. Cross-linking and native gels of purified homomeric full-length and a C-terminal Panx2 truncation mutant showed a banding pattern more consistent with an octamer. We purified Panx1/Panx2 heteromeric channels and found that they were unstable over time, possibly because Panx1 and Panx2 homomeric pannexons have different monomer sizes and oligomeric symmetry from each other.
Collapse
Affiliation(s)
- Cinzia Ambrosi
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California 92093-0608, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
MacVicar BA, Thompson RJ. Non-junction functions of pannexin-1 channels. Trends Neurosci 2009; 33:93-102. [PMID: 20022389 DOI: 10.1016/j.tins.2009.11.007] [Citation(s) in RCA: 210] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 11/11/2009] [Accepted: 11/20/2009] [Indexed: 02/07/2023]
Abstract
Pannexins are large-pore ion channels with broad expression in the central nervous system (CNS). The channels function by releasing large signaling molecules, such ATP and arachidonic acid derivatives, from neurons and possibly astrocytes. They might also contribute to novel forms of non-synaptic communication in the CNS, thereby affecting synaptic function, astrocytic Ca(2+) wave propagation and possibly regulation of vascular tone in the brain. Panx1 activation in various in vitro pathological conditions implicates these channels in ischemic, excitotoxic and ATP-dependent cell death, whereas Panx coupling with purinergic receptors triggers the inflammasome. Novel functions for the pannexin channels are likely to be discovered as current understanding of how they are regulated in physiological and pathological situations improves.
Collapse
Affiliation(s)
- Brian A MacVicar
- Brain Research Centre, Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
49
|
Blum AE, Walsh BC, Dubyak GR. Extracellular osmolarity modulates G protein-coupled receptor-dependent ATP release from 1321N1 astrocytoma cells. Am J Physiol Cell Physiol 2009; 298:C386-96. [PMID: 19907018 DOI: 10.1152/ajpcell.00430.2009] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We previously reported that ATP release from 1321N1 human astrocytoma cells could be stimulated either by activation of G protein-coupled receptors (GPCR) or by hypotonic stress. Cheema et al. (Cheema TA, Ward CE, Fisher SK. J Pharmacol Exp Ther 315: 755-763, 2005) have demonstrated that thrombin activation of protease-activated receptor 1 (PAR1) in 1321N1 cells and primary astrocytes acts synergistically with hypotonic stress to gate the opening of volume-sensitive organic osmolyte and anion channels (VSOAC) and that hypertonic stress strongly inhibits PAR1 gating of VSOAC. We tested the hypothesis that a VSOAC-type permeability might comprise a GPCR-regulated pathway for ATP export by determining whether PAR1-sensitive ATP release from 1321N1 cells is similarly potentiated by hypotonicity but suppressed by hypertonic conditions. Strong hypotonic stress by itself elicited ATP release and positively modulated the response to thrombin. Thrombin-dependent ATP release was also potentiated by mild hypotonic stress that by itself did not stimulate ATP export. Notably, PAR1-sensitive ATP export was greatly inhibited in hypertonic medium. Neither the potency nor efficacy of thrombin as an activator of proximal PAR1 signaling was affected by hypotonicity or hypertonicity. 1,9-Dideoxyforskolin and carbenoxolone similarly attenuated PAR1-dependent ATP release and suppressed the PAR1-independent ATP elicited by strong hypotonic stress. Probenecid attenuated PAR1-stimulated ATP release under isotonic but not mild hypotonic conditions and had no effect on PAR1-independent release stimulated by strong hypotonicity. PAR1-dependent ATP export under all osmotic conditions required concurrent signaling by Ca(2+) mobilization and Rho-GTPase activation. In contrast, PAR1-independent ATP release triggered by strong hypotonicity required neither of these intracellular signals. Thus, we provide the new finding that GPCR-regulated ATP release from 1321N1 astrocytoma cells is remarkably sensitive to both positive and negative modulation by extracellular osmolarity. This supports a model wherein GPCR stimulation and osmotic stress converge on an ATP release pathway in astrocytes that exhibits several features of VSOAC-type channels.
Collapse
Affiliation(s)
- Andrew E Blum
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44120, USA
| | | | | |
Collapse
|
50
|
Civan MM. Transporters beyond transport. Focus on "Deregulation of apoptotic volume decrease and ionic movements in multidrug-resistant tumor cells: role of chloride channels". Am J Physiol Cell Physiol 2009; 298:C11-3. [PMID: 19846758 DOI: 10.1152/ajpcell.00459.2009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Mortimer M Civan
- Department of Physiology, A303 Richards Bldg., Univ. of Pennsylvania, Philadelphia, PA 19104-6085, USA.
| |
Collapse
|