1
|
Vitureira N, Rafael A, Abudara V. P2X7 receptors and pannexin1 hemichannels shape presynaptic transmission. Purinergic Signal 2024; 20:223-236. [PMID: 37713157 PMCID: PMC11189373 DOI: 10.1007/s11302-023-09965-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023] Open
Abstract
Over the last decades, since the discovery of ATP as a transmitter, accumulating evidence has been reported about the role of this nucleotide and purinergic receptors, in particular P2X7 receptors, in the modulation of synaptic strength and plasticity. Purinergic signaling has emerged as a crucial player in orchestrating the molecular interaction between the components of the tripartite synapse, and much progress has been made in how this neuron-glia interaction impacts neuronal physiology under basal and pathological conditions. On the other hand, pannexin1 hemichannels, which are functionally linked to P2X7 receptors, have appeared more recently as important modulators of excitatory synaptic function and plasticity under diverse contexts. In this review, we will discuss the contribution of ATP, P2X7 receptors, and pannexin hemichannels to the modulation of presynaptic strength and its impact on motor function, sensory processing, synaptic plasticity, and neuroglial communication, with special focus on the P2X7 receptor/pannexin hemichannel interplay. We also address major hypotheses about the role of this interaction in physiological and pathological circumstances.
Collapse
Affiliation(s)
- Nathalia Vitureira
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - Alberto Rafael
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Verónica Abudara
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
2
|
Qiu X, Yang Y, Da X, Wang Y, Chen Z, Xu C. Satellite glial cells in sensory ganglia play a wider role in chronic pain via multiple mechanisms. Neural Regen Res 2024; 19:1056-1063. [PMID: 37862208 PMCID: PMC10749601 DOI: 10.4103/1673-5374.382986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/26/2023] [Accepted: 07/10/2023] [Indexed: 10/22/2023] Open
Abstract
Satellite glial cells are unique glial cells that surround the cell body of primary sensory neurons. An increasing body of evidence suggests that in the presence of inflammation and nerve damage, a significant number of satellite glial cells become activated, thus triggering a series of functional changes. This suggests that satellite glial cells are closely related to the occurrence of chronic pain. In this review, we first summarize the morphological structure, molecular markers, and physiological functions of satellite glial cells. Then, we clarify the multiple key roles of satellite glial cells in chronic pain, including gap junction hemichannel Cx43, membrane channel Pannexin1, K channel subunit 4.1, ATP, purinergic P2 receptors, and a series of additional factors and their receptors, including tumor necrosis factor, glutamate, endothelin, and bradykinin. Finally, we propose that future research should focus on the specific sorting of satellite glial cells, and identify genomic differences between physiological and pathological conditions. This review provides an important perspective for clarifying mechanisms underlying the peripheral regulation of chronic pain and will facilitate the formulation of new treatment plans for chronic pain.
Collapse
Affiliation(s)
- Xiaoyun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yuanzhi Yang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaoli Da
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
3
|
Xing Q, Cibelli A, Yang GL, Dohare P, Li QH, Scemes E, Guan FX, Spray DC. Neuronal Panx1 drives peripheral sensitization in experimental plantar inflammatory pain. Mil Med Res 2024; 11:27. [PMID: 38685116 PMCID: PMC11057180 DOI: 10.1186/s40779-024-00525-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The channel-forming protein Pannexin1 (Panx1) has been implicated in both human studies and animal models of chronic pain, but the underlying mechanisms remain incompletely understood. METHODS Wild-type (WT, n = 24), global Panx1 KO (n = 24), neuron-specific Panx1 KO (n = 20), and glia-specific Panx1 KO (n = 20) mice were used in this study at Albert Einstein College of Medicine. The von Frey test was used to quantify pain sensitivity in these mice following complete Freund's adjuvant (CFA) injection (7, 14, and 21 d). The qRT-PCR was employed to measure mRNA levels of Panx1, Panx2, Panx3, Cx43, Calhm1, and β-catenin. Laser scanning confocal microscopy imaging, Sholl analysis, and electrophysiology were utilized to evaluate the impact of Panx1 on neuronal excitability and morphology in Neuro2a and dorsal root ganglion neurons (DRGNs) in which Panx1 expression or function was manipulated. Ethidium bromide (EtBr) dye uptake assay and calcium imaging were employed to investigate the role of Panx1 in adenosine triphosphate (ATP) sensitivity. β-galactosidase (β-gal) staining was applied to determine the relative cellular expression levels of Panx1 in trigeminal ganglia (TG) and DRG of transgenic mice. RESULTS Global or neuron-specific Panx1 deletion markedly decreased pain thresholds after CFA stimuli (7, 14, and 21 d; P < 0.01 vs. WT group), indicating that Panx1 was positively correlated with pain sensitivity. In Neuro2a, global Panx1 deletion dramatically reduced neurite extension and inward currents compared to the WT group (P < 0.05), revealing that Panx1 enhanced neurogenesis and excitability. Similarly, global Panx1 deletion significantly suppressed Wnt/β-catenin dependent DRG neurogenesis following 5 d of nerve growth factor (NGF) treatment (P < 0.01 vs. WT group). Moreover, Panx1 channels enhanced DRG neuron response to ATP after CFA injection (P < 0.01 vs. Panx1 KO group). Furthermore, ATP release increased Ca2+ responses in DRGNs and satellite glial cells surrounding them following 7 d of CFA treatment (P < 0.01 vs. Panx1 KO group), suggesting that Panx1 in glia also impacts exaggerated neuronal excitability. Interestingly, neuron-specific Panx1 deletion was found to markedly reduce differentiation in cultured DRGNs, as evidenced by stunted neurite outgrowth (P < 0.05 vs. Panx1 KO group; P < 0.01 vs. WT group or GFAP-Cre group), blunted activation of Wnt/β-catenin signaling (P < 0.01 vs. WT, Panx1 KO and GFAP-Cre groups), and diminished cell excitability (P < 0.01 vs. GFAP-Cre group) and response to ATP stimulation (P < 0.01 vs. WT group). Analysis of β-gal staining showed that cellular expression levels of Panx1 in neurons are significantly higher (2.5-fold increase) in the DRG than in the TG. CONCLUSIONS The present study revealed that neuronal Panx1 is a prominent driver of peripheral sensitivity in the setting of inflammatory pain through cell-autonomous effects on neuronal excitability. This hyperexcitability dependence on neuronal Panx1 contrasts with inflammatory orofacial pain, where similar studies revealed a prominent role for glial Panx1. The apparent differences in Panx1 expression in neuronal and non-neuronal TG and DRG cells are likely responsible for the distinct impact of these cell types in the two pain models.
Collapse
Affiliation(s)
- Qu Xing
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Antonio Cibelli
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, 70125, Italy
| | - Greta Luyuan Yang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, 06459, USA
| | - Preeti Dohare
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, 12208, USA
| | - Qing-Hua Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Eliana Scemes
- Department of Anatomy and Cell Biology, New York Medical College, Valhalla, NY, 10595, USA
| | - Fang-Xia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450001, China.
| | - David C Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
4
|
BzATP Activates Satellite Glial Cells and Increases the Excitability of Dorsal Root Ganglia Neurons In Vivo. Cells 2022; 11:cells11152280. [PMID: 35892578 PMCID: PMC9330736 DOI: 10.3390/cells11152280] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 12/14/2022] Open
Abstract
The purinergic system plays an important role in pain transmission. Recent studies have suggested that activation of P2-purinergic receptors (P2Rs) may be involved in neuron-satellite glial cell (SGC) interactions in the dorsal root ganglia (DRG), but the details remain unclear. In DRG, P2X7R is selectively expressed in SGCs, which closely surround neurons, and is highly sensitive to 3’-O-(4-Benzoyl) benzoyl-ATP (BzATP). Using calcium imaging in intact mice to survey a large number of DRG neurons and SGCs, we examined how intra-ganglionic purinergic signaling initiated by BzATP affects neuronal activities in vivo. We developed GFAP-GCaMP6s and Pirt-GCaMP6s mice to express the genetically encoded calcium indicator GGCaM6s in SGCs and DRG neurons, respectively. The application of BzATP to the ganglion induced concentration-dependent activation of SGCs in GFAP-GCaMP6s mice. In Pirt-GCaMP6s mice, BzATP initially activated more large-size neurons than small-size ones. Both glial and neuronal responses to BzATP were blocked by A438079, a P2X7R-selective antagonist. Moreover, blockers to pannexin1 channels (probenecid) and P2X3R (A317491) also reduced the actions of BzATP, suggesting that P2X7R stimulation may induce the opening of pannexin1 channels, leading to paracrine ATP release, which could further excite neurons by acting on P2X3Rs. Importantly, BzATP increased the responses of small-size DRG neurons and wide-dynamic range spinal neurons to subsequent peripheral stimuli. Our findings suggest that intra-ganglionic purinergic signaling initiated by P2X7R activation could trigger SGC-neuron interaction in vivo and increase DRG neuron excitability.
Collapse
|
5
|
Sinegubov A, Andreeva D, Burzak N, Vasyutina M, Murashova L, Dyachuk V. Heterogeneity and Potency of Peripheral Glial Cells in Embryonic Development and Adults. Front Mol Neurosci 2022; 15:737949. [PMID: 35401107 PMCID: PMC8990813 DOI: 10.3389/fnmol.2022.737949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
This review describes the heterogeneity of peripheral glial cell populations, from the emergence of Schwann cells (SCs) in early development, to their involvement, and that of their derivatives in adult glial populations. We focus on the origin of the first glial precursors from neural crest cells (NCCs), and their ability to differentiate into several cell types during development. We also discuss the heterogeneity of embryonic glia in light of the latest data from genetic tracing and transcriptome analysis. Special attention has been paid to the biology of glial populations in adult animals, by highlighting common features of different glial cell types and molecular differences that modulate their functions. Finally, we consider the communication of glial cells with axons of neurons in normal and pathological conditions. In conclusion, the present review details how information available on glial cell types and their functions in normal and pathological conditions may be utilized in the development of novel therapeutic strategies for the treatment of patients with neurodiseases.
Collapse
|
6
|
Long JY, Wang XJ, Li XY, Kong XH, Yang G, Zhang D, Yang YT, Shi Z, Ma XP. Spinal Microglia and Astrocytes: Two Key Players in Chronic Visceral Pain Pathogenesis. Neurochem Res 2022; 47:545-551. [PMID: 34797501 DOI: 10.1007/s11064-021-03486-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 10/19/2022]
Abstract
Chronic visceral pain (CVP) is one of the common symptoms of many diseases triggered by underlying diseases of the internal organs of the human body. Its causes include vascular mechanisms, mechanical factors, persistent inflammation, and unexplained functional mechanisms. Although the pathogenesis is unclear, more and more research has begun to shift from the neuronal aspect to the glial cells in recent years. Some data highlight that the spinal glial cells, particularly the microglia and astrocytes, play an essential role in CVP. Based on this, we highlight the mechanisms of microglia and astrocytes in CVP concerning the release of cytokines, chemokines, and neuroactive substances and alterations in intracellular signaling pathways during the process. Finally, because CVP is widespread in various diseases, we present future perspectives targeting microglia and astrocytes for treatment.
Collapse
Affiliation(s)
- Jun-Yi Long
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Xue-Jun Wang
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Xiao-Ying Li
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Xie-He Kong
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Guang Yang
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Dan Zhang
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Yan-Ting Yang
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Zheng Shi
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Xiao-Peng Ma
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China.
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China.
| |
Collapse
|
7
|
Kurisu R, Saigusa T, Aono Y, Hayashi Y, Hitomi S, Shimada M, Iwata K, Shinoda M. Pannexin 1 role in the trigeminal ganglion in infraorbital nerve injury-induced mechanical allodynia. Oral Dis 2022; 29:1770-1781. [PMID: 35029007 DOI: 10.1111/odi.14129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES The detailed pathological mechanism of orofacial neuropathic pain remains unknown. We aimed to examine the pannexin 1 (Panx1) signaling in the trigeminal ganglion (TG) involvement in infraorbital nerve injury (IONI)-induced orofacial neuropathic pain. MATERIALS AND METHODS Mechanical head-withdrawal threshold (MHWT) was measured in IONI-treated rats receiving intra-TG Panx1 inhibitor or metabotropic glutamate receptor 5 (mGluR5) antagonist administration and MHWTs in naive rats receiving intra-TG mGluR5 agonist administration post-IONI. Glutamate and Panx1 in the TG were measured post-IONI. Panx1, mGluR5, and glutamine synthetase expression in TG were immunohistochemically identified, and changes in the number of mGluR5-P2X3 -expressed TG neurons were examined. RESULTS MHWT was significantly decreased post-IONI, and this decrease was reversed by Panx1 inhibition or mGluR5 antagonism. mGluR5 agonism induced a decrease in the MHWT. IONI increased extracellular glutamate in TG. Panx1 was expressed in satellite glial cells and TG neurons, and intra-TG mGluR5 antagonism decreased the number of mGluR5 and P2X3 positive TG neurons post-IONI. CONCLUSIONS IONI facilitates glutamate release via Panx1 that activates mGluR5 which was expressed in the nociceptive TG neurons innervating the orofacial region. In turn, P2X3 receptor-expressed TG neurons is enhanced via mGluR5 signaling, resulting in orofacial neuropathic pain.
Collapse
Affiliation(s)
- Ryoko Kurisu
- Dental Anesthesiology and Orofacial Pain Management, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tadashi Saigusa
- Department of Pharmacology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Yuri Aono
- Department of Pharmacology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Masahiko Shimada
- Dental Anesthesiology and Orofacial Pain Management, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
8
|
Liu YD, Tang G, Qian F, Liu L, Huang JR, Tang FR. Astroglial Connexins in Neurological and Neuropsychological Disorders and Radiation Exposure. Curr Med Chem 2021; 28:1970-1986. [PMID: 32520676 DOI: 10.2174/0929867327666200610175037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 11/22/2022]
Abstract
Radiotherapy is a common treatment for brain and spinal cord tumors and also a risk factor for neuropathological changes in the brain leading to different neurological and neuropsychological disorders. Astroglial connexins are involved in brain inflammation, development of Alzheimer's Disease (AD), depressive, epilepsy, and amyotrophic lateral sclerosis, and are affected by radiation exposure. Therefore, it is speculated that radiation-induced changes of astroglial connexins may be related to the brain neuropathology and development of neurological and neuropsychological disorders. In this paper, we review the functional expression and regulation of astroglial connexins expressed between astrocytes and different types of brain cells (including oligodendrocytes, microglia, neurons and endothelial cells). The roles of these connexins in the development of AD, depressive, epilepsy, amyotrophic lateral sclerosis and brain inflammation have also been summarized. The radiation-induced astroglial connexins changes and development of different neurological and neuropsychological disorders are then discussed. Based on currently available data, we propose that radiation-induced astroglial connexins changes may be involved in the genesis of different neurological and neuropsychological disorders which depends on the age, brain regions, and radiation doses/dose rates. The abnormal astroglial connexins may be novel therapeutic targets for the prevention of radiation-induced cognitive impairment, neurological and neuropsychological disorders.
Collapse
Affiliation(s)
- Yuan Duo Liu
- Medical School of Yangtze University, Jingzhou 434000, China
| | - Ge Tang
- Woodlands Health Campus, National Healthcare Group Singapore, Singapore
| | - Feng Qian
- Medical School of Yangtze University, Jingzhou 434000, China
| | - Lian Liu
- Medical School of Yangtze University, Jingzhou 434000, China
| | | | - Feng Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| |
Collapse
|
9
|
Obot P, Velíšek L, Velíšková J, Scemes E. The Contribution of Astrocyte and Neuronal Panx1 to Seizures Is Model and Brain Region Dependent. ASN Neuro 2021; 13:17590914211007273. [PMID: 33910381 PMCID: PMC8718119 DOI: 10.1177/17590914211007273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Pannexin1 (Panx1) is an ATP release channel expressed in neurons and astrocytes that plays important roles in CNS physiology and pathology. Evidence for the involvement of Panx1 in seizures includes the reduction of epileptiform activity and ictal discharges following Panx1 channel blockade or deletion. However, very little is known about the relative contribution of astrocyte and neuronal Panx1 channels to hyperexcitability. To this end, mice with global and cell type specific deletion of Panx1 were used in one in vivo and two in vitro seizure models. In the low-Mg2+ in vitro model, global deletion but not cell-type specific deletion of Panx1 reduced the frequency of epileptiform discharges. This reduced frequency of discharges did not impact the overall power spectra obtained from local field potentials. In the in vitro KA model, in contrast, global or cell type specific deletion of Panx1 did not affect the frequency of discharges, but reduced the overall power spectra. EEG recordings following KA-injection in vivo revealed that although global deletion of Panx1 did not affect the onset of status epilepticus (SE), SE onset was delayed in mice lacking neuronal Panx1 and accelerated in mice lacking astrocyte Panx1. EEG power spectral analysis disclosed a Panx1-dependent cortical region effect; while in the occipital region, overall spectral power was reduced in all three Panx1 genotypes; in the frontal cortex, the overall power was not affected by deletion of Panx1. Together, our results show that the contribution of Panx1 to ictal activity is model, cell-type and brain region dependent.
Collapse
Affiliation(s)
- Price Obot
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States
| | - Libor Velíšek
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States.,Department of Neurology, New York Medical College, Valhalla, New York, United States.,Department of Pediatrics, New York Medical College, Valhalla, New York, United States
| | - Jana Velíšková
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States.,Department of Neurology, New York Medical College, Valhalla, New York, United States.,Department of Obstetrics & Gynecology, New York Medical College, Valhalla, New York, United States
| | - Eliana Scemes
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States
| |
Collapse
|
10
|
Natha CM, Vemulapalli V, Fiori MC, Chang CWT, Altenberg GA. Connexin hemichannel inhibitors with a focus on aminoglycosides. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166115. [PMID: 33711451 DOI: 10.1016/j.bbadis.2021.166115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/06/2021] [Accepted: 02/22/2021] [Indexed: 12/31/2022]
Abstract
Connexins are membrane proteins involved directly in cell-to-cell communication through the formation of gap-junctional channels. These channels result from the head-to-head docking of two hemichannels, one from each of two adjacent cells. Undocked hemichannels are also present at the plasma membrane where they mediate the efflux of molecules that participate in autocrine and paracrine signaling, but abnormal increase in hemichannel activity can lead to cell damage in disorders such as cardiac infarct, stroke, deafness, cataracts, and skin diseases. For this reason, connexin hemichannels have emerged as a valid therapeutic target. Know small molecule hemichannel inhibitors are not ideal leads for the development of better drugs for clinical use because they are not specific and/or have toxic effects. Newer inhibitors are more selective and include connexin mimetic peptides, anti-connexin antibodies and drugs that reduce connexin expression such as antisense oligonucleotides. Re-purposed drugs and their derivatives are also promising because of the significant experience with their clinical use. Among these, aminoglycoside antibiotics have been identified as inhibitors of connexin hemichannels that do not inhibit gap-junctional channels. In this review, we discuss connexin hemichannels and their inhibitors, with a focus on aminoglycoside antibiotics and derivatives of kanamycin A that inhibit connexin hemichannels, but do not have antibiotic effect.
Collapse
Affiliation(s)
- Cristina M Natha
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Varun Vemulapalli
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Mariana C Fiori
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Cheng-Wei T Chang
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, USA
| | - Guillermo A Altenberg
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
11
|
Mitroshina EV, Krivonosov MI, Burmistrov DE, Savyuk MO, Mishchenko TA, Ivanchenko MV, Vedunova MV. Signatures of the Consolidated Response of Astrocytes to Ischemic Factors In Vitro. Int J Mol Sci 2020; 21:E7952. [PMID: 33114758 PMCID: PMC7672566 DOI: 10.3390/ijms21217952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/20/2022] Open
Abstract
Whether and under what conditions astrocytes can mount a collective network response has recently become one of the central questions in neurobiology. Here, we address this problem, investigating astrocytic reactions to different biochemical stimuli and ischemic-like conditions in vitro. Identifying an emergent astrocytic network is based on a novel mathematical approach that extracts calcium activity from time-lapse fluorescence imaging and estimates the connectivity of astrocytes. The developed algorithm represents the astrocytic network as an oriented graph in which the nodes correspond to separate astrocytes, and the edges indicate high dynamical correlations between astrocytic events. We demonstrate that ischemic-like conditions decrease network connectivity in primary cultures in vitro, although calcium events persist. Importantly, we found that stimulation under normal conditions with 10 µM ATP increases the number of long-range connections and the degree of corresponding correlations in calcium activity, apart from the frequency of calcium events. This result indicates that astrocytes can form a large functional network in response to certain stimuli. In the post-ischemic interval, the response to ATP stimulation is not manifested, which suggests a deep lesion in functional astrocytic networks. The blockade of Connexin 43 during ischemic modeling preserves the connectivity of astrocytes in the post-hypoxic period.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, 603950 Nizhny Novgorod, Russia; (D.E.B.); (M.O.S.); (T.A.M.)
| | - Mikhail I. Krivonosov
- Institute of Information, Technology, Mathematics and Mechanics, Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, 603950 Nizhny Novgorod, Russia; (M.I.K.); (M.V.I.)
| | - Dmitriy E. Burmistrov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, 603950 Nizhny Novgorod, Russia; (D.E.B.); (M.O.S.); (T.A.M.)
| | - Maria O. Savyuk
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, 603950 Nizhny Novgorod, Russia; (D.E.B.); (M.O.S.); (T.A.M.)
| | - Tatiana A. Mishchenko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, 603950 Nizhny Novgorod, Russia; (D.E.B.); (M.O.S.); (T.A.M.)
| | - Mikhail V. Ivanchenko
- Institute of Information, Technology, Mathematics and Mechanics, Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, 603950 Nizhny Novgorod, Russia; (M.I.K.); (M.V.I.)
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, 603950 Nizhny Novgorod, Russia; (D.E.B.); (M.O.S.); (T.A.M.)
| |
Collapse
|
12
|
Tedoldi A, Argent L, Montgomery JM. The role of the tripartite synapse in the heart: how glial cells may contribute to the physiology and pathophysiology of the intracardiac nervous system. Am J Physiol Cell Physiol 2020; 320:C1-C14. [PMID: 33085497 DOI: 10.1152/ajpcell.00363.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the major roles of the intracardiac nervous system (ICNS) is to act as the final site of signal integration for efferent information destined for the myocardium to enable local control of heart rate and rhythm. Multiple subtypes of neurons exist in the ICNS where they are organized into clusters termed ganglionated plexi (GP). The majority of cells in the ICNS are actually glial cells; however, despite this, ICNS glial cells have received little attention to date. In the central nervous system, where glial cell function has been widely studied, glia are no longer viewed simply as supportive cells but rather have been shown to play an active role in modulating neuronal excitability and synaptic plasticity. Pioneering studies have demonstrated that in addition to glia within the brain stem, glial cells within multiple autonomic ganglia in the peripheral nervous system, including the ICNS, can also act to modulate cardiovascular function. Clinically, patients with atrial fibrillation (AF) undergoing catheter ablation show high plasma levels of S100B, a protein produced by cardiac glial cells, correlated with decreased AF recurrence. Interestingly, S100B also alters GP neuron excitability and neurite outgrowth in the ICNS. These studies highlight the importance of understanding how glial cells can affect the heart by modulating GP neuron activity or synaptic inputs. Here, we review studies investigating glia both in the central and peripheral nervous systems to discuss the potential role of glia in controlling cardiac function in health and disease, paying particular attention to the glial cells of the ICNS.
Collapse
Affiliation(s)
- Angelo Tedoldi
- Department of Physiology, University of Auckland, Auckland, New Zealand.,Manaaki Mānawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Liam Argent
- Department of Physiology, University of Auckland, Auckland, New Zealand.,Manaaki Mānawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology, University of Auckland, Auckland, New Zealand.,Manaaki Mānawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
13
|
Singh N, Driessen AK, McGovern AE, Moe AAK, Farrell MJ, Mazzone SB. Peripheral and central mechanisms of cough hypersensitivity. J Thorac Dis 2020; 12:5179-5193. [PMID: 33145095 PMCID: PMC7578480 DOI: 10.21037/jtd-2020-icc-007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chronic cough is a difficult to treat symptom of many respiratory and some non-respiratory diseases, indicating that varied pathologies can underpin the development of chronic cough. However, clinically and experimentally it has been useful to collate these different pathological processes into the single unifying concept of cough hypersensitivity. Cough hypersensitivity syndrome is reflected by troublesome cough often precipitated by levels of stimuli that ordinarily don't cause cough in healthy people, and this appears to be a hallmark feature in many patients with chronic cough. Accordingly, a strong argument has emerged that changes in the excitability and/or normal regulation of the peripheral and central neural circuits responsible for cough are instrumental in establishing cough hypersensitivity and for causing excessive cough in disease. In this review, we explore the current peripheral and central neural mechanisms that are believed to be involved in altered cough sensitivity and present possible links to the mechanism of action of novel therapies that are currently undergoing clinical trials for chronic cough.
Collapse
Affiliation(s)
- Nabita Singh
- Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Australia
| | - Alexandria K. Driessen
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Alice E. McGovern
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Aung Aung Kywe Moe
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Michael J. Farrell
- Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Australia
- Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Stuart B. Mazzone
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| |
Collapse
|
14
|
Driessen AK, Devlin AC, Lundy FT, Martin SL, Sergeant GP, Mazzone SB, McGarvey LP. Perspectives on neuroinflammation contributing to chronic cough. Eur Respir J 2020; 56:13993003.00758-2020. [DOI: 10.1183/13993003.00758-2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
Abstract
Chronic cough can be a troublesome clinical problem. Current thinking is that increased activity and/or enhanced sensitivity of the peripheral and central neural pathways mediates chronic cough via processes similar to those associated with the development of chronic pain. While inflammation is widely thought to be involved in the development of chronic cough, the true mechanisms causing altered neural activity and sensitisation remain largely unknown. In this back-to-basics perspective article we explore evidence that inflammation in chronic cough may, at least in part, involve neuroinflammation orchestrated by glial cells of the nervous system. We summarise the extensive evidence for the role of both peripheral and central glial cells in chronic pain, and hypothesise that the commonalities between pain and cough pathogenesis and clinical presentation warrant investigations into the neuroinflammatory mechanisms that contribute to chronic cough. We open the debate that glial cells may represent an underappreciated therapeutic target for controlling troublesome cough in disease.
Collapse
|
15
|
Enes J, Haburčák M, Sona S, Gerard N, Mitchell AC, Fu W, Birren SJ. Satellite glial cells modulate cholinergic transmission between sympathetic neurons. PLoS One 2020; 15:e0218643. [PMID: 32017764 PMCID: PMC6999876 DOI: 10.1371/journal.pone.0218643] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 01/15/2020] [Indexed: 02/07/2023] Open
Abstract
Postganglionic sympathetic neurons and satellite glial cells are the two major cell types of the peripheral sympathetic ganglia. Sympathetic neurons project to and provide neural control of peripheral organs and have been implicated in human disorders ranging from cardiovascular disease to peripheral neuropathies. Here we show that satellite glia regulate synaptic activity of cultured postnatal sympathetic neurons, providing evidence for local ganglionic control of sympathetic drive. In addition to modulating neuron-to-neuron cholinergic neurotransmission, satellite glia promote synapse formation and contribute to neuronal survival. Examination of the cellular architecture of the rat sympathetic ganglia in vivo shows this regulation of neuronal properties takes place during a developmental period in which neuronal morphology and density are actively changing and satellite glia enwrap sympathetic neuronal somata. Cultured satellite glia make and release factors that promote neuronal activity and that can partially rescue the neurons from cell death following nerve growth factor deprivation. Thus, satellite glia play an early and ongoing role within the postnatal sympathetic ganglia, expanding our understanding of the contributions of local and target-derived factors in the regulation of sympathetic neuron function.
Collapse
Affiliation(s)
- Joana Enes
- Department of Biology, Brandeis University, Waltham, MA, United States of America
- Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States of America
| | - Marián Haburčák
- Department of Biology, Brandeis University, Waltham, MA, United States of America
- Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States of America
| | - Surbhi Sona
- Department of Biology, Brandeis University, Waltham, MA, United States of America
- Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States of America
| | - Nega Gerard
- Department of Biology, Brandeis University, Waltham, MA, United States of America
- Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States of America
| | - Alexander C. Mitchell
- Department of Biology, Brandeis University, Waltham, MA, United States of America
- Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States of America
| | - Wenqi Fu
- Department of Biology, Brandeis University, Waltham, MA, United States of America
- Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States of America
| | - Susan J. Birren
- Department of Biology, Brandeis University, Waltham, MA, United States of America
- Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States of America
- * E-mail:
| |
Collapse
|
16
|
Delvaeye T, De Smet MAJ, Verwaerde S, Decrock E, Czekaj A, Vandenbroucke RE, Lemeire K, Gonçalves A, Declercq W, Vandenabeele P, Krysko DV, Leybaert L. Blocking connexin43 hemichannels protects mice against tumour necrosis factor-induced inflammatory shock. Sci Rep 2019; 9:16623. [PMID: 31719598 PMCID: PMC6851386 DOI: 10.1038/s41598-019-52900-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Upon intravenous injection of tumour necrosis factor (TNF) in mice, a systemic inflammatory response syndrome (SIRS) is initiated, characterized by an acute cytokine storm and induction of vascular hyperpermeability. Connexin43 hemichannels have been implicated in various pathological conditions, e.g. ischemia and inflammation, and can lead to detrimental cellular outcomes. Here, we explored whether targeting connexin43 hemichannels could alleviate TNF-induced endothelial barrier dysfunction and lethality in SIRS. Therefore, we verified whether administration of connexin43-targeting-peptides affected survival, body temperature and vascular permeability in vivo. In vitro, TNF-effects on connexin43 hemichannel function were investigated by single-channel studies and Ca2+-imaging. Blocking connexin43 hemichannels with TAT-Gap19 protected mice against TNF-induced mortality, hypothermia and vascular leakage, while enhancing connexin43 hemichannel function with TAT-CT9 provoked opposite sensitizing effects. In vitro patch-clamp studies revealed that TNF acutely activated connexin43 hemichannel opening in endothelial cells, which was promoted by CT9, and inhibited by Gap19 and intracellular Ca2+-buffering. In vivo experiments aimed at buffering intracellular Ca2+, and pharmacologically targeting Ca2+/calmodulin-dependent protein kinase-II, a known modulator of endothelial barrier integrity, demonstrated their involvement in permeability alterations. Our results demonstrate significant benefits of inhibiting connexin43 hemichannels to counteract TNF-induced SIRS-associated vascular permeability and lethality.
Collapse
Affiliation(s)
- Tinneke Delvaeye
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Maarten A J De Smet
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Stijn Verwaerde
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Elke Decrock
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Aleksandra Czekaj
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Kelly Lemeire
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Amanda Gonçalves
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,VIB BioImaging Core, Ghent, Belgium
| | - Wim Declercq
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- VIB Center for Inflammation Research, Ghent, Belgium. .,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium. .,Methusalem Program, Ghent University, Ghent, Belgium.
| | - Dmitri V Krysko
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium.
| | - Luc Leybaert
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
17
|
Ahmadian E, Eftekhari A, Samiei M, Maleki Dizaj S, Vinken M. The role and therapeutic potential of connexins, pannexins and their channels in Parkinson's disease. Cell Signal 2019; 58:111-118. [DOI: 10.1016/j.cellsig.2019.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/10/2019] [Accepted: 03/10/2019] [Indexed: 02/07/2023]
|
18
|
Xing L, Yang T, Cui S, Chen G. Connexin Hemichannels in Astrocytes: Role in CNS Disorders. Front Mol Neurosci 2019; 12:23. [PMID: 30787868 PMCID: PMC6372977 DOI: 10.3389/fnmol.2019.00023] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
In the central nervous system (CNS), astrocytes form networks interconnected by gap junctions made from connexins of the subtypes Cx30 and Cx43. When unopposed by an adjoining hemichannel, astrocytic connexins can act as hemichannels to control the release of small molecules such as ATP and glutamate into the extracellular space. Accruing evidence indicates that astrocytic connexins are crucial for the coordination and maintenance of physiologic CNS activity. Here we provide an update on the role of astrocytic connexins in neurodegenerative disorders, glioma, and ischemia. In addition, we address the regulation of Cx43 in chronic pain.
Collapse
Affiliation(s)
- LingYan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - ShuSen Cui
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Gang Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
19
|
Horton SM, Luna Lopez C, Blevins E, Howarth H, Weisberg J, Shestopalov VI, Makarenkova HP, Shah SB. Pannexin 1 Modulates Axonal Growth in Mouse Peripheral Nerves. Front Cell Neurosci 2017; 11:365. [PMID: 29213230 PMCID: PMC5702652 DOI: 10.3389/fncel.2017.00365] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/06/2017] [Indexed: 01/29/2023] Open
Abstract
The pannexin family of channels consists of three members—pannexin-1 (Panx1), pannexin-2 (Panx2), and pannexin-3 (Panx3) that enable the exchange of metabolites and signaling molecules between intracellular and extracellular compartments. Pannexin-mediated release of intracellular ATP into the extracellular space has been tied to a number of cellular activities, primarily through the activity of type P2 purinergic receptors. Previous work indicates that the opening of Panx1 channels and activation of purinergic receptors by extracellular ATP may cause inflammation and apoptosis. In the CNS (central nervous system) and PNS (peripheral nervous system), coupled pannexin, and P2 functions have been linked to peripheral sensitization (pain) pathways. Purinergic pathways are also essential for other critical processes in the PNS, including myelination and neurite outgrowth. However, whether such pathways are pannexin-dependent remains to be determined. In this study, we use a Panx1 knockout mouse model and pharmacological inhibitors of the Panx1 and the ATP-mediated signaling pathway to fill gaps in our understanding of Panx1 localization in peripheral nerves, roles for Panx1 in axonal outgrowth and myelination, and neurite extension. Our data show that Panx1 is localized to axonal, myelin, and vascular compartments of the peripheral nerves. Knockout of Panx1 gene significantly increased axonal caliber in vivo and axonal growth rate in cultured dorsal root ganglia (DRG) neurons. Furthermore, genetic knockout of Panx1 or inhibition of components of purinergic signaling, by treatment with probenecid and apyrase, resulted in denser axonal outgrowth from cultured DRG explants compared to untreated wild-types. Our findings suggest that Panx1 regulates axonal growth in the peripheral nervous system.
Collapse
Affiliation(s)
- Steven M Horton
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, CA, United States
| | - Carlos Luna Lopez
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, CA, United States
| | - Elisabeth Blevins
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, CA, United States.,Research Service, Veterans Affairs San Diego Healthcare System, San Diego, La Jolla, CA, United States
| | - Holly Howarth
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Jake Weisberg
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, CA, United States
| | | | - Helen P Makarenkova
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Sameer B Shah
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, CA, United States.,Research Service, Veterans Affairs San Diego Healthcare System, San Diego, La Jolla, CA, United States.,Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
20
|
Retamal MA, Riquelme MA, Stehberg J, Alcayaga J. Connexin43 Hemichannels in Satellite Glial Cells, Can They Influence Sensory Neuron Activity? Front Mol Neurosci 2017; 10:374. [PMID: 29200997 PMCID: PMC5696352 DOI: 10.3389/fnmol.2017.00374] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022] Open
Abstract
In this review article, we summarize the current insight on the role of Connexin- and Pannexin-based channels as modulators of sensory neurons. The somas of sensory neurons are located in sensory ganglia (i.e., trigeminal and nodose ganglia). It is well known that within sensory ganglia, sensory neurons do not form neither electrical nor chemical synapses. One of the reasons for this is that each soma is surrounded by glial cells, known as satellite glial cells (SGCs). Recent evidence shows that connexin43 (Cx43) hemichannels and probably pannexons located at SGCs have an important role in paracrine communication between glial cells and sensory neurons. This communication may be exerted via the release of bioactive molecules from SGCs and their subsequent action on receptors located at the soma of sensory neurons. The glio-neuronal communication seems to be relevant for the establishment of chronic pain, hyperalgesia and pathologies associated with tissue inflammation. Based on the current literature, it is possible to propose that Cx43 hemichannels expressed in SGCs could be a novel pharmacological target for treating chronic pain, which need to be directly evaluated in future studies.
Collapse
Affiliation(s)
- Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clinica Alemana Universidad del Desarrollo, Santiago, Chile.,Department of Cell Physiology and Molecular Biophysics, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - Jimmy Stehberg
- Laboratorio de Neurobiología, Centro de Investigaciones Biomedicas, Universidad Andres Bello, Santiago, Chile
| | - Julio Alcayaga
- Department of Biology, Cell Physiology Center, University of Chile, Santiago, Chile
| |
Collapse
|
21
|
Neuronal P2X7 Receptors Revisited: Do They Really Exist? J Neurosci 2017; 37:7049-7062. [PMID: 28747388 DOI: 10.1523/jneurosci.3103-16.2017] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/22/2017] [Accepted: 05/24/2017] [Indexed: 12/13/2022] Open
Abstract
P2X7 receptors (Rs) constitute a subclass of ATP-sensitive ionotropic receptors (P2X1-P2X7). P2X7Rs have many distinguishing features, mostly based on their long intracellular C terminus regulating trafficking to the cell membrane, protein-protein interactions, and post-translational modification. Their C-terminal tail is especially important in enabling the transition from the nonselective ion channel mode to a membrane pore allowing the passage of large molecules. There is an ongoing dispute on the existence of neuronal P2X7Rs with consequences for our knowledge on their involvement in neuroinflammation, aggravating stroke, temporal lobe epilepsy, neuropathic pain, and various neurodegenerative diseases. Whereas early results appeared to support the operation of P2X7Rs at neurons, more recently glial P2X7Rs are increasingly considered as indirect causes of neuronal effects. Specific tools for P2X7Rs are of limited value because of the poor selectivity of agonists, and the inherent failure of antibodies to differentiate between the large number of active and inactive splice variants, or gain-of-function and loss-of-function small nucleotide polymorphisms of the receptor. Unfortunately, the available P2RX7 knock-out mice generated by pharmaceutical companies possess certain splice variants, which evade inactivation. In view of the recently discovered bidirectional dialogue between astrocytes and neurons (and even microglia and neurons), we offer an alternative explanation for previous data, which assumedly support the existence of P2X7Rs at neurons. We think that the unbiased reader will follow our argumentation on astrocytic or microglial P2X7Rs being the primary targets of pathologically high extracellular ATP concentrations, although a neuronal localization of these receptors cannot be fully excluded either.
Collapse
|
22
|
Abstract
Enhanced expression and function of gap junctions and pannexin (Panx) channels have been associated with both peripheral and central mechanisms of pain sensitization. At the level of the sensory ganglia, evidence includes augmented gap junction and pannexin1 expression in glial cells and neurons in inflammatory and neuropathic pain models and increased synchrony and enhanced cross-excitation among sensory neurons by gap junction-mediated coupling. In spinal cord and in suprapinal areas, evidence is largely limited to increased expression of relevant proteins, although in several rodent pain models, hypersensitivity is reduced by treatment with gap junction/Panx1 channel blocking compounds. Moreover, targeted modulation of Cx43 expression was shown to modulate pain thresholds, albeit in somewhat contradictory ways, and mice lacking Panx1 expression globally or in specific cell types show depressed hyperalgesia. We here review the evidence for involvement of gap junctions and Panx channels in a variety of animal pain studies and then discuss ways in which gap junctions and Panx channels may mediate their action in pain processing. This discussion focusses on spread of signals among satellite glial cells, in particular intercellular Ca2+ waves, which are propagated through both gap junction and Panx1-dependent routes and have been associated with the phenomenon of spreading depression and the malady of migraine headache with aura.
Collapse
|
23
|
Beyer EC, Berthoud VM. Gap junction gene and protein families: Connexins, innexins, and pannexins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:5-8. [PMID: 28559187 DOI: 10.1016/j.bbamem.2017.05.016] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 05/18/2017] [Accepted: 05/25/2017] [Indexed: 02/08/2023]
Abstract
Gap junction channels facilitate the intercellular exchange of ions and small molecules. While this process is critical to all multicellular organisms, the proteins that form gap junction channels are not conserved. Vertebrate gap junctions are formed by connexins, while invertebrate gap junctions are formed by innexins. Interestingly, vertebrates and lower chordates contain innexin homologs, the pannexins, which also form channels, but rarely (if ever) make intercellular channels. While the connexin and the innexin/pannexin polypeptides do not share significant sequence similarity, all three of these protein families share a similar membrane topology and some similarities in quaternary structure. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Eric C Beyer
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, United States.
| | - Viviana M Berthoud
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, United States
| |
Collapse
|
24
|
Bargiello TA, Oh S, Tang Q, Bargiello NK, Dowd TL, Kwon T. Gating of Connexin Channels by transjunctional-voltage: Conformations and models of open and closed states. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:22-39. [PMID: 28476631 DOI: 10.1016/j.bbamem.2017.04.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/26/2017] [Accepted: 04/28/2017] [Indexed: 11/18/2022]
Abstract
Voltage is an important physiologic regulator of channels formed by the connexin gene family. Connexins are unique among ion channels in that both plasma membrane inserted hemichannels (undocked hemichannels) and intercellular channels (aggregates of which form gap junctions) have important physiological roles. The hemichannel is the fundamental unit of gap junction voltage-gating. Each hemichannel displays two distinct voltage-gating mechanisms that are primarily sensitive to a voltage gradient formed along the length of the channel pore (the transjunctional voltage) rather than sensitivity to the absolute membrane potential (Vm or Vi-o). These transjunctional voltage dependent processes have been termed Vj- or fast-gating and loop- or slow-gating. Understanding the mechanism of voltage-gating, defined as the sequence of voltage-driven transitions that connect open and closed states, first and foremost requires atomic resolution models of the end states. Although ion channels formed by connexins were among the first to be characterized structurally by electron microscopy and x-ray diffraction in the early 1980's, subsequent progress has been slow. Much of the current understanding of the structure-function relations of connexin channels is based on two crystal structures of Cx26 gap junction channels. Refinement of crystal structure by all-atom molecular dynamics and incorporation of charge changing protein modifications has resulted in an atomic model of the open state that arguably corresponds to the physiologic open state. Obtaining validated atomic models of voltage-dependent closed states is more challenging, as there are currently no methods to solve protein structure while a stable voltage gradient is applied across the length of an oriented channel. It is widely believed that the best approach to solve the atomic structure of a voltage-gated closed ion channel is to apply different but complementary experimental and computational methods and to use the resulting information to derive a consensus atomic structure that is then subjected to rigorous validation. In this paper, we summarize our efforts to obtain and validate atomic models of the open and voltage-driven closed states of undocked connexin hemichannels. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Thaddeus A Bargiello
- Dominic P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| | - Seunghoon Oh
- Department of Physiology, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Qingxiu Tang
- Dominic P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Nicholas K Bargiello
- Dominic P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Terry L Dowd
- Department of Chemistry, Brooklyn College, Brooklyn, NY 11210, United States
| | - Taekyung Kwon
- Dominic P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| |
Collapse
|
25
|
Nielsen BS, Hansen DB, Ransom BR, Nielsen MS, MacAulay N. Connexin Hemichannels in Astrocytes: An Assessment of Controversies Regarding Their Functional Characteristics. Neurochem Res 2017; 42:2537-2550. [DOI: 10.1007/s11064-017-2243-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/17/2017] [Accepted: 03/17/2017] [Indexed: 12/19/2022]
|
26
|
Batir Y, Bargiello TA, Dowd TL. Structural studies of N-terminal mutants of Connexin 26 and Connexin 32 using (1)H NMR spectroscopy. Arch Biochem Biophys 2016; 608:8-19. [PMID: 27378082 PMCID: PMC5051353 DOI: 10.1016/j.abb.2016.06.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/25/2022]
Abstract
Alterations in gap junctions underlie the etiologies of syndromic deafness (KID) and Charcot-Marie Tooth disease (CMTX). Functional gap junctions are composed of connexin molecules with N-termini containing a flexible turn around G12, inserting the N-termini into the channel pore allowing voltage gating. The loss of this turn correlates with loss of Connexin 32 (Cx32) function by impaired trafficking to the cell membrane. Using (1)H NMR we show the N-terminus of a syndromic deafness mutation Cx26G12R, producing "leaky channels", contains a turn around G12 which is less structured and more flexible than wild-type. In contrast, the N-terminal structure of the same mutation in Cx32 chimera, Cx32*43E1G12R shows a larger constricted turn and no membrane current expression but forms membrane inserted hemichannels. Their function was rescued by formation of heteromeric channels with wild type subunits. We suggest the inflexible Cx32G12R N-terminus blocks ion conduction in homomeric channels and this channel block is relieved by incorporation of wild type subunits. In contrast, the increased open probability of Cx26G12R hemichannels is likely due to the addition of positive charge in the channel pore changing pore electrostatics and impairing hemichannel regulation by Ca(2+). These results provide mechanistic information on aberrant channel activity observed in disease.
Collapse
Affiliation(s)
- Yuksel Batir
- Department of Chemistry, Brooklyn College, Brooklyn, NY 11210, United States
| | - Thaddeus A Bargiello
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Terry L Dowd
- Department of Chemistry, Brooklyn College, Brooklyn, NY 11210, United States; Ph.D. Program in Chemistry and Biochemistry, The Graduate Center of the City University of New York, New York, NY, 10016, United States.
| |
Collapse
|
27
|
Molecular Signaling and Dysfunction of the Human Reactive Enteric Glial Cell Phenotype: Implications for GI Infection, IBD, POI, Neurological, Motility, and GI Disorders. Inflamm Bowel Dis 2016; 22:1812-34. [PMID: 27416040 PMCID: PMC4993196 DOI: 10.1097/mib.0000000000000854] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Clinical observations or animal studies implicate enteric glial cells in motility disorders, irritable bowel syndrome, inflammatory bowel disease, gastrointestinal (GI) infections, postoperative ileus, and slow transit constipation. Mechanisms underlying glial responses to inflammation in human GI tract are not understood. Our goal was to identify the "reactive human enteric glial cell (rhEGC) phenotype" induced by inflammation, and probe its functional relevance. METHODS Human enteric glial cells in culture from 15 GI-surgical specimens were used to study gene expression, Ca, and purinergic signaling by Ca/fluo-4 imaging and mechanosensitivity. A nanostring panel of 107 genes was designed as a read out of inflammation, transcription, purinergic signaling, vesicular transport protein, channel, antioxidant, and other pathways. A 24-hour treatment with lipopolysaccharide (200 μg/mL) and interferon-γ (10 μg/mL) was used to induce inflammation and study molecular signaling, flow-dependent Ca responses from 3 mL/min to 10 mL/min, adenosine triphosphate (ATP) release, and ATP responses. RESULTS Treatment induced a "rhEGC phenotype" and caused up-regulation in messenger RNA transcripts of 58% of 107 genes analyzed. Regulated genes included inflammatory genes (54%/IP10; IFN-γ; CxCl2; CCL3; CCL2; C3; s100B; IL-1β; IL-2R; TNF-α; IL-4; IL-6; IL-8; IL-10; IL-12A; IL-17A; IL-22; and IL-33), purine-genes (52%/AdoR2A; AdoR2B; P2RY1; P2RY2; P2RY6; P2RX3; P2RX7; AMPD3; ENTPD2; ENTPD3; and NADSYN1), channels (40%/Panx1; CHRNA7; TRPV1; and TRPA1), vesicular transporters (SYT1, SYT2, SNAP25, and SYP), transcription factors (relA/relB, SOCS3, STAT3, GATA_3, and FOXP3), growth factors (IGFBP5 and GMCSF), antioxidant genes (SOD2 and HMOX1), and enzymes (NOS2; TPH2; and CASP3) (P < 0.0001). Treatment disrupted Ca signaling, ATP, and mechanical/flow-dependent Ca responses in human enteric glial cells. ATP release increased 5-fold and s100B decreased 33%. CONCLUSIONS The "rhEGC phenotype" is identified by a complex cascade of pro-inflammatory pathways leading to alterations of important molecular and functional signaling pathways (Ca, purinergic, and mechanosensory) that could disrupt GI motility. Inflammation induced a "purinergic switch" from ATP to adenosine diphosphate/adenosine/uridine triphosphate signaling. Findings have implications for GI infection, inflammatory bowel disease, postoperative ileus, motility, and GI disorders.
Collapse
|
28
|
Orellana JA, Retamal MA, Moraga-Amaro R, Stehberg J. Role of Astroglial Hemichannels and Pannexons in Memory and Neurodegenerative Diseases. Front Integr Neurosci 2016; 10:26. [PMID: 27489539 PMCID: PMC4951483 DOI: 10.3389/fnint.2016.00026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/06/2016] [Indexed: 11/13/2022] Open
Abstract
Under physiological conditions, astroglial hemichannels and pannexons allow the release of gliotransmitters from astrocytes. These gliotransmitters are critical in modulating synaptic transmission, plasticity and memory. However, recent evidence suggests that under pathological conditions, they may be central in the development of various neurodegenerative diseases. Here we review current literature on the role of astroglial hemichannels and pannexons in memory, stress and the development of neurodegenerative diseases, and propose that they are not only crucial for normal brain function, including memory, but also a potential target for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Juan A Orellana
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo Santiago, Chile
| | - Rodrigo Moraga-Amaro
- Laboratorio de Neurobiología, Centro de Investigaciones Biomédicas, Universidad Andres Bello Santiago, Chile
| | - Jimmy Stehberg
- Laboratorio de Neurobiología, Centro de Investigaciones Biomédicas, Universidad Andres Bello Santiago, Chile
| |
Collapse
|
29
|
Bultynck G. The anti-metastatic micro-environment of the bone: Importance of osteocyte Cx43 hemichannels. Biochim Biophys Acta Rev Cancer 2016; 1866:121-7. [PMID: 27400952 DOI: 10.1016/j.bbcan.2016.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/04/2016] [Accepted: 07/07/2016] [Indexed: 12/25/2022]
Abstract
Bone metastases of tumor cells are a common and life-threatening feature of a variety of late-stage cancers, including breast cancers. However, until now, much less has been known about the intrinsic anti-metastatic properties of the bones and how these could be exploited to prevent or treat bone metastases. Very recently, native Cx43 hemichannels present in osteocytes have been identified as important anti-metastatic signaling complexes by establishing high local extracellular ATP levels. Moreover, bisphosphonate drugs, applied as adjuvant therapies in the treatment of breast cancer patients and bone diseases, are known to display anti-metastatic properties. Now, it became clear that these compounds exert their effects through osteocyte Cx43 hemichannels, thereby triggering their opening and promoting ATP release in the extracellular micro-environment. Hence, endogenous osteocyte Cx43 hemichannels emerge as important and promising therapeutic targets for the prevention of bone metastases and/or clinical treatment of bone-metastasized breast cancers.
Collapse
Affiliation(s)
- Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE 3000 Leuven, Belgium.
| |
Collapse
|
30
|
Bele T, Fabbretti E. The scaffold protein calcium/calmodulin-dependent serine protein kinase controls ATP release in sensory ganglia upon P2X3 receptor activation and is part of an ATP keeper complex. J Neurochem 2016; 138:587-97. [PMID: 27217099 DOI: 10.1111/jnc.13680] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/06/2016] [Accepted: 05/11/2016] [Indexed: 12/27/2022]
Abstract
P2X3 receptors, gated by extracellular ATP, are expressed by sensory neurons and are involved in peripheral nociception and pain sensitization. The ability of P2X3 receptors to transduce extracellular stimuli into neuronal signals critically depends on the dynamic molecular partnership with the calcium/calmodulin-dependent serine protein kinase (CASK). The present work used trigeminal sensory neurons to study the impact that activation of P2X3 receptors (evoked by the agonist α,β-meATP) has on the release of endogenous ATP and how CASK modulates this phenomenon. P2X3 receptor function was followed by ATP efflux via Pannexin1 (Panx1) hemichannels, a mechanism that was blocked by the P2X3 receptor antagonist A-317491, and by P2X3 silencing. ATP efflux was enhanced by nerve growth factor, a treatment known to potentiate P2X3 receptor function. Basal ATP efflux was not controlled by CASK, and carbenoxolone or Pannexin silencing reduced ATP release upon P2X3 receptor function. CASK-controlled ATP efflux followed P2X3 receptor activity, but not depolarization-evoked ATP release. Molecular biology experiments showed that CASK was essential for the transactivation of Panx1 upon P2X3 receptor activation. These data suggest that P2X3 receptor function controls a new type of feed-forward purinergic signaling on surrounding cells, with consequences at peripheral and spinal cord level. Thus, P2X3 receptor-mediated ATP efflux may be considered for the future development of pharmacological strategies aimed at containing neuronal sensitization. P2X3 receptors are involved in sensory transduction and associate to CASK. We have studied in primary sensory neurons the molecular mechanisms downstream P2X3 receptor activation, namely ATP release and partnership with CASK or Panx1. Our data suggest that CASK and P2X3 receptors are part of an ATP keeper complex, with important feed-forward consequences at peripheral and central level.
Collapse
Affiliation(s)
- Tanja Bele
- Center for Biomedical Sciences and Engineering, University of Nova Gorica, Nova Gorica, Slovenia
| | - Elsa Fabbretti
- Center for Biomedical Sciences and Engineering, University of Nova Gorica, Nova Gorica, Slovenia
| |
Collapse
|
31
|
Orellana JA. Editorial: Single membrane channels formed by connexins or pannexins: focus on the nervous system. Front Cell Neurosci 2015; 9:402. [PMID: 26528132 PMCID: PMC4606011 DOI: 10.3389/fncel.2015.00402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 09/22/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Juan A Orellana
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
32
|
Hanani M. Role of satellite glial cells in gastrointestinal pain. Front Cell Neurosci 2015; 9:412. [PMID: 26528140 PMCID: PMC4602093 DOI: 10.3389/fncel.2015.00412] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/28/2015] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal (GI) pain is a common clinical problem, for which effective therapy is quite limited. Sensations from the GI tract, including pain, are mediated largely by neurons in the dorsal root ganglia (DRG), and to a smaller extent by vagal afferents emerging from neurons in the nodose/jugular ganglia. Neurons in rodent DRG become hyperexcitable in models of GI pain (e.g., gastric or colonic inflammation), and can serve as a source for chronic pain. Glial cells are another element in the pain signaling pathways, and there is evidence that spinal glial cells (microglia and astrocytes) undergo activation (gliosis) in various pain models and contribute to pain. Recently it was found that satellite glial cells (SGCs), the main type of glial cells in sensory ganglia, might also contribute to chronic pain in rodent models. Most of that work focused on somatic pain, but in several studies GI pain was also investigated, and these are discussed in the present review. We have shown that colonic inflammation induced by dinitrobenzene sulfonic acid (DNBS) in mice leads to the activation of SGCs in DRG and increases gap junction-mediated coupling among these cells. This coupling appears to contribute to the hyperexcitability of DRG neurons that innervate the colon. Blocking gap junctions (GJ) in vitro reduced neuronal hyperexcitability induced by inflammation, suggesting that glial GJ participate in SGC-neuron interactions. Moreover, blocking GJ by carbenoxolone and other agents reduces pain behavior. Similar changes in SGCs were also found in the mouse nodose ganglia (NG), which provide sensory innervation to most of the GI tract. Following systemic inflammation, SGCs in these ganglia were activated, and displayed augmented coupling and greater sensitivity to the pain mediator ATP. The contribution of these changes to visceral pain remains to be determined. These results indicate that although visceral pain is unique, it shares basic mechanisms with somatic pain, suggesting that therapeutic approaches to both pain types may be similar. Future research in this field should include additional types of GI injury and also other types of visceral pain.
Collapse
Affiliation(s)
- Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus Jerusalem, Israel
| |
Collapse
|
33
|
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
|
34
|
Retamal MA, Reyes EP, García IE, Pinto B, Martínez AD, González C. Diseases associated with leaky hemichannels. Front Cell Neurosci 2015; 9:267. [PMID: 26283912 PMCID: PMC4515567 DOI: 10.3389/fncel.2015.00267] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/29/2015] [Indexed: 01/10/2023] Open
Abstract
Hemichannels (HCs) and gap junction channels (GJCs) formed by protein subunits called connexins (Cxs) are major pathways for intercellular communication. While HCs connect the intracellular compartment with the extracellular milieu, GJCs allow the interchange of molecules between cytoplasm of two contacting cells. Under physiological conditions, HCs are mostly closed, but they can open under certain stimuli allowing the release of autocrine and paracrine molecules. Moreover, some pathological conditions, like ischemia or other inflammation conditions, significantly increase HCs activity. In addition, some mutations in Cx genes associated with human diseases, such as deafness or cataracts, lead to the formation of more active HCs or “leaky HCs.” In this article we will revise cellular and molecular mechanisms underlying the appearance of leaky HCs, and the consequences of their expression in different cellular systems and animal models, in seeking a common pattern or pathological mechanism of disease.
Collapse
Affiliation(s)
- Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo Santiago, Chile
| | - Edison P Reyes
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo Santiago, Chile ; Centro de Investigación Biomédica, Universidad Autónoma de Chile Santiago, Chile
| | - Isaac E García
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso Valparaíso, Chile
| | - Bernardo Pinto
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso Valparaíso, Chile
| | - Agustín D Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso Valparaíso, Chile
| | - Carlos González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso Valparaíso, Chile
| |
Collapse
|
35
|
Feldman-Goriachnik R, Belzer V, Hanani M. Systemic inflammation activates satellite glial cells in the mouse nodose ganglion and alters their functions. Glia 2015; 63:2121-2132. [DOI: 10.1002/glia.22881] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 06/08/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Rachel Feldman-Goriachnik
- Laboratory of Experimental Surgery; Hadassah-Hebrew University Medical Center; Mount Scopus Jerusalem 91240 Israel
| | - Vitali Belzer
- Laboratory of Experimental Surgery; Hadassah-Hebrew University Medical Center; Mount Scopus Jerusalem 91240 Israel
| | - Menachem Hanani
- Laboratory of Experimental Surgery; Hadassah-Hebrew University Medical Center; Mount Scopus Jerusalem 91240 Israel
| |
Collapse
|
36
|
Retamal MA, Reyes EP, Alcayaga J. Petrosal ganglion: a more complex role than originally imagined. Front Physiol 2014; 5:474. [PMID: 25538627 PMCID: PMC4255496 DOI: 10.3389/fphys.2014.00474] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 11/17/2014] [Indexed: 11/13/2022] Open
Abstract
The petrosal ganglion (PG) is a peripheral sensory ganglion, composed of pseudomonopolar sensory neurons that innervate the posterior third of the tongue and the carotid sinus and body. According to their electrical properties PG neurons can be ascribed to one of two categories: (i) neurons with action potentials presenting an inflection (hump) on its repolarizing phase and (ii) neurons with fast and brisk action potentials. Although there is some correlation between the electrophysiological properties and the sensory modality of the neurons in some species, no general pattern can be easily recognized. On the other hand, petrosal neurons projecting to the carotid body are activated by several transmitters, with acetylcholine and ATP being the most conspicuous in most species. Petrosal neurons are completely surrounded by a multi-cellular sheet of glial (satellite) cells that prevents the formation of chemical or electrical synapses between neurons. Thus, PG neurons are regarded as mere wires that communicate the periphery (i.e., carotid body) and the central nervous system. However, it has been shown that in other sensory ganglia satellite glial cells and their neighboring neurons can interact, partly by the release of chemical neuro-glio transmitters. This intercellular communication can potentially modulate the excitatory status of sensory neurons and thus the afferent discharge. In this mini review, we will briefly summarize the general properties of PG neurons and the current knowledge about the glial-neuron communication in sensory neurons and how this phenomenon could be important in the chemical sensory processing generated in the carotid body.
Collapse
Affiliation(s)
- Mauricio A Retamal
- Facultad de Medicina, Centro de Fisiología Celular e Integrativa, Clínica Alemana Universidad del Desarrollo Santiago, Chile
| | - Edison P Reyes
- Facultad de Medicina, Centro de Fisiología Celular e Integrativa, Clínica Alemana Universidad del Desarrollo Santiago, Chile ; Dirección de Investigación, Universidad Autónoma de Chile Temuco, Chile
| | - Julio Alcayaga
- Laboratorio de Fisiología Celular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile Santiago, Chile
| |
Collapse
|
37
|
D'hondt C, Iyyathurai J, Himpens B, Leybaert L, Bultynck G. Cx43-hemichannel function and regulation in physiology and pathophysiology: insights from the bovine corneal endothelial cell system and beyond. Front Physiol 2014; 5:348. [PMID: 25309448 PMCID: PMC4162354 DOI: 10.3389/fphys.2014.00348] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/25/2014] [Indexed: 12/13/2022] Open
Abstract
Intercellular communication in primary bovine corneal endothelial cells (BCECs) is mainly driven by the release of extracellular ATP through Cx43 hemichannels. Studying the characteristics of Ca2+-wave propagation in BCECs, an important form of intercellular communication, in response to physiological signaling events has led to the discovery of important insights in the functional properties and regulation of native Cx43 hemichannels. Together with ectopic expression models for Cx43 hemichannels and truncated/mutated Cx43 versions, it became very clear that loop/tail interactions play a key role in controlling the activity of Cx43 hemichannels. Interestingly, the negative regulation of Cx43 hemichannels by enhanced actin/myosin contractility seems to impinge upon loss of these loop/tail interactions essential for opening Cx43 hemichannels. Finally, these molecular insights have spurred the development of novel peptide tools that can selectively inhibit Cx43 hemichannels, but neither Cx43 gap junctions nor hemichannels formed by other Cx isoforms. These tools now set the stage to hunt for novel physiological functions for Cx43 hemichannels in primary cells and tissues and to tackle disease conditions associated with excessive, pathological Cx43-hemichannel openings.
Collapse
Affiliation(s)
- Catheleyne D'hondt
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven Leuven, Belgium
| | - Jegan Iyyathurai
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven Leuven, Belgium
| | - Bernard Himpens
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven Leuven, Belgium
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University Ghent, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven Leuven, Belgium
| |
Collapse
|