1
|
Su AX, Ma ZJ, Li ZY, Li XY, Xia L, Ge YJ, Chen GH. Serum levels of neurotensin, pannexin-1, and sestrin-2 and the correlations with sleep quality or/and cognitive function in the patients with chronic insomnia disorder. Front Psychiatry 2024; 15:1360305. [PMID: 38803679 PMCID: PMC11128551 DOI: 10.3389/fpsyt.2024.1360305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Objectives To examine serum concentrations of neurotensin, pannexin-1 and sestrin-2, and their correlations with subjective and objective sleep quality and cognitive function in the patients with chronic insomnia disorder (CID). Methods Sixty-five CID patients were enrolled continuously and fifty-six good sleepers in the same period were served as healthy controls (HCs). Serum levels of neurotensin, pannexin-1 and sestrin-2 were measured by enzyme-linked immunosorbent assays. Sleep quality was assessed with the Pittsburgh Sleep Quality Index (PSQI) and polysomnography, and mood was evaluated by 17-item Hamilton Depression Rating Scale. General cognitive function was assessed with the Chinese-Beijing Version of Montreal Cognitive Assessment and spatial memory was evaluated by Blue Velvet Arena Test (BVAT). Results Relative to the HCs, the CID sufferers had higher levels of neurotensin (t=5.210, p<0.001) and pannexin-1 (Z=-4.169, p<0.001), and lower level of sestrin-2 (Z=-2.438, p=0.015). In terms of objective sleep measures, pannexin-1 was positively associated with total sleep time (r=0.562, p=0.002) and sleep efficiency (r=0.588, p=0.001), and negatively with wake time after sleep onset (r=-0.590, p=0.001) and wake time (r=-0.590, p=0.001); sestrin-2 was positively associated with percentage of rapid eye movement sleep (r=0.442, p=0.016) and negatively with non-rapid eye movement sleep stage 2 in the percentage (r=-0.394, p=0.034). Adjusted for sex, age and HAMD, pannexin-1 was still associated with the above objective sleep measures, but sestrin-2 was only negatively with wake time (r=-0.446, p=0.022). However, these biomarkers showed no significant correlations with subjective sleep quality (PSQI score). Serum concentrations of neurotensin and pannexin-1 were positively associated with the mean erroneous distance in the BVAT. Adjusted for sex, age and depression, neurotensin was negatively associated with MoCA score (r=-0.257, p=0.044), pannexin-1 was positively associated with the mean erroneous distance in the BVAT (r=0.270, p=0.033). Conclusions The CID patients had increased neurotensin and pannexin-1 and decreased sestrin-2 in the serum levels, indicating neuron dysfunction, which could be related to poor sleep quality and cognitive dysfunction measured objectively.
Collapse
Affiliation(s)
- Ai-Xi Su
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
- Department of General Medicine, the First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Zi-Jie Ma
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Zong-Yin Li
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Xue-Yan Li
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Lan Xia
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Yi-Jun Ge
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| |
Collapse
|
2
|
Khalili A, Safarian N, van Wijngaarden E, Zoidl GS, Zoidl GR, Rezai P. Loss of Panx1 function in zebrafish alters motor behavior in a lab-on-chip model of Parkinson's disease. J Neurosci Res 2023; 101:1814-1825. [PMID: 37688406 DOI: 10.1002/jnr.25241] [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: 12/20/2022] [Revised: 08/08/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023]
Abstract
Pannexin 1 (Panx1) forms ATP-permeable membrane channels that play roles in purinergic signaling in the nervous system. A link between Panx1 activity and neurodegenerative disorders including Parkinson's disease (PD) has been suggested, but experimental evidence is limited. Here, a zebrafish model of PD was produced by exposing panx1a+/+ and panx1a-/- zebrafish larvae to 6-hydroxydopamine (6-OHDA). Electrical stimulation in a microfluidic chip and quantitative real-time-qPCR of zebrafish larvae tested the role of Panx1 in both pathological and normal conditions. After 72-h treatment with 6-OHDA, the electric-induced locomotor activity of 5 days post fertilization (5dpf) panx1a+/+ larvae were reduced, while the stimulus did not affect locomotor activity of age-matched panx1a-/- larvae. A RT-qPCR analysis showed an increase in the expression of genes that are functionally related to dopaminergic signaling, like the tyrosine hydroxylase (th2) and the leucine-rich repeat kinase 2 (lrrk2). Extending the 6-OHDA treatment duration to 120 h caused a significant reduction in the locomotor response of 7dpf panx1a-/- larvae compared to the untreated panx1a-/- group. The RT-qPCR data showed a reduced expression of dopaminergic signaling genes in both genotypes. It was concluded that the absence of Panx1a channels compromised dopaminergic signaling in 6-OHDA-treated zebrafish larvae and that the increase in the expression of dopaminergic genes was transient, most likely due to a compensatory upregulation. We propose that zebrafish Panx1a models offer opportunities to shed light on PD's physiological and molecular basis. Panx1a might play a role on the progression of PD, and therefore deserves further investigation.
Collapse
Affiliation(s)
- Arezoo Khalili
- Department of Mechanical Engineering, York University, Toronto, Ontario, Canada
| | - Nickie Safarian
- Department of Biology, York University, Toronto, Ontario, Canada
| | | | - Georg S Zoidl
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Georg R Zoidl
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Pouya Rezai
- Department of Mechanical Engineering, York University, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Seo JH, Dalal MS, Contreras JE. Pannexin-1 Channels as Mediators of Neuroinflammation. Int J Mol Sci 2021; 22:ijms22105189. [PMID: 34068881 PMCID: PMC8156193 DOI: 10.3390/ijms22105189] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is a major component of central nervous system (CNS) injuries and neurological diseases, including Alzheimer’s disease, multiple sclerosis, neuropathic pain, and brain trauma. The activation of innate immune cells at the damage site causes the release of pro-inflammatory cytokines and chemokines, which alter the functionality of nearby tissues and might mediate the recruitment of leukocytes to the injury site. If this process persists or is exacerbated, it prevents the adequate resolution of the inflammation, and ultimately enhances secondary damage. Adenosine 5′ triphosphate (ATP) is among the molecules released that trigger an inflammatory response, and it serves as a chemotactic and endogenous danger signal. Extracellular ATP activates multiple purinergic receptors (P2X and P2Y) that have been shown to promote neuroinflammation in a variety of CNS diseases. Recent studies have shown that Pannexin-1 (Panx1) channels are the principal conduits of ATP release from dying cells and innate immune cells in the brain. Herein, we review the emerging evidence that directly implicates Panx-1 channels in the neuroinflammatory response in the CNS.
Collapse
Affiliation(s)
- Joon Ho Seo
- Department of Neurology and Nash Family, Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA;
| | - Miloni S. Dalal
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA;
| | - Jorge E. Contreras
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA;
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
- Correspondence: ; Tel.: +1-530-754-2770
| |
Collapse
|
4
|
Whyte-Fagundes P, Kurtenbach S, Zoidl C, Shestopalov VI, Carlen PL, Zoidl G. A Potential Compensatory Role of Panx3 in the VNO of a Panx1 Knock Out Mouse Model. Front Mol Neurosci 2018; 11:135. [PMID: 29780304 PMCID: PMC5946002 DOI: 10.3389/fnmol.2018.00135] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 04/03/2018] [Indexed: 12/30/2022] Open
Abstract
Pannexins (Panx) are integral membrane proteins, with Panx1 being the best-characterized member of the protein family. Panx1 is implicated in sensory processing, and knockout (KO) animal models have become the primary tool to investigate the role(s) of Panx1 in sensory systems. Extending previous work from our group on primary olfaction, the expression patterns of Panxs in the vomeronasal organ (VNO), an auxiliary olfactory sense organ with a role in reproduction and social behavior, were compared. Using qRT-PCR and Immunohistochemistry (IHC), we confirmed the loss of Panx1, found similar Panx2 expression levels in both models, and a significant upregulation of Panx3 in mice with a global ablation of Panx1. Specifically, Panx3 showed upregulated expression in nerve fibers of the non-sensory epithelial layer in juvenile and adult KO mice and in the sensory layer of adults, which overlaps with Panx1 expression areas in WT populations. Since both social behavior and evoked ATP release in the VNO was not compromised in KO animals, we hypothesized that Panx3 could compensate for the loss of Panx1. This led us to compare Panx1 and Panx3 channels in vitro, demonstrating similar dye uptake and ATP release properties. Outcomes of this study strongly suggest that Panx3 may functionally compensate for the loss of Panx1 in the VNO of the olfactory system, ensuring sustained chemosensory processing. This finding extends previous reports on the upregulation of Panx3 in arterial walls and the skin of Panx1 KO mice, suggesting that roles of Panx1 warrant uncharacterized safeguarding mechanisms involving Panx3.
Collapse
Affiliation(s)
- Paige Whyte-Fagundes
- Department of Biology, York University, Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Stefan Kurtenbach
- Department of Biology, York University, Toronto, ON, Canada.,Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | - Valery I Shestopalov
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Peter L Carlen
- Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Georg Zoidl
- Department of Biology, York University, Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Psychology, York University, Toronto, ON, Canada
| |
Collapse
|
5
|
Robinson JC, Chapman CA, Courtemanche R. Gap Junction Modulation of Low-Frequency Oscillations in the Cerebellar Granule Cell Layer. THE CEREBELLUM 2018; 16:802-811. [PMID: 28421552 DOI: 10.1007/s12311-017-0858-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Local field potential (LFP) oscillations in the granule cell layer (GCL) of the cerebellar cortex have been identified previously in the awake rat and monkey during immobility. These low-frequency oscillations are thought to be generated through local circuit interactions between Golgi cells and granule cells within the GCL. Golgi cells display rhythmic firing and pacemaking properties, and also are electrically coupled through gap junctions within the GCL. Here, we tested if gap junctions in the rat cerebellar cortex contribute to the generation of LFP oscillations in the GCL. We recorded LFP oscillations under urethane anesthesia, and examined the effects of local infusion of gap junction blockers on 5-15 Hz oscillations. Local infusion of the gap junction blockers carbenoxolone and mefloquine resulted in significant decreases in the power of oscillations over a 30-min period, but the power of oscillations was unchanged in control experiments following vehicle injections. In addition, infusion of gap junction blockers had no significant effect on multi-unit activity, suggesting that the attenuation of low-frequency oscillations was likely due to reductions in electrical coupling rather than a decreased excitability within the granule cell layer. Our results indicate that electrical coupling among the Golgi cell networks in the cerebellar cortex contributes to the local circuit mechanisms that promote the occurrence of GCL LFP slow oscillations in the anesthetized rat.
Collapse
Affiliation(s)
- Jennifer Claire Robinson
- Department of Exercise Science, and the FRQS Groupe de Recherche en Neurobiologie Comportementale (CSBN), Concordia University, SP-165-03, 7141 Sherbrooke Street West, Montreal, QC, H4B 1R6, Canada
| | - C Andrew Chapman
- Department of Psychology, and the FRQS Groupe de Recherche en Neurobiologie Comportementale (CSBN), Concordia University, Montreal, Canada
| | - Richard Courtemanche
- Department of Exercise Science, and the FRQS Groupe de Recherche en Neurobiologie Comportementale (CSBN), Concordia University, SP-165-03, 7141 Sherbrooke Street West, Montreal, QC, H4B 1R6, Canada.
| |
Collapse
|
6
|
Pannexin-1 channels in epilepsy. Neurosci Lett 2017; 695:71-75. [PMID: 28886985 DOI: 10.1016/j.neulet.2017.09.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 07/20/2017] [Accepted: 09/01/2017] [Indexed: 02/07/2023]
Abstract
Pannexin-1 (Panx1) expression is raised in several animal seizure models and in resected human epileptic brain tissue, suggesting relevance to epilepsy. Multiple factors that are characteristic of seizures are thought to regulate Panx1 channel opening, including elevated levels of extracellular K+. Panx1, when open, 1) releases ATP, glutamate, and other metabolites into the extracellular medium, and 2) may depolarize the membrane due to a channel reversal potential around 0mV. Resultant ATP release from stimulated Panx1 can activate purinergic receptors, including P2X7 receptors. Glutamate and other signaling molecules released by Panx1 opening may have both excitatory and inhibitory actions on seizure generation. This review examines the critical and complex roles of Panx1 channels in epilepsy, which could provide a basis for future therapeutics.
Collapse
|
7
|
Whyte-Fagundes P, Zoidl G. Mechanisms of pannexin1 channel gating and regulation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:65-71. [PMID: 28735901 DOI: 10.1016/j.bbamem.2017.07.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 01/07/2023]
Abstract
Pannexins are a family of integral membrane proteins with distinct post-translational modifications, sub-cellular localization and tissue distribution. Panx1 is the most studied and best-characterized isoform of this gene family. The ubiquitous expression, as well as its function as a major ATP release and nucleotide permeation channel, makes Panx1 a primary candidate for participating in the pathophysiology of CNS disorders. While many investigations revolve around Panx1 functions in health and disease, more recently, details started emerging about mechanisms that control Panx1 channel activity. These advancements in Panx1 biology have revealed that beyond its classical role as an unopposed plasma membrane channel, it participates in alternative pathways involving multiple intracellular compartments, protein complexes and a myriad of extracellular participants. Here, we review recent progress in our understanding of Panx1 at the center of these pathways, highlighting its modulation in a context specific manner. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
| | - Georg Zoidl
- Biology, York University, Toronto, Canada; Psychology, York University, Toronto, Canada.
| |
Collapse
|
8
|
Valdebenito S, Barreto A, Eugenin EA. The role of connexin and pannexin containing channels in the innate and acquired immune response. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:154-165. [PMID: 28559189 DOI: 10.1016/j.bbamem.2017.05.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/17/2017] [Accepted: 05/25/2017] [Indexed: 12/20/2022]
Abstract
Connexin (Cx) and pannexin (Panx) containing channels - gap junctions (GJs) and hemichannels (HCs) - are present in virtually all cells and tissues. Currently, the role of these channels under physiological conditions is well defined. However, their role in the immune response and pathological conditions has only recently been explored. Data from several laboratories demonstrates that infectious agents, including HIV, have evolved to take advantage of GJs and HCs to improve viral/bacterial replication, enhance inflammation, and help spread toxicity into neighboring areas. In the current review, we discuss the role of Cx and Panx containing channels in immune activation and the pathogenesis of several infectious diseases. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Public Health Research Institute (PHRI), Newark, NJ, USA; Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA
| | - Andrea Barreto
- Public Health Research Institute (PHRI), Newark, NJ, USA; Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA
| | - Eliseo A Eugenin
- Public Health Research Institute (PHRI), Newark, NJ, USA; Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA.
| |
Collapse
|
9
|
Xu J, Chen L, Li L. Pannexin hemichannels: A novel promising therapy target for oxidative stress related diseases. J Cell Physiol 2017; 233:2075-2090. [PMID: 28295275 DOI: 10.1002/jcp.25906] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 12/16/2022]
Abstract
Pannexins, which contain three subtypes: pannexin-1, -2, and -3, are vertebrate glycoproteins that form non-junctional plasma membrane intracellular hemichannels via oligomerization. Oxidative stress refers to an imbalance of the generation and elimination of reactive oxygen species (ROS). Studies have shown that elevated ROS levels are pivotal in the development of a variety of diseases. Recent studies indicate that the occurrence of these oxidative stress related diseases is associated with pannexin hemichannels. It is also reported that pannexins regulate the production of ROS which in turn may increase the opening of pannexin hemichannels. In this paper, we review recent researches about the important role of pannexin hemichannels in oxidative stress related diseases. Thus, pannexin hemichannels, novel therapeutic targets, hold promise in managing oxidative stress related diseases such as the tumor, inflammatory bowel diseases (IBD), pulmonary fibrosis, chronic obstructive pulmonary disease (COPD), cardiovascular disease, insulin resistance (IR), and neural degeneration diseases.
Collapse
Affiliation(s)
- Jin Xu
- Learning Key Laboratory for Pharmacoproteomics, Institute of Pharmacy and Pharmacology, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| | - Linxi Chen
- Learning Key Laboratory for Pharmacoproteomics, Institute of Pharmacy and Pharmacology, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| | - Lanfang Li
- Learning Key Laboratory for Pharmacoproteomics, Institute of Pharmacy and Pharmacology, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| |
Collapse
|
10
|
Scemes E, Velíšková J. Exciting and not so exciting roles of pannexins. Neurosci Lett 2017; 695:25-31. [PMID: 28284836 DOI: 10.1016/j.neulet.2017.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/02/2017] [Accepted: 03/06/2017] [Indexed: 01/24/2023]
Abstract
It is the current view that purinergic signaling regulates many physiological functions. Pannexin1 (Panx1), a member of the gap junction family of proteins is an ATP releasing channel that plays important physio-pathological roles in various tissues, including the CNS. Upon binding to purinergic receptors expressed in neural cells, ATP triggers cellular responses including increased cell proliferation, cell morphology changes, release of cytokines, and regulation of neuronal excitability via release of glutamate, GABA and ATP itself. Under pathological conditions such as ischemia, trauma, inflammation, and epilepsy, extracellular ATP concentrations increases drastically but the consequences of this surge is still difficult to characterize due to its rapid metabolism in ADP and adenosine, the latter having inhibitory action on neuronal activity. For seizures, for instance, the excitatory effect of ATP on neuronal activity is mainly related to its action of P2X receptors, while the inhibitory effects are related to activation of P1, adenosine receptors. Here we provide a mini review on the properties of pannexins with a main focus on Panx1 and its involvement in seizure activity. Although there are only few studies implicating Panx1 in seizures, they are illustrative of the dual role that Panx1 has on neuronal excitability.
Collapse
Affiliation(s)
- Eliana Scemes
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - Jana Velíšková
- Departments of Cell Biology & Anatomy, Obstetrics & Gynecology and Neurology, New York Medical College, Valhalla, NY, 10595, USA.
| |
Collapse
|
11
|
Abstract
Cell-cell and cell-matrix communications play important roles in both cell proliferation and differentiation. Gap junction proteins mediate signaling communication by exchanging small molecules and dramatically stimulating intracellular signaling pathways to determine cell fate. Vertebrates have 2 gap junction families: pannexins (Panxs) and connexins (Cxs). Unlike Cxs, the functions of Panxs are not fully understood. In skeletal formation, Panx3 and Cx43 are the most abundantly expressed gap junction proteins from each family. Panx3 is induced in the transient stage from the proliferation and differentiation of chondrocytes and osteoprogenitor cells. Panx3 regulates both chondrocyte and osteoblast differentiation via the activation of intracellular Ca2+ signaling pathways through multiple channel activities: hemichannels, endoplasmic reticulum (ER) Ca2+ channels, and gap junctions. Moreover, Panx3 also inhibits osteoprogenitor cell proliferation and promotes cell cycle exit through the inactivation of Wnt/β-catenin signaling and the activation of p21. Panx3-knockout (KO) mice have more severe skeletal abnormalities than those of Cx43-KO mice. A phenotypic analysis of Panx3-KO mice indicates that Panx3 regulates the terminal differentiation of chondrocytes by promoting vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMP) 13. Based on the generation of Panx3-/-; Cx43-/- mice, Panx3 is upstream of Cx43 in osteogenesis. Panx3 promotes Cx43 expression by regulating Wnt/β-catenin signaling and osterix expression. Further, although Panx3 can function in 3 ways, Cx43 cannot function through the ER Ca2+ channel, only via the hemichannels and gap junction routes. In this review, we discuss the current knowledge regarding the roles of Panx3 in skeletal formation and address the potential for new therapies in the treatment of diseases and pathologies associated with Panx3, such as osteoarthritis (OA).
Collapse
Affiliation(s)
- M Ishikawa
- 1 Division of Operative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan.,2 Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Y Yamada
- 2 Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
12
|
Velasquez S, Malik S, Lutz SE, Scemes E, Eugenin EA. Pannexin1 Channels Are Required for Chemokine-Mediated Migration of CD4+ T Lymphocytes: Role in Inflammation and Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2016; 196:4338-47. [PMID: 27076682 DOI: 10.4049/jimmunol.1502440] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/09/2016] [Indexed: 12/19/2022]
Abstract
Pannexin1 (Panx1) channels are large high conductance channels found in all vertebrates that can be activated under several physiological and pathological conditions. Our published data indicate that HIV infection results in the extended opening of Panx1 channels (5-60 min), allowing for the secretion of ATP through the channel pore with subsequent activation of purinergic receptors, which facilitates HIV entry and replication. In this article, we demonstrate that chemokines, which bind CCR5 and CXCR4, especially SDF-1α/CXCL12, result in a transient opening (peak at 5 min) of Panx1 channels found on CD4(+) T lymphocytes, which induces ATP secretion, focal adhesion kinase phosphorylation, cell polarization, and subsequent migration. Increased migration of immune cells is key for the pathogenesis of several inflammatory diseases including multiple sclerosis (MS). In this study, we show that genetic deletion of Panx1 reduces the number of the CD4(+) T lymphocytes migrating into the spinal cord of mice subjected to experimental autoimmune encephalomyelitis, an animal model of MS. Our results indicate that opening of Panx1 channels in response to chemokines is required for CD4(+) T lymphocyte migration, and we propose that targeting Panx1 channels could provide new potential therapeutic approaches to decrease the devastating effects of MS and other inflammatory diseases.
Collapse
Affiliation(s)
- Stephani Velasquez
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ 07103; Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ 07103
| | - Shaily Malik
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ 07103; Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ 07103
| | - Sarah E Lutz
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697; and
| | - Eliana Scemes
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Eliseo A Eugenin
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ 07103; Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ 07103;
| |
Collapse
|
13
|
Raslan A, Hainz N, Beckmann A, Tschernig T, Meier C. Pannexin-1 expression in developing mouse nervous system: new evidence for expression in sensory ganglia. Cell Tissue Res 2015; 364:29-41. [PMID: 26453396 DOI: 10.1007/s00441-015-2294-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022]
Abstract
Pannexin1 (Panx1) is one of three members of the pannexin protein family. The expression of Panx1 mRNA has been extensively investigated from late embryonic to adult stages. In contrast, expression during early embryonic development is largely unknown. Our aim is to examine the temporal and spatial expression of Panx1 in mouse embryonic development by focusing on embryonic days (E) 9.5 to 12.5. Whole embryos are investigated in order to provide a comprehensive survey. Analyses were performed at the mRNA level by using reverse transcription plus the polymerase chain reaction and whole-mount in situ hybridization. Panx1 mRNA was detected in the heads and bodies of embryos at all developmental stages investigated (E9.5, E10.5, E11.5, E12.5). In particular, the nervous system expressed Panx1 at an early time point. Interestingly, Panx1 expression was found in afferent ganglia of the cranial nerves and spinal cord. This finding is of particular interest in the context of neuropathic pain and other Panx1-related neurological disorders. Our study shows, for the first time, that Panx1 is expressed in the central and peripheral nervous system during early developmental stages. The consequences of Panx1 deficiency or inhibition in a number of experimental paradigms might therefore be predicated on changes during early development.
Collapse
Affiliation(s)
- Abdulrahman Raslan
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66424, Homburg, Saar, Germany
| | - Nadine Hainz
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66424, Homburg, Saar, Germany
| | - Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66424, Homburg, Saar, Germany
| | - Thomas Tschernig
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66424, Homburg, Saar, Germany
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66424, Homburg, Saar, Germany.
| |
Collapse
|
14
|
Bravo D, Maturana CJ, Pelissier T, Hernández A, Constandil L. Interactions of pannexin 1 with NMDA and P2X7 receptors in central nervous system pathologies: Possible role on chronic pain. Pharmacol Res 2015. [PMID: 26211949 DOI: 10.1016/j.phrs.2015.07.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pannexin 1 (Panx1) is a glycoprotein that acts as a membrane channel in a wide variety of tissues in mammals. In the central nervous system (CNS) Panx1 is expressed in neurons, astrocytes and microglia, participating in the pathophysiology of some CNS diseases, such as epilepsy, anoxic depolarization after stroke and neuroinflammation. In these conditions Panx1 acts as an important modulator of the neuroinflammatory response, by secreting ATP, by interacting with the P2X7 receptor (P2X7R), and as an amplifier of NMDA receptor (NMDAR) currents, particularly in conditions of pathological neuronal hyperexcitability. Here, we briefly reviewed the current evidences that support the interaction of Panx1 with NMDAR and P2X7R in pathological contexts of the CNS, with special focus in recent data supporting that Panx1 is involved in chronic pain signaling by interacting with NMDAR in neurons and with P2X7R in glia. The participation of Panx1 in chronic pain constitutes a novel topic for research in the field of clinical neurosciences and a potential target for pharmacological interventions in chronic pain.
Collapse
Affiliation(s)
- D Bravo
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Chile; School of Kinesiology, Faculty of Sport, Health and Recreation, University Bernardo O'Higgins, Chile.
| | - C J Maturana
- Departamento de Fisiología, Pontificia Universidad Católica De Chile, Chile
| | - T Pelissier
- Program of Molecular and Clinical Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Chile
| | - A Hernández
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Chile
| | - L Constandil
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Chile
| |
Collapse
|
15
|
Li L, He L, Wu D, Chen L, Jiang Z. Pannexin-1 channels and their emerging functions in cardiovascular diseases. Acta Biochim Biophys Sin (Shanghai) 2015; 47:391-6. [PMID: 25921414 DOI: 10.1093/abbs/gmv028] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 02/04/2015] [Indexed: 11/15/2022] Open
Abstract
Pannexin-1, Pannexin-2, and Pannexin-3 are three members of the Pannexin family of channel-forming glycoprotein. Their primary function is defined by their ability to form single-membrane channels. Pannexin-1 ubiquitously exists in many cells and organs throughout the body and is specially distributed in the circulatory system, while the expressions of Pannexin-2 and Pannexin-3 are mostly restricted to organs and tissues. Pannexin-1 oligomers have been shown to be functional single membrane channels that connect intracellular and extracellular compartments and are not intercellular channels in appositional membranes. The physiological functions of Pannexin-1 are to link to the adenosine triphosphate efflux that acts as a paracrine signal, and regulate cellular inflammasomes in a variety of cell types under physiological and pathophysiological conditions. However, there are still many functions to be explored. This review summarizes recent reports and discusses the role of Pannexin-1 in cardiovascular diseases, including ischemia, arrhythmia, cardiac fibrosis, and hypertension. Pannexin-1 has been suggested as an exciting, clinically relevant target in cardiovascular diseases.
Collapse
Affiliation(s)
- Lanfang Li
- Post-doctoral Mobile Stations for Basic Medicine, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Lu He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Di Wu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Linxi Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Zhisheng Jiang
- Post-doctoral Mobile Stations for Basic Medicine, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China
| |
Collapse
|
16
|
Cisneros-Mejorado A, Gottlieb M, Cavaliere F, Magnus T, Koch-Nolte F, Scemes E, Pérez-Samartín A, Matute C. Blockade of P2X7 receptors or pannexin-1 channels similarly attenuates postischemic damage. J Cereb Blood Flow Metab 2015; 35:843-50. [PMID: 25605289 PMCID: PMC4420860 DOI: 10.1038/jcbfm.2014.262] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/19/2014] [Accepted: 12/22/2014] [Indexed: 11/09/2022]
Abstract
The role of P2X7 receptors and pannexin-1 channels in ischemic damage remains controversial. Here, we analyzed their contribution to postanoxic depolarization after ischemia in cultured neurons and in brain slices. We observed that pharmacological blockade of P2X7 receptors or pannexin-1 channels delayed the onset of postanoxic currents and reduced their slope, and that simultaneous inhibition did not further enhance the effects of blocking either one. These results were confirmed in acute cortical slices from P2X7 and pannexin-1 knockout mice. Oxygen-glucose deprivation in cortical organotypic cultures caused neuronal death that was reduced with P2X7 and pannexin-1 blockers as well as in organotypic cultures derived from mice lacking P2X7 and pannexin 1. Subsequently, we used transient middle cerebral artery occlusion to monitor the neuroprotective effect of those drugs in vivo. We found that P2X7 and pannexin-1 antagonists, and their ablation in knockout mice, substantially attenuated the motor symptoms and reduced the infarct volume to ~50% of that in vehicle-treated or wild-type animals. These results show that P2X7 receptors and pannexin-1 channels are major mediators of postanoxic depolarization in neurons and of brain damage after ischemia, and that they operate in the same deleterious signaling cascade leading to neuronal and tissue demise.
Collapse
Affiliation(s)
- Abraham Cisneros-Mejorado
- Achucarro Basque Center for Neuroscience, Departamento de Neurociencias and CIBERNED, Universidad del País Vasco (UPV/EHU), Leioa, Spain
| | - Miroslav Gottlieb
- 1] Achucarro Basque Center for Neuroscience, Departamento de Neurociencias and CIBERNED, Universidad del País Vasco (UPV/EHU), Leioa, Spain [2] Institute of Neurobiology, Slovak Academy of Sciences, Kosice, Slovak Republic
| | - Fabio Cavaliere
- 1] Achucarro Basque Center for Neuroscience, Departamento de Neurociencias and CIBERNED, Universidad del País Vasco (UPV/EHU), Leioa, Spain [2] Neurotek-UPV/EHU, Parque Tecnológico de Bizkaia, Zamudio, Spain
| | - Tim Magnus
- Department of Neurology, University Hospital Hamburg, Hamburg, Germany
| | | | - Eliana Scemes
- Dominick P. Purpura Department of Neurosciences, Albert Einstein College of Medicine, New York, New York, USA
| | - Alberto Pérez-Samartín
- 1] Achucarro Basque Center for Neuroscience, Departamento de Neurociencias and CIBERNED, Universidad del País Vasco (UPV/EHU), Leioa, Spain [2] Neurotek-UPV/EHU, Parque Tecnológico de Bizkaia, Zamudio, Spain
| | - Carlos Matute
- 1] Achucarro Basque Center for Neuroscience, Departamento de Neurociencias and CIBERNED, Universidad del País Vasco (UPV/EHU), Leioa, Spain [2] Neurotek-UPV/EHU, Parque Tecnológico de Bizkaia, Zamudio, Spain
| |
Collapse
|
17
|
Diezmos EF, Sandow SL, Perera DS, King DW, Bertrand PP, Liu L. Pannexin-2 is expressed in the human colon with extensive localization in the enteric nervous system. Neurogastroenterol Motil 2015; 27:672-83. [PMID: 25773474 DOI: 10.1111/nmo.12541] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 02/16/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Pannexin-2 (Panx2) is a member of the novel group of membrane spanning protein channels present in the central nervous system. Limited studies have examined Panx2 in the intestine, where it may have important physiological roles. The present study characterized Panx2 expression and localization in the human colon in health and disease states. METHODS Immunofluorescence determined Panx2 localization and co-localization, and quantitative real-time PCR and Western blot determined gene and protein expression in ulcerative colitis (UC), Crohn's disease (CD), and control human colon. KEY RESULTS Panx2 was widely expressed in myenteric and submucosal ganglia, particularly in the cytoplasm of neurons. Panx2 was also expressed on smooth muscle of the muscularis and blood vessels, some non-lymphoid leukocytes, mast cells, and mucosal epithelial cells. Co-localization of Panx2 occurred with β-tubulin, neuronal nitric oxide synthase, substance P, vesicular acetylcholine transporter, and calcitonin gene-related peptide, indicating widespread Panx2 expression in extrinsic and intrinsic neurons. Molecular studies revealed a 3.4-fold higher level of Panx2 mRNA in ascending compared to sigmoid muscularis (p < 0.05), despite similar protein levels. Similarly, UC muscularis showed a 35-fold up-regulation in Panx2 mRNA, but not in protein (p < 0.05). CONCLUSIONS & INFERENCES Here, we demonstrated the dense expression of Panx2 in the enteric nervous system and the co-localization of Panx2 with a spectrum of neuronal markers, indicating that Panx2 may be involved in mediating neurotransmission in the colon. The substantial increase in Panx2 mRNA in UC muscle but not protein suggests that the Panx2 translation process may be disrupted in UC.
Collapse
Affiliation(s)
- E F Diezmos
- School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | | | | | | | | | | |
Collapse
|
18
|
Boassa D, Nguyen P, Hu J, Ellisman MH, Sosinsky GE. Pannexin2 oligomers localize in the membranes of endosomal vesicles in mammalian cells while Pannexin1 channels traffic to the plasma membrane. Front Cell Neurosci 2015; 8:468. [PMID: 25698922 PMCID: PMC4313697 DOI: 10.3389/fncel.2014.00468] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/27/2014] [Indexed: 12/13/2022] Open
Abstract
Pannexin2 (Panx2) is the largest of three members of the pannexin proteins. Pannexins are topologically related to connexins and innexins, but serve different functional roles than forming gap junctions. We previously showed that pannexins form oligomeric channels but unlike connexins and innexins, they form only single membrane channels. High levels of Panx2 mRNA and protein in the Central Nervous System (CNS) have been documented. Whereas Pannexin1 (Panx1) is fairly ubiquitous and Pannexin3 (Panx3) is found in skin and connective tissue, both are fully glycosylated, traffic to the plasma membrane and have functions correlated with extracellular ATP release. Here, we describe trafficking and subcellular localizations of exogenous Panx2 and Panx1 protein expression in MDCK, HeLa, and HEK 293T cells as well as endogenous Panx1 and Panx2 patterns in the CNS. Panx2 was found in intracellular localizations, was partially N-glycosylated, and localizations were non-overlapping with Panx1. Confocal images of hippocampal sections immunolabeled for the astrocytic protein GFAP, Panx1 and Panx2 demonstrated that the two isoforms, Panx1 and Panx2, localized at different subcellular compartments in both astrocytes and neurons. Using recombinant fusions of Panx2 with appended genetic tags developed for correlated light and electron microscopy and then expressed in different cell lines, we determined that Panx2 is localized in the membrane of intracellular vesicles and not in the endoplasmic reticulum as initially indicated by calnexin colocalization experiments. Dual immunofluorescence imaging with protein markers for specific vesicle compartments showed that Panx2 vesicles are early endosomal in origin. In electron tomographic volumes, cross-sections of these vesicles displayed fine structural details and close proximity to actin filaments. Thus, pannexins expressed at different subcellular compartments likely exert distinct functional roles, particularly in the nervous system.
Collapse
Affiliation(s)
- Daniela Boassa
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA
| | - Phuong Nguyen
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA
| | - Junru Hu
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA ; Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Gina E Sosinsky
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA ; Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
19
|
Cisneros-Mejorado A, Pérez-Samartín A, Gottlieb M, Matute C. ATP signaling in brain: release, excitotoxicity and potential therapeutic targets. Cell Mol Neurobiol 2015; 35:1-6. [PMID: 25096398 DOI: 10.1007/s10571-014-0092-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 07/19/2014] [Indexed: 10/24/2022]
Abstract
Adenosine 5'-triphosphate (ATP) is released as a genuine co-transmitter, or as a principal purinergic neurotransmitter, in an exocytotic and non-exocytotic manner. It activates ionotropic (P2X) and metabotropic (P2Y) receptors which mediate a plethora of functions in the brain. In particular, P2X7 receptor (P2X7R) are expressed in all brain cells and its activation can form a large pore allowing the passage of organic cations, the leakage of metabolites of up to 900 Da and the release of ATP itself. In turn, pannexins (Panx) are a family of proteins forming hemichannels that can release ATP. In this review, we summarize the progress in the understanding of the mechanisms of ATP release both in physiological and pathophysiological stages. We also provide data suggesting that P2X7R and pannexin 1 (Panx1) may form a large pore in cortical neurons as assessed by electrophysiology. Finally, the participation of calcium homeostasis modulator 1 is also suggested, another non-selective ion channel that can release ATP, and that could play a role in ischemic events, together with P2X7 and Panx1 during excitotoxicity by ATP.
Collapse
Affiliation(s)
- Abraham Cisneros-Mejorado
- Achucarro Basque Center for Neuroscience, Departamento de Neurociencias and CIBERNED, Universidad Del País Vasco, Leioa, Vizcaya, Spain
| | | | | | | |
Collapse
|
20
|
Mondal P, Trigun SK. Pannexin1 as a novel cerebral target in pathogenesis of hepatic encephalopathy. Metab Brain Dis 2014; 29:1007-15. [PMID: 24807590 DOI: 10.1007/s11011-014-9556-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 04/28/2014] [Indexed: 12/11/2022]
Abstract
Hepatic encephalopathy (HE) represents a nervous system disorder caused due to liver dysfunction. HE is broadly classified as acute/overt and moderate-minimal HE. Since HE syndrome severely affects quality of life of the patients and it may be life threatening, it is important to develop effective therapeutic strategy against HE. Mainly ammonia neurotoxicity is considered accountable for HE. Increased level of ammonia in the brain activates glutamate-NMDA (N-methyl-D-aspartate) receptor (NMDAR) pathway leading to Ca(2+) influx, energy deficit and oxidative stress in the post synaptic neurons. Moreover, NMDAR blockage has been found to be a poor therapeutic option, as this neurotransmitter receptor plays important role in maintaining normal neurophysiology of the brain. Thus, searching new molecular players in HE pathogenesis is of current concern. There is an evolving concept about roles of the trans-membrane channels in the pathogenesis of a number of neurological complications. Pannexin1 (Panx1) is one of them and has been described to be implicated in stroke, epilepsy and ischemia. Importantly, the pathogenesis of these complications relates to some extent with NMDAR over activation. Thus, it is speculated that HE pathogenesis might also involve Panx1. Indeed, some recent observations in the animal models of HE provide support to this argument. Since opening of Panx1 channel is mostly associated with the neuronal dysfunctions, down regulation of this channel could serve as a relevant therapeutic strategy without producing any serious side effects. In the review article an attempt has been made to summarize the current information on implication of Panx1 in the brain disorders and its prospects for being examined as pharmacological target in HE pathogenesis.
Collapse
Affiliation(s)
- Papia Mondal
- Biochemistry Section Centre of Advanced Study in Zoology, Banaras Hindu university, Varanasi, 221005, India
| | | |
Collapse
|
21
|
Kurtenbach S, Kurtenbach S, Zoidl G. Emerging functions of pannexin 1 in the eye. Front Cell Neurosci 2014; 8:263. [PMID: 25309318 PMCID: PMC4163987 DOI: 10.3389/fncel.2014.00263] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/14/2014] [Indexed: 01/23/2023] Open
Abstract
Pannexin 1 (Panx1) is a high-conductance, voltage-gated channel protein found in vertebrates. Panx1 is widely expressed in many organs and tissues, including sensory systems. In the eye, Panx1 is expressed in major divisions including the retina, lens and cornea. Panx1 is found in different neuronal and non-neuronal cell types. The channel is mechanosensitive and responds to changes in extracellular ATP, intracellular calcium, pH, or ROS/nitric oxide. Since Panx1 channels operate at the crossroad of major signaling pathways, physiological functions in important autocrine and paracrine feedback signaling mechanisms were hypothesized. This review starts with describing in depth the initial Panx1 expression and localization studies fostering functional studies that uncovered distinct roles in processing visual information in subsets of neurons in the rodent and fish retina. Panx1 is expressed along the entire anatomical axis from optical nerve to retina and cornea in glia, epithelial and endothelial cells as well as in neurons. The expression and diverse localizations throughout the eye points towards versatile functions of Panx1 in neuronal and non-neuronal cells, implicating Panx1 in the crosstalk between immune and neural cells, pressure related pathological conditions like glaucoma, wound repair or neuronal cell death caused by ischemia. Summarizing the literature on Panx1 in the eye highlights the diversity of emerging Panx1 channel functions in health and disease.
Collapse
Affiliation(s)
- Sarah Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Stefan Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Georg Zoidl
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada ; Department of Biology, Faculty of Science, York University Toronto, ON, Canada
| |
Collapse
|
22
|
Kurtenbach S, Whyte-Fagundes P, Gelis L, Kurtenbach S, Brazil E, Zoidl C, Hatt H, Shestopalov VI, Zoidl G. Investigation of olfactory function in a Panx1 knock out mouse model. Front Cell Neurosci 2014; 8:266. [PMID: 25309319 PMCID: PMC4162419 DOI: 10.3389/fncel.2014.00266] [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: 05/27/2014] [Accepted: 08/18/2014] [Indexed: 01/01/2023] Open
Abstract
Pannexin 1 (Panx1), the most extensively investigated member of a channel-forming protein family, is able to form pores conducting molecules up to 1.5 kDa, like ATP, upon activation. In the olfactory epithelium (OE), ATP modulates olfactory responsiveness and plays a role in proliferation and differentiation of olfactory sensory neurons (OSNs). This process continuously takes place in the OE, as neurons are replaced throughout the whole lifespan. The recent discovery of Panx1 expression in the OE raises the question whether Panx1 mediates ATP release responsible for modulating chemosensory function. In this study, we analyzed pannexin expression in the OE and a possible role of Panx1 in olfactory function using a Panx1−/− mouse line with a global ablation of Panx1. This mouse model has been previously used to investigate Panx1 functions in the retina and adult hippocampus. Here, qPCR, in-situ hybridization, and immunohistochemistry (IHC) demonstrated that Panx1 is expressed in axon bundles deriving from sensory neurons of the OE. The localization, distribution, and expression of major olfactory signal transduction proteins were not significantly altered in Panx1−/− mice. Further, functional analysis of Panx1−/− animals does not reveal any major impairment in odor perception, indicated by electroolfactogram (EOG) measurements and behavioral testing. However, ATP release evoked by potassium gluconate application was reduced in Panx1−/− mice. This result is consistent with previous reports on ATP release in isolated erythrocytes and spinal or lumbar cord preparations from Panx1−/− mice, suggesting that Panx1 is one of several alternative pathways to release ATP in the olfactory system.
Collapse
Affiliation(s)
- Stefan Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Paige Whyte-Fagundes
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Lian Gelis
- Department of Cell Physiology, Ruhr University Bochum Bochum, Germany
| | - Sarah Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Emerson Brazil
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Christiane Zoidl
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Hanns Hatt
- Department of Cell Physiology, Ruhr University Bochum Bochum, Germany
| | - Valery I Shestopalov
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miller School of Medicine, University of Miami Miami, FL, USA ; Vavilov Institute of General Genetics, Russian Academy of Sciences Moscow, Russia
| | - Georg Zoidl
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| |
Collapse
|
23
|
Kwon TJ, Kim DB, Bae JW, Sagong B, Choi SY, Cho HJ, Kim UK, Lee KY. Molecular cloning, characterization, and expression of pannexin genes in chicken. Poult Sci 2014; 93:2253-61. [PMID: 25002553 DOI: 10.3382/ps.2013-03867] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pannexins (Panx) are a family of proteins that share sequences with the invertebrate gap junction proteins, innexins, and have a similar structure to that of the vertebrate gap junction proteins, connexins. To date, the Panx family consists of 3 members, but their genetic sequences have only been completely determined in a few vertebrate species. Moreover, expression of the Panx family has been reported in several rodent tissues: Panx1 is ubiquitously expressed in mammals, whereas Panx2 and Panx3 expressions are more restricted. Although members of the Panx family have been detected in mammals, their genetic sequences in avian species have not yet been fully elucidated. Here, we obtained the full-length mRNA sequences of chicken PANX genes and evaluated the homology of the amino acids from these sequences with those of other species. Furthermore, PANX gene expression in several chicken tissues was investigated based on mRNA levels. PANX1 was detected in the brain, cochlea, chondrocytes, eye, lung, skin, and intestine, and PANX2 was expressed in the brain, eye, and intestine. PANX3 was observed in the cochlea, chondrocytes, and bone. In addition, expression of PANX3 was higher than PANX1 in the cochlea. Immunofluorescent staining revealed PANX1 in hair cells, as well as the supporting cells, ganglion neurons, and the tegmentum vasculosum in chickens, whereas PANX3 was only detected in the bone surrounding the cochlea. Overall, the results of this study provide the first identification and characterization of the sequence and expression of the PANX family in an avian species, and fundamental data for confirmation of Panx function.
Collapse
Affiliation(s)
- Tae-Jun Kwon
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Dong-Bin Kim
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Jae Woong Bae
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Borum Sagong
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Soo-Young Choi
- Department of Medicine, University of Pennsylvania, Philadelphia 19104-4539
| | - Hyun-Ju Cho
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Un-Kyung Kim
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Kyu-Yup Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medicine, Kyungpook National University, Daegu, 700-721, South Korea
| |
Collapse
|
24
|
Velasquez S, Eugenin EA. Role of Pannexin-1 hemichannels and purinergic receptors in the pathogenesis of human diseases. Front Physiol 2014; 5:96. [PMID: 24672487 PMCID: PMC3953678 DOI: 10.3389/fphys.2014.00096] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 02/24/2014] [Indexed: 12/20/2022] Open
Abstract
In the last decade several groups have determined the key role of hemichannels formed by pannexins or connexins, extracellular ATP and purinergic receptors in physiological and pathological conditions. Our work and the work of others, indicate that the opening of Pannexin-1 hemichannels and activation of purinergic receptors by extracellular ATP is essential for HIV infection, cellular migration, inflammation, atherosclerosis, stroke, and apoptosis. Thus, this review discusses the importance of purinergic receptors, Panx-1 hemichannels and extracellular ATP in the pathogenesis of several human diseases and their potential use to design novel therapeutic approaches.
Collapse
Affiliation(s)
- Stephani Velasquez
- Public Health Research Institute, Rutgers the State University of New Jersey Newark, NJ, USA ; Department of Microbiology and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey Newark, NJ, USA
| | - Eliseo A Eugenin
- Public Health Research Institute, Rutgers the State University of New Jersey Newark, NJ, USA ; Department of Microbiology and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey Newark, NJ, USA
| |
Collapse
|
25
|
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
|
26
|
Ishikawa M, Iwamoto T, Fukumoto S, Yamada Y. Pannexin 3 inhibits proliferation of osteoprogenitor cells by regulating Wnt and p21 signaling. J Biol Chem 2013; 289:2839-51. [PMID: 24338011 DOI: 10.1074/jbc.m113.523241] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Canonical Wnt signaling and BMP promote the proliferation and differentiation of osteoprogenitors, respectively. However, the regulatory mechanism involved in the transition from proliferation to differentiation is unclear. Here, we show that Panx3 (pannexin 3) plays a key role in this transition by inhibiting the proliferation and promoting the cell cycle exit. Using primary calvarial cells and explants, C3H10T1/2 cells, and C2C12 cells, we found that Panx3 expression inhibited cell growth, whereas the inhibition of endogenous Panx3 expression increased it. We also found that the Panx3 hemichannel inhibited cell growth by promoting β-catenin degradation through GSK3β activation. Additionally, the Panx3 hemichannel inhibited cyclin D1 transcription and Rb phosphorylation through reduced cAMP/PKA/CREB signaling. Furthermore, the Panx3 endoplasmic reticulum Ca(2+) channel induced the transcription and phosphorylation of p21, through the calmodulin/Smad pathway, and resulted in the cell cycle exit. Our results reveal that Panx3 is a new regulator that promotes the switch from proliferation to differentiation of osteoprogenitors via multiple Panx3 signaling pathways.
Collapse
Affiliation(s)
- Masaki Ishikawa
- From the Laboratory of Cell and Developmental Biology, NIDCR, National Institutes of Health, Bethesda, Maryland 20892-4370
| | | | | | | |
Collapse
|
27
|
Courtemanche R, Robinson JC, Aponte DI. Linking oscillations in cerebellar circuits. Front Neural Circuits 2013; 7:125. [PMID: 23908606 PMCID: PMC3725427 DOI: 10.3389/fncir.2013.00125] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 07/11/2013] [Indexed: 11/13/2022] Open
Abstract
In many neuroscience fields, the study of local and global rhythmicity has been receiving increasing attention. These network influences could directly impact on how neuronal groups interact together, organizing for different contexts. The cerebellar cortex harbors a variety of such local circuit rhythms, from the rhythms in the cerebellar cortex per se, or those dictated from important afferents. We present here certain cerebellar oscillatory phenomena that have been recorded in rodents and primates. Those take place in a range of frequencies: from the more known oscillations in the 4-25 Hz band, such as the olivocerebellar oscillatory activity and the granule cell layer oscillations, to the more recently reported slow (<1 Hz oscillations), and the fast (>150 Hz) activity in the Purkinje cell layer. Many of these oscillations appear spontaneously in the circuits, and are modulated by behavioral imperatives. We review here how those oscillations are recorded, some of their modulatory mechanisms, and also identify some of the cerebellar nodes where they could interact. A particular emphasis has been placed on how these oscillations could be modulated by movement and certain neuropathological manifestations. Many of those oscillations could have a definite impact on the way information is processed in the cerebellum and how it interacts with other structures in a variety of contexts.
Collapse
Affiliation(s)
- Richard Courtemanche
- Department of Exercise Science, Groupe de Recherche en Neurobiologie Comportementale/Center for Studies in Behavioral Neurobiology, Concordia UniversityMontréal, QC, Canada
| | | | | |
Collapse
|
28
|
Lutz SE, González-Fernández E, Ventura JCC, Pérez-Samartín A, Tarassishin L, Negoro H, Patel NK, Suadicani SO, Lee SC, Matute C, Scemes E. Contribution of pannexin1 to experimental autoimmune encephalomyelitis. PLoS One 2013; 8:e66657. [PMID: 23885286 PMCID: PMC3688586 DOI: 10.1371/journal.pone.0066657] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 05/08/2013] [Indexed: 11/19/2022] Open
Abstract
Pannexin1 (Panx1) is a plasma membrane channel permeable to relatively large molecules, such as ATP. In the central nervous system (CNS) Panx1 is found in neurons and glia and in the immune system in macrophages and T-cells. We tested the hypothesis that Panx1-mediated ATP release contributes to expression of Experimental Autoimmune Encephalomyelitis (EAE), an animal model for multiple sclerosis, using wild-type (WT) and Panx1 knockout (KO) mice. Panx1 KO mice displayed a delayed onset of clinical signs of EAE and decreased mortality compared to WT mice, but developed as severe symptoms as the surviving WT mice. Spinal cord inflammatory lesions were also reduced in Panx1 KO EAE mice during acute disease. Additionally, pharmacologic inhibition of Panx1 channels with mefloquine (MFQ) reduced severity of acute and chronic EAE when administered before or after onset of clinical signs. ATP release and YoPro uptake were significantly increased in WT mice with EAE as compared to WT non-EAE and reduced in tissues of EAE Panx1 KO mice. Interestingly, we found that the P2X7 receptor was upregulated in the chronic phase of EAE in both WT and Panx1 KO spinal cords. Such increase in receptor expression is likely to counterbalance the decrease in ATP release recorded from Panx1 KO mice and thus contribute to the development of EAE symptoms in these mice. The present study shows that a Panx1 dependent mechanism (ATP release and/or inflammasome activation) contributes to disease progression, and that inhibition of Panx1 using pharmacology or gene disruption delays and attenuates clinical signs of EAE.
Collapse
Affiliation(s)
- Sarah E. Lutz
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | | | | | | | - Leonid Tarassishin
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York United States of America
| | - Hiromitsu Negoro
- Department of Urology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Naman K. Patel
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Sylvia O. Suadicani
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Urology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Sunhee C. Lee
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York United States of America
| | - Carlos Matute
- Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - Eliana Scemes
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
29
|
Cone AC, Ambrosi C, Scemes E, Martone ME, Sosinsky GE. A comparative antibody analysis of pannexin1 expression in four rat brain regions reveals varying subcellular localizations. Front Pharmacol 2013; 4:6. [PMID: 23390418 PMCID: PMC3565217 DOI: 10.3389/fphar.2013.00006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 01/09/2013] [Indexed: 11/13/2022] Open
Abstract
Pannexin1 (Panx1) channels release cytosolic ATP in response to signaling pathways. Panx1 is highly expressed in the central nervous system. We used four antibodies with different Panx1 anti-peptide epitopes to analyze four regions of rat brain. These antibodies labeled the same bands in Western blots and had highly similar patterns of immunofluorescence in tissue culture cells expressing Panx1, but Western blots of brain lysates from Panx1 knockout and control mice showed different banding patterns. Localizations of Panx1 in brain slices were generated using automated wide field mosaic confocal microscopy for imaging large regions of interest while retaining maximum resolution for examining cell populations and compartments. We compared Panx1 expression over the cerebellum, hippocampus with adjacent cortex, thalamus, and olfactory bulb. While Panx1 localizes to the same neuronal cell types, subcellular localizations differ. Two antibodies with epitopes against the intracellular loop and one against the carboxy terminus preferentially labeled cell bodies, while an antibody raised against an N-terminal peptide highlighted neuronal processes more than cell bodies. These labeling patterns may be a reflection of different cellular and subcellular localizations of full-length and/or modified Panx1 channels where each antibody is highlighting unique or differentially accessible Panx1 populations. However, we cannot rule out that one or more of these antibodies have specificity issues. All data associated with experiments from these four antibodies are presented in a manner that allows them to be compared and our claims thoroughly evaluated, rather than eliminating results that were questionable. Each antibody is given a unique identifier through the NIF Antibody Registry that can be used to track usage of individual antibodies across papers and all image and metadata are made available in the public repository, the Cell Centered Database, for on-line viewing, and download.
Collapse
Affiliation(s)
- Angela C Cone
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | |
Collapse
|
30
|
Lohman AW, Weaver JL, Billaud M, Sandilos JK, Griffiths R, Straub AC, Penuela S, Leitinger N, Laird DW, Bayliss DA, Isakson BE. S-nitrosylation inhibits pannexin 1 channel function. J Biol Chem 2012; 287:39602-12. [PMID: 23033481 DOI: 10.1074/jbc.m112.397976] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
S-nitrosylation is a post-translational modification on cysteine(s) that can regulate protein function, and pannexin 1 (Panx1) channels are present in the vasculature, a tissue rich in nitric oxide (NO) species. Therefore, we investigated whether Panx1 can be S-nitrosylated and whether this modification can affect channel activity. Using the biotin switch assay, we found that application of the NO donor S-nitrosoglutathione (GSNO) or diethylammonium (Z)-1-1(N,N-diethylamino)diazen-1-ium-1,2-diolate (DEA NONOate) to human embryonic kidney (HEK) 293T cells expressing wild type (WT) Panx1 and mouse aortic endothelial cells induced Panx1 S-nitrosylation. Functionally, GSNO and DEA NONOate attenuated Panx1 currents; consistent with a role for S-nitrosylation, current inhibition was reversed by the reducing agent dithiothreitol and unaffected by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, a blocker of guanylate cyclase activity. In addition, ATP release was significantly inhibited by treatment with both NO donors. To identify which cysteine residue(s) was S-nitrosylated, we made single cysteine-to-alanine substitutions in Panx1 (Panx1(C40A), Panx1(C346A), and Panx1(C426A)). Mutation of these single cysteines did not prevent Panx1 S-nitrosylation; however, mutation of either Cys-40 or Cys-346 prevented Panx1 current inhibition and ATP release by GSNO. This observation suggested that multiple cysteines may be S-nitrosylated to regulate Panx1 channel function. Indeed, we found that mutation of both Cys-40 and Cys-346 (Panx1(C40A/C346A)) prevented Panx1 S-nitrosylation by GSNO as well as the GSNO-mediated inhibition of Panx1 current and ATP release. Taken together, these results indicate that S-nitrosylation of Panx1 at Cys-40 and Cys-346 inhibits Panx1 channel currents and ATP release.
Collapse
Affiliation(s)
- Alexander W Lohman
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Cea LA, Riquelme MA, Cisterna BA, Puebla C, Vega JL, Rovegno M, Sáez JC. Connexin- and pannexin-based channels in normal skeletal muscles and their possible role in muscle atrophy. J Membr Biol 2012; 245:423-36. [PMID: 22850938 DOI: 10.1007/s00232-012-9485-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 06/28/2012] [Indexed: 12/13/2022]
Abstract
Precursor cells of skeletal muscles express connexins 39, 43 and 45 and pannexin1. In these cells, most connexins form two types of membrane channels, gap junction channels and hemichannels, whereas pannexin1 forms only hemichannels. All these channels are low-resistance pathways permeable to ions and small molecules that coordinate developmental events. During late stages of skeletal muscle differentiation, myofibers become innervated and stop expressing connexins but still express pannexin1 hemichannels that are potential pathways for the ATP release required for potentiation of the contraction response. Adult injured muscles undergo regeneration, and connexins are reexpressed and form membrane channels. In vivo, connexin reexpression occurs in undifferentiated cells that form new myofibers, favoring the healing process of injured muscle. However, differentiated myofibers maintained in culture for 48 h or treated with proinflammatory cytokines for less than 3 h also reexpress connexins and only form functional hemichannels at the cell surface. We propose that opening of these hemichannels contributes to drastic changes in electrochemical gradients, including reduction of membrane potential, increases in intracellular free Ca(2+) concentration and release of diverse metabolites (e.g., NAD(+) and ATP) to the extracellular milieu, contributing to multiple metabolic and physiologic alterations that characterize muscles undergoing atrophy in several acquired and genetic human diseases. Consequently, inhibition of connexin hemichannels expressed by injured or denervated skeletal muscles might reduce or prevent deleterious changes triggered by conditions that promote muscle atrophy.
Collapse
Affiliation(s)
- Luis A Cea
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile,
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Pannexins are a recently discovered protein family with the isoform Panx1 ubiquitously expressed and therefore extensively studied. Panx1 proteins form membrane channels known to release purines such as ATP. Because ATP and, more generally, purinergic signaling plays an important role in the vasculature, it became evident that Panx1 could have a key role in vascular functions. This article reviews recent findings on the pivotal role of Panx1 in smooth muscle cells in the contraction of arteries as well as recent insights into Panx1 channel regulation.
Collapse
Affiliation(s)
- Marie Billaud
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
33
|
Iglesias RM, Spray DC. Pannexin1-mediated ATP release provides signal transmission between Neuro2A cells. Neurochem Res 2012; 37:1355-63. [PMID: 22359052 DOI: 10.1007/s11064-012-0720-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 01/13/2012] [Accepted: 01/28/2012] [Indexed: 01/22/2023]
Abstract
Pannexin1 (Panx1), a protein related to the gap junction proteins of invertebrates, forms nonjunctional channels that open upon depolarization and in response to mechanical stretch and purinergic receptor stimulation. Importantly, ATP can be released through Panx1 channels, providing a possible role for these channels in non-vesicular signal transmission. In this study we expressed exogenous human and mouse Panx1 in the gap junction deficient Neuro2A neuroblastoma cell line and explored the contribution of Panx1 channels to cell-cell communication as sites of ATP release. Electrophysiological (patch clamp) recordings from Panx1 transfected Neuro2A cells revealed membrane conductance that increased beyond 0 mV when applying voltage ramps from -60 to +100 mV; threshold was correlated with extracellular K+, so that at 10 mM K+, channels began to open at -30 mV. Evaluation of cell-cell communication using dual whole cell recordings from cell pairs revealed that activation of Panx1 current in one cell of the pair induced an inward current in the second cell after a latency of 10-20 s. This paracrine response was amplified by an ATPase inhibitor (ARL67156, 100 μM) and was blocked by the ATP-degrading enzyme apyrase (6.7 U/ml), by the P2 receptor antagonist suramin (50 μM) and by the Panx1 channel blocker carbenoxolone. These results provide additional evidence that ATP release through Panx1 channels can mediate nonsynaptic bidirectional intercellular communication. Furthermore, current potentiation by elevated K+ provides a mechanism for enhancement of ATP release under pathological conditions.
Collapse
Affiliation(s)
- Rodolfo M Iglesias
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, K 840, Bronx, NY 10461, USA
| | | |
Collapse
|
34
|
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: 294] [Impact Index Per Article: 22.6] [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
|
35
|
Prochnow N, Hoffmann S, Dermietzel R, Zoidl G. Replacement of a single cysteine in the fourth transmembrane region of zebrafish pannexin 1 alters hemichannel gating behavior. Exp Brain Res 2012; 199:255-64. [PMID: 19701745 DOI: 10.1007/s00221-009-1957-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 07/18/2009] [Indexed: 01/09/2023]
Abstract
Pannexin1 (Panx1) is a novel candidate for an electrical synapse protein in the retina. At present Panx1 is considered to function as a hemichannel. Since information about the gating properties of Panx1 channels to date rely on blocker pharmacology, we have begun to establish a structural context of channel function starting with site directed mutagenesis of cysteine residues in transmembrane domains of Panx1. Dye uptake and whole cell voltage clamp recordings of transfected N2a cells demonstrate that zfPanx1 forms voltage activated hemichannels with a large unitary conductance in vitro. The function of this channel was significantly reduced following mutation of a single cysteine residue (C282W) in the fourth transmembrane region. This result suggests a role of this domain in gating of the Panx1 hemichannel.
Collapse
Affiliation(s)
- Nora Prochnow
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum 44780, Germany.
| | | | | | | |
Collapse
|
36
|
Sosinsky GE, Boassa D, Dermietzel R, Duffy HS, Laird DW, MacVicar B, Naus CC, Penuela S, Scemes E, Spray DC, Thompson RJ, Zhao HB, Dahl G. Pannexin channels are not gap junction hemichannels. Channels (Austin) 2011; 5:193-7. [PMID: 21532340 DOI: 10.4161/chan.5.3.15765] [Citation(s) in RCA: 276] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Pannexins, a class of membrane channels, bear significant sequence homology with the invertebrate gap junction proteins, innexins and more distant similarities in their membrane topologies and pharmacological sensitivities with the gap junction proteins, connexins. However, the functional role for the pannexin oligomers, or pannexons, is different from connexin oligomers, the connexons. Many pannexin publications have used the term "hemichannels" to describe pannexin oligomers while others use the term "channels" instead. This has led to confusion within the literature about the function of pannexins that promotes the idea that pannexons serve as gap junction hemichannels and thus have an assembly and functional state as gap junctional intercellular channels. Here we present the case that unlike the connexin gap junction intercellular channels, so far, pannexin oligomers have repeatedly been shown to be channels that are functional in single membranes, but not as intercellular channel in appositional membranes. Hence, they should be referred to as channels and not hemichannels. Thus, we advocate that in the absence of firm evidence that pannexins form gap junctions, the use of the term "hemichannel" be discontinued within the pannexin literature.
Collapse
Affiliation(s)
- Gina E Sosinsky
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
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
|
38
|
Simões de Souza FM, De Schutter E. Robustness effect of gap junctions between Golgi cells on cerebellar cortex oscillations. NEURAL SYSTEMS & CIRCUITS 2011; 1:7. [PMID: 22330240 PMCID: PMC3278348 DOI: 10.1186/2042-1001-1-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Accepted: 12/20/2010] [Indexed: 11/25/2022]
Abstract
Background Previous one-dimensional network modeling of the cerebellar granular layer has been successfully linked with a range of cerebellar cortex oscillations observed in vivo. However, the recent discovery of gap junctions between Golgi cells (GoCs), which may cause oscillations by themselves, has raised the question of how gap-junction coupling affects GoC and granular-layer oscillations. To investigate this question, we developed a novel two-dimensional computational model of the GoC-granule cell (GC) circuit with and without gap junctions between GoCs. Results Isolated GoCs coupled by gap junctions had a strong tendency to generate spontaneous oscillations without affecting their mean firing frequencies in response to distributed mossy fiber input. Conversely, when GoCs were synaptically connected in the granular layer, gap junctions increased the power of the oscillations, but the oscillations were primarily driven by the synaptic feedback loop between GoCs and GCs, and the gap junctions did not change oscillation frequency or the mean firing rate of either GoCs or GCs. Conclusion Our modeling results suggest that gap junctions between GoCs increase the robustness of cerebellar cortex oscillations that are primarily driven by the feedback loop between GoCs and GCs. The robustness effect of gap junctions on synaptically driven oscillations observed in our model may be a general mechanism, also present in other regions of the brain.
Collapse
Affiliation(s)
- Fabio M Simões de Souza
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology, Okinawa 904-0411, Japan.
| | | |
Collapse
|
39
|
Turmel P, Dufresne J, Hermo L, Smith CE, Penuela S, Laird DW, Cyr DG. Characterization of pannexin1 and pannexin3 and their regulation by androgens in the male reproductive tract of the adult rat. Mol Reprod Dev 2011; 78:124-38. [DOI: 10.1002/mrd.21280] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
40
|
Fontainhas AM, Wang M, Liang KJ, Chen S, Mettu P, Damani M, Fariss RN, Li W, Wong WT. Microglial morphology and dynamic behavior is regulated by ionotropic glutamatergic and GABAergic neurotransmission. PLoS One 2011; 6:e15973. [PMID: 21283568 PMCID: PMC3026789 DOI: 10.1371/journal.pone.0015973] [Citation(s) in RCA: 243] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 12/01/2010] [Indexed: 02/01/2023] Open
Abstract
PURPOSE Microglia represent the primary resident immune cells in the CNS, and have been implicated in the pathology of neurodegenerative diseases. Under basal or "resting" conditions, microglia possess ramified morphologies and exhibit dynamic surveying movements in their processes. Despite the prominence of this phenomenon, the function and regulation of microglial morphology and dynamic behavior are incompletely understood. We investigate here whether and how neurotransmission regulates "resting" microglial morphology and behavior. METHODS We employed an ex vivo mouse retinal explant system in which endogenous neurotransmission and dynamic microglial behavior are present. We utilized live-cell time-lapse confocal imaging to study the morphology and behavior of GFP-labeled retinal microglia in response to neurotransmitter agonists and antagonists. Patch clamp electrophysiology and immunohistochemical localization of glutamate receptors were also used to investigate direct-versus-indirect effects of neurotransmission by microglia. RESULTS Retinal microglial morphology and dynamic behavior were not cell-autonomously regulated but are instead modulated by endogenous neurotransmission. Morphological parameters and process motility were differentially regulated by different modes of neurotransmission and were increased by ionotropic glutamatergic neurotransmission and decreased by ionotropic GABAergic neurotransmission. These neurotransmitter influences on retinal microglia were however unlikely to be directly mediated; local applications of neurotransmitters were unable to elicit electrical responses on microglia patch-clamp recordings and ionotropic glutamatergic receptors were not located on microglial cell bodies or processes by immunofluorescent labeling. Instead, these influences were mediated indirectly via extracellular ATP, released in response to glutamatergic neurotransmission through probenecid-sensitive pannexin hemichannels. CONCLUSIONS Our results demonstrate that neurotransmission plays an endogenous role in regulating the morphology and behavior of "resting" microglia in the retina. These findings illustrate a mode of constitutive signaling between the neural and immune compartments of the CNS through which immune cells may be regulated in concert with levels of neural activity.
Collapse
Affiliation(s)
- Aurora M. Fontainhas
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Minhua Wang
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Katharine J. Liang
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Shan Chen
- Unit on Retinal Neurophysiology, National Eye Institute, Bethesda, Maryland, United States of America
| | - Pradeep Mettu
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mausam Damani
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert N. Fariss
- Biological Imaging Core, Office of the Scientific Director, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wei Li
- Unit on Retinal Neurophysiology, National Eye Institute, Bethesda, Maryland, United States of America
| | - Wai T. Wong
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
41
|
Pannexin channels in ATP release and beyond: an unexpected rendezvous at the endoplasmic reticulum. Cell Signal 2010; 23:305-16. [PMID: 20688156 DOI: 10.1016/j.cellsig.2010.07.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Accepted: 07/22/2010] [Indexed: 01/13/2023]
Abstract
The pannexin (Panx) family of proteins, which is co-expressed with connexins (Cxs) in vertebrates, was found to be a new GJ-forming protein family related to invertebrate innexins. During the past ten years, different studies showed that Panxs mainly form hemichannels in the plasma membrane and mediate paracrine signalling by providing a flux pathway for ions such as Ca²(+), for ATP and perhaps for other compounds, in response to physiological and pathological stimuli. Although the physiological role of Panxs as a hemichannel was questioned, there is increasing evidence that Panx play a role in vasodilatation, initiation of inflammatory responses, ischemic death of neurons, epilepsy and in tumor suppression. Moreover, it is intriguing that Panxs may also function at the endoplasmic reticulum (ER) as intracellular Ca²(+)-leak channel and may be involved in ER-related functions. Although the physiological significance and meaning of such Panx-regulated intracellular Ca²(+) leak requires further exploration, this functional property places Panx at the centre of many physiological and pathophysiological processes, given the fundamental role of intracellular Ca²(+) homeostasis and dynamics in a plethora of physiological processes. In this review, we therefore want to focus on Panx as channels at the plasma membrane and at the ER membranes with a particular emphasis on the potential implications of the latter in intracellular Ca²(+) signalling.
Collapse
|
42
|
Swayne LA, Sorbara CD, Bennett SAL. Pannexin 2 is expressed by postnatal hippocampal neural progenitors and modulates neuronal commitment. J Biol Chem 2010; 285:24977-86. [PMID: 20529862 DOI: 10.1074/jbc.m110.130054] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The pannexins (Panx1, -2, and -3) are a mammalian family of putative single membrane channels discovered through homology to invertebrate gap junction-forming proteins, the innexins. Because connexin gap junction proteins are known regulators of neural stem and progenitor cell proliferation, migration, and specification, we asked whether pannexins, specifically Panx2, play a similar role in the postnatal hippocampus. We show that Panx2 protein is differentially expressed by multipotential progenitor cells and mature neurons. Both in vivo and in vitro, Type I and IIa stem-like neural progenitor cells express an S-palmitoylated Panx2 species localizing to Golgi and endoplasmic reticulum membranes. Protein expression is down-regulated during neurogenesis in neuronally committed Type IIb and III progenitor cells and immature neurons. Panx2 is re-expressed by neurons following maturation. Protein expressed by mature neurons is not palmitoylated and localizes to the plasma membrane. To assess the impact of Panx2 on neuronal differentiation, we used short hairpin RNA to suppress Panx2 expression in Neuro2a cells. Knockdown significantly accelerated the rate of neuronal differentiation. Neuritic extension and the expression of antigenic markers of mature neurons occurred earlier in stable lines expressing Panx2 short hairpin RNA than in controls. Together, these findings describe an endogenous post-translational regulation of Panx2, specific to early neural progenitor cells, and demonstrate that this expression plays a role in modulating the timing of their commitment to a neuronal lineage.
Collapse
Affiliation(s)
- Leigh Anne Swayne
- Department of Biochemistry, Neural Regeneration Laboratory and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1M 1E5, Canada.
| | | | | |
Collapse
|
43
|
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
|
44
|
Bunse S, Locovei S, Schmidt M, Qiu F, Zoidl G, Dahl G, Dermietzel R. The potassium channel subunit Kvbeta3 interacts with pannexin 1 and attenuates its sensitivity to changes in redox potentials. FEBS J 2009; 276:6258-70. [PMID: 19780818 DOI: 10.1111/j.1742-4658.2009.07334.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pannexin 1 (Panx1), a member of the second gap junction protein family identified in vertebrates, appears to preferentially form non-junctional membrane channels. A candidate regulatory protein of Panx1 is the potassium channel subunit Kvbeta3, previously identified by bacterial two-hybrid strategies. Here, we report on the physical association of Panx1 with Kvbeta3 by immunoprecipitation when co-expressed in a neuroblastoma cell line (Neuro2A). Furthermore, in vivo co-expression of Panx1 and Kvbeta3 was shown to occur in murine hippocampus and cerebellum. Kvbeta3 is known to accelerate inactivation of otherwise slowly inactivating potassium channels under reducing conditions. We subsequently found that Panx1 channel currents exhibit a significant reduction when exposed to reducing agents, and that this effect is attenuated in the presence of Kvbeta3. Apparently, Kvbeta3 is involved in regulating the susceptibility of Panx1 channels to redox potential. Furthermore, the Panx1 channel blockers carbenoxolone and Probenecid were less effective in inhibiting Panx1 currents when Kvbeta3 was co-expressed. The influence of Kvbeta3 on Panx1 is the first example of modulation of Panx1 channel function(s) by interacting proteins, and suggests the physiological importance of sensing changes in redox potentials.
Collapse
Affiliation(s)
- Stefanie Bunse
- Department of Neuroanatomy & Molecular Brain Research, Ruhr University, Bochum, Germany
| | | | | | | | | | | | | |
Collapse
|
45
|
Lai CPK, Bechberger JF, Naus CC. Pannexin2 as a novel growth regulator in C6 glioma cells. Oncogene 2009; 28:4402-8. [DOI: 10.1038/onc.2009.283] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Penuela S, Bhalla R, Nag K, Laird DW. Glycosylation regulates pannexin intermixing and cellular localization. Mol Biol Cell 2009; 20:4313-23. [PMID: 19692571 DOI: 10.1091/mbc.e09-01-0067] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The pannexin family of mammalian proteins, composed of Panx1, Panx2, and Panx3, has been postulated to be a new class of single-membrane channels with functional similarities to connexin gap junction proteins. In this study, immunolabeling and coimmunoprecipitation assays revealed that Panx1 can interact with Panx2 and to a lesser extent, with Panx3 in a glycosylation-dependent manner. Panx2 strongly interacts with the core and high-mannose species of Panx1 but not with Panx3. Biotinylation and dye uptake assays indicated that all three pannexins, as well as the N-glycosylation-defective mutants of Panx1 and Panx3, can traffic to the cell surface and form functional single-membrane channels. Interestingly, Panx2, which is also a glycoprotein and seems to only be glycosylated to a high-mannose form, is more abundant in intracellular compartments, except when coexpressed with Panx1, when its cell surface distribution increases by twofold. Functional assays indicated that the combination of Panx1 and Panx2 results in compromised channel function, whereas coexpressing Panx1 and Panx3 does not affect the incidence of dye uptake in 293T cells. Collectively, these results reveal that the functional state and cellular distribution of mouse pannexins are regulated by their glycosylation status and interactions among pannexin family members.
Collapse
Affiliation(s)
- Silvia Penuela
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | | | | | |
Collapse
|
47
|
Abstract
Purinergic signaling plays distinct and important roles in the CNS, including the transmission of calcium signals between astrocytes. Gap junction hemichannels are among the mechanisms proposed by which astrocytes might release ATP; however, whether the gap junction protein connexin43 (Cx43) forms these "hemichannels" remains controversial. Recently, a new group of proteins, the pannexins, have been shown to form nonselective, high-conductance plasmalemmal channels permeable to ATP, thereby offering an alternative for the hemichannel protein. Here, we provide strong evidence that, in cultured astrocytes, pannexin1 (Panx1) but not Cx43 forms hemichannels. Electrophysiological and fluorescence microscope recordings performed in wild-type and Cx43-null astrocytes did not reveal any differences in hemichannel activity, which was mostly eliminated by treating Cx43-null astrocytes with Panx1-short interfering RNA [Panx1-knockdown (Panx1-KD)]. Moreover, quantification of the amount of ATP released from wild-type, Cx43-null, and Panx1-KD astrocytes indicates that downregulation of Panx1, but not of Cx43, prevented ATP release from these cells.
Collapse
|
48
|
The cerebellum, cerebellar disorders, and cerebellar research--two centuries of discoveries. THE CEREBELLUM 2009; 7:505-16. [PMID: 18855093 DOI: 10.1007/s12311-008-0063-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Research on the cerebellum is evolving rapidly. The exquisiteness of the cerebellar circuitry with a unique geometric arrangement has fascinated researchers from numerous disciplines. The painstaking works of pioneers of these last two centuries, such as Rolando, Flourens, Luciani, Babinski, Holmes, Cajal, Larsell, or Eccles, still exert a strong influence in the way we approach cerebellar functions. Advances in genetic studies, detailed molecular and cellular analyses, profusion of brain imaging techniques, emergence of behavioral assessments, and reshaping of models of cerebellar function are generating an immense amount of knowledge. Simultaneously, a better definition of cerebellar disorders encountered in the clinic is emerging. The essentials of a trans-disciplinary blending are expanding. The analysis of the literature published these last two decades indicates that the gaps between domains of research are vanishing. The launch of the society for research on the cerebellum (SRC) illustrates how cerebellar research is burgeoning. This special issue gathers the contributions of the inaugural conference of the SRC dedicated to the mechanisms of cerebellar function. Contributions were brought together around five themes: (1) cerebellar development, death, and regeneration; (2) cerebellar circuitry: processing and function; (3) mechanisms of cerebellar plasticity and learning; (4) cerebellar function: timing, prediction, and/or coordination?; (5) anatomical and disease perspectives on cerebellar function.
Collapse
|
49
|
Dugué GP, Brunel N, Hakim V, Schwartz E, Chat M, Lévesque M, Courtemanche R, Léna C, Dieudonné S. Electrical coupling mediates tunable low-frequency oscillations and resonance in the cerebellar Golgi cell network. Neuron 2009; 61:126-39. [PMID: 19146818 DOI: 10.1016/j.neuron.2008.11.028] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 08/01/2008] [Accepted: 11/06/2008] [Indexed: 10/21/2022]
Abstract
Tonic motor control involves oscillatory synchronization of activity at low frequency (5-30 Hz) throughout the sensorimotor system, including cerebellar areas. We investigated the mechanisms underpinning cerebellar oscillations. We found that Golgi interneurons, which gate information transfer in the cerebellar cortex input layer, are extensively coupled through electrical synapses. When depolarized in vitro, these neurons displayed low-frequency oscillatory synchronization, imposing rhythmic inhibition onto granule cells. Combining experiments and modeling, we show that electrical transmission of the spike afterhyperpolarization is the essential component for oscillatory population synchronization. Rhythmic firing arises in spite of strong heterogeneities, is frequency tuned by the mean excitatory input to Golgi cells, and displays pronounced resonance when the modeled network is driven by oscillating inputs. In vivo, unitary Golgi cell activity was found to synchronize with low-frequency LFP oscillations occurring during quiet waking. These results suggest a major role for Golgi cells in coordinating cerebellar sensorimotor integration during oscillatory interactions.
Collapse
|
50
|
Wang XH, Streeter M, Liu YP, Zhao HB. Identification and characterization of pannexin expression in the mammalian cochlea. J Comp Neurol 2009; 512:336-46. [PMID: 19009624 DOI: 10.1002/cne.21898] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The gap junction in vertebrates is encoded by the connexin gene family. Recently, a new gene family termed pannexin (Panx) has been identified in vertebrates and found to encode gap junctional proteins as well. To date, three pannexin isoforms (Panx1, 2, and 3) have been cloned from mouse and human genomes. In this study, expression of pannexins in the mouse and rat cochlea was investigated. Polymerase chain reaction and Western blot analysis showed that all three pannexin isoforms were expressed in the cochlea. Immunofluorescent staining showed that Panx1 expression was extensive. In the organ of Corti, Panx1 labeling was found in supporting cells, including pillar cells, Hensen cells, Claudius cells, and Boettcher cells. Both surface plaque-like punctate labeling and diffuse-cytoplasmic labeling were visible. However, the labeling was weak and rare in Deiters cells. No labeling was found in the hair cells. Intense labeling for Panx1 was also observed in the interdental cells in the spiral limbus, the inner and outer sulcus cells, and the type II fibrocytes in the spiral prominence and central region in the cochlear lateral wall. In addition, Panx1 labeling was detectable in Reissner's membrane and strial blood vessel cells. Panx2 labeling was restricted to the basal cells in the stria vascularis and was also detectable in the spiral ganglion neurons. However, no overlapping labeling for Panx1 and Panx2 was observed. Finally, Panx3 labeling was exclusively observed in the cochlear bone. Thus, Panx1, 2, and 3 are abundantly expressed in the mammalian cochlea and demonstrate distinct cellular distributions. Like connexins, they may play an important role in hearing.
Collapse
Affiliation(s)
- Xiao-Hui Wang
- Department of Surgery-Otolaryngology, University of Kentucky Medical Center, Lexington, Kentucky 40536, USA
| | | | | | | |
Collapse
|