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
|
Ruan Z, Lee J, Li Y, Du J, Lü W. Human pannexin 1 channel is not phosphorylated by Src tyrosine kinase at Tyr199 and Tyr309. eLife 2024; 13:RP95118. [PMID: 38780416 PMCID: PMC11115448 DOI: 10.7554/elife.95118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Protein phosphorylation is one of the major molecular mechanisms regulating protein activity and function throughout the cell. Pannexin 1 (PANX1) is a large-pore channel permeable to ATP and other cellular metabolites. Its tyrosine phosphorylation and subsequent activation have been found to play critical roles in diverse cellular conditions, including neuronal cell death, acute inflammation, and smooth muscle contraction. Specifically, the non-receptor kinase Src has been reported to phosphorylate Tyr198 and Tyr308 of mouse PANX1 (equivalent to Tyr199 and Tyr309 of human PANX1), resulting in channel opening and ATP release. Although the Src-dependent PANX1 activation mechanism has been widely discussed in the literature, independent validation of the tyrosine phosphorylation of PANX1 has been lacking. Here, we show that commercially available antibodies against the two phosphorylation sites mentioned above-which were used to identify endogenous PANX1 phosphorylation at these two sites-are nonspecific and should not be used to interpret results related to PANX1 phosphorylation. We further provide evidence that neither tyrosine residue is a major phosphorylation site for Src kinase in heterologous expression systems. We call on the field to re-examine the existing paradigm of tyrosine phosphorylation-dependent activation of the PANX1 channel.
Collapse
Affiliation(s)
- Zheng Ruan
- Department of Structural Biology, Van Andel InstituteGrand RapidsUnited States
| | - Junuk Lee
- Department of Structural Biology, Van Andel InstituteGrand RapidsUnited States
| | - Yangyang Li
- Department of Structural Biology, Van Andel InstituteGrand RapidsUnited States
| | - Juan Du
- Department of Structural Biology, Van Andel InstituteGrand RapidsUnited States
| | - Wei Lü
- Department of Structural Biology, Van Andel InstituteGrand RapidsUnited States
| |
Collapse
|
3
|
Huang Y, Shi Y, Wang M, Liu B, Chang X, Xiao X, Yu H, Cui X, Bai Y. Pannexin1 Channel-Mediated Inflammation in Acute Ischemic Stroke. Aging Dis 2024; 15:1296-1307. [PMID: 37196132 PMCID: PMC11081155 DOI: 10.14336/ad.2023.0303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/03/2023] [Indexed: 05/19/2023] Open
Abstract
Emerging evidence suggests that inflammation mediated by the pannexin1 channel contributes significantly to acute ischemic stroke. It is believed that the pannexin1 channel is key in initiating central system inflammation during the early stages of acute ischemic stroke. Moreover, the pannexin1 channel is involved in the inflammatory cascade to maintain the inflammation levels. Specifically, the interaction of pannexin1 channels with ATP-sensitive P2X7 purinoceptors or promotion of potassium efflux mediates the activation of the NLRP3 inflammasome, triggering the release of pro-inflammatory factors such as IL-1 and IL-18, exacerbating and sustaining inflammation of brain. Also, increased release of ATP induced by cerebrovascular injury activates pannexin1 in vascular endothelial cells. This signal directs peripheral leukocytes to migrate into ischemic brain tissue, leading to an expansion of the inflammatory zone. Intervention strategies targeting pannexin1 channels may greatly alleviate inflammation after acute ischemic stroke to improve this patient population's clinical outcomes. In this review, we sought to summarize relevant studies on inflammation mediated by the pannexin1 channel in acute ischemic stroke and discussed the possibility of using brain organoid-on-a-chip technology to screen miRNAs that exclusively target the pannexin1 channel to provide new therapeutic measures for targeted regulation of pannexin1 channel to reduce inflammation in acute ischemic stroke.
Collapse
Affiliation(s)
- Yubing Huang
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Yutong Shi
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Mengmeng Wang
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Medical College, Institute of Microanalysis, Dalian University, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Bingyi Liu
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Xueqin Chang
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Xia Xiao
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Huihui Yu
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Xiaodie Cui
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
- Graduate School, Dalian University, Dalian, Liaoning, China
| | - Ying Bai
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| |
Collapse
|
4
|
Fukuyama K, Motomura E, Okada M. Age-Dependent Activation of Pannexin1 Function Contributes to the Development of Epileptogenesis in Autosomal Dominant Sleep-related Hypermotor Epilepsy Model Rats. Int J Mol Sci 2024; 25:1619. [PMID: 38338895 PMCID: PMC10855882 DOI: 10.3390/ijms25031619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
To explore the processes of epileptogenesis/ictogenesis, this study determined the age-dependent development of the functional abnormalities in astroglial transmission associated with pannexin1-hemichannel using a genetic rat model of autosomal dominant sleep-related hypermotor epilepsy (ADSHE) named 'S286L-TG'. Pannexin1 expression in the plasma membrane of primary cultured cortical astrocytes and the orbitofrontal cortex (OFC), which is an ADSHE focus region, were determined using capillary immunoblotting. Astroglial D-serine releases induced by artificial high-frequency oscillation (HFO)-evoked stimulation, the removal of extracellular Ca2+, and the P2X7 receptor agonist (BzATP) were determined using ultra-high performance liquid chromatography (UHPLC). The expressions of pannexin1 in the plasma membrane fraction of the OFC in S286L-TG at four weeks old were almost equivalent when compared to the wild type. The pannexin1 expression in the OFC of the wild type non-statistically decreased age-dependently, whereas that in S286L-TG significantly increased age-dependently, resulting in relatively increasing pannexin1 expression from the 7- (at the onset of interictal discharge) and 10-week-old (after the ADSHE seizure onset) S286L-TG compared to the wild type. However, no functional abnormalities of astroglial pannexin1 expression or D-serine release through the pannexin1-hemichannels from the cultured astrocytes of S286L-TG could be detected. Acutely HFO-evoked stimulation, such as physiological ripple burst (200 Hz) and epileptogenic fast ripple burst (500 Hz), frequency-dependently increased both pannexin1 expression in the astroglial plasma membrane and astroglial D-serine release. Neither the selective inhibitors of pannexin1-hemichannel (10PANX) nor connexin43-hemichannel (Gap19) affected astroglial D-serine release during the resting stage, whereas HFO-evoked D-serine release was suppressed by both inhibitors. The inhibitory effect of 10PANX on the ripple burst-evoked D-serine release was more predominant than that of Gap19, whereas fast ripple burst-evoked D-serine release was predominantly suppressed by Gap19 rather than 10PANX. Astroglial D-serine release induced by acute exposure to BzATP was suppressed by 10PANX but not by Gap19. These results suggest that physiological ripple burst during the sleep spindle plays important roles in the organization of some components of cognition in healthy individuals, but conversely, it contributes to the initial development of epileptogenesis/ictogenesis in individuals who have ADSHE vulnerability via activation of the astroglial excitatory transmission associated with pannexin1-hemichannels.
Collapse
Affiliation(s)
| | | | - Motohiro Okada
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan; (K.F.); (E.M.)
| |
Collapse
|
5
|
Wang X, Dong YT, Hu XM, Zhang JZ, Shi NR, Zuo YQ, Wang X. The circadian regulation of extracellular ATP. Purinergic Signal 2023; 19:283-295. [PMID: 35939197 PMCID: PMC9984637 DOI: 10.1007/s11302-022-09881-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 06/24/2022] [Indexed: 11/25/2022] Open
Abstract
Extracellular ATP is a potent signaling molecule released from various cells throughout the body and is intimately involved in the pathophysiological functions of the nervous system and immune system by activating P2 purinergic receptors. Recent increasingly studies showed that extracellular ATP exhibits circadian oscillation with an approximately 24-h periodicity, which participates in regulatory pathways of central oscillator suprachiasmatic nucleus and peripheral oscillator bladder, respectively. Oscillators modulate the protein expression of ATP release channels and ectonucleotidase activity through clock genes; indeed, real-time alterations of ATP release and degradation determine outcomes of temporal character on extracellular ATP rhythm. The regulatory pathways on extracellular ATP rhythm are different in central and peripheral systems. In this review, we summarize the circadian rhythm of extracellular ATP and discuss several circadian regulatory pathways in different organs via ATP release and degradation, to provide a new understanding for purinergic signaling in the regulatory mechanism of circadian rhythm and a potential target to research the circadian regulation of extracellular ATP in other circadian oscillators.
Collapse
Affiliation(s)
- Xin Wang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shi-er Qiao Road, Chengdu, 610075 China
| | - Yu-Ting Dong
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shi-er Qiao Road, Chengdu, 610075 China
| | - Xiu-Ming Hu
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shi-er Qiao Road, Chengdu, 610075 China
| | - Ji-Zhou Zhang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shi-er Qiao Road, Chengdu, 610075 China
| | - Nan-Rui Shi
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shi-er Qiao Road, Chengdu, 610075 China
| | - Yan-Qin Zuo
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shi-er Qiao Road, Chengdu, 610075 China
| | - Xu Wang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shi-er Qiao Road, Chengdu, 610075 China
| |
Collapse
|
6
|
Baracaldo-Santamaría D, Corrales-Hernández MG, Ortiz-Vergara MC, Cormane-Alfaro V, Luque-Bernal RM, Calderon-Ospina CA, Cediel-Becerra JF. Connexins and Pannexins: Important Players in Neurodevelopment, Neurological Diseases, and Potential Therapeutics. Biomedicines 2022; 10:2237. [PMID: 36140338 PMCID: PMC9496069 DOI: 10.3390/biomedicines10092237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Cell-to-cell communication is essential for proper embryonic development and its dysfunction may lead to disease. Recent research has drawn attention to a new group of molecules called connexins (Cxs) and pannexins (Panxs). Cxs have been described for more than forty years as pivotal regulators of embryogenesis; however, the exact mechanism by which they provide this regulation has not been clearly elucidated. Consequently, Cxs and Panxs have been linked to congenital neurodegenerative diseases such as Charcot-Marie-Tooth disease and, more recently, chronic hemichannel opening has been associated with adult neurodegenerative diseases (e.g., Alzheimer's disease). Cell-to-cell communication via gap junctions formed by hexameric assemblies of Cxs, known as connexons, is believed to be a crucial component in developmental regulation. As for Panxs, despite being topologically similar to Cxs, they predominantly seem to form channels connecting the cytoplasm to the extracellular space and, despite recent research into Panx1 (Pannexin 1) expression in different regions of the brain during the embryonic phase, it has been studied to a lesser degree. When it comes to the nervous system, Cxs and Panxs play an important role in early stages of neuronal development with a wide span of action ranging from cellular migration during early stages to neuronal differentiation and system circuitry formation. In this review, we describe the most recent available evidence regarding the molecular and structural aspects of Cx and Panx channels, their role in neurodevelopment, congenital and adult neurological diseases, and finally propose how pharmacological modulation of these channels could modify the pathogenesis of some diseases.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - María Gabriela Corrales-Hernández
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Maria Camila Ortiz-Vergara
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Valeria Cormane-Alfaro
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Ricardo-Miguel Luque-Bernal
- Anatomy and Embriology Units, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos-Alberto Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- GENIUROS Research Group, Center for Research in Genetics and Genomics (CIGGUR), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Juan-Fernando Cediel-Becerra
- Histology and Embryology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| |
Collapse
|
7
|
Zhang X, Lee MD, Buckley C, Wilson C, McCarron JG. Mitochondria regulate TRPV4-mediated release of ATP. Br J Pharmacol 2022; 179:1017-1032. [PMID: 34605007 DOI: 10.1111/bph.15687] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/10/2021] [Accepted: 09/02/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Ca2+ influx via TRPV4 channels triggers Ca2+ release from the IP3 -sensitive internal store to generate repetitive oscillations. Although mitochondria are acknowledged regulators of IP3 -mediated Ca2+ release, how TRPV4-mediated Ca2+ signals are regulated by mitochondria is unknown. We show that depolarised mitochondria switch TRPV4 signalling from relying on Ca2+ -induced Ca2+ release at IP3 receptors to being independent of Ca2+ influx and instead mediated by ATP release via pannexins. EXPERIMENTAL APPROACH TRPV4-evoked Ca2+ signals were individually examined in hundreds of cells in the endothelium of rat mesenteric resistance arteries using the indicator Cal520. KEY RESULTS TRPV4 activation with GSK1016790A (GSK) generated repetitive Ca2+ oscillations that required Ca2+ influx. However, when the mitochondrial membrane potential was depolarised, by the uncoupler CCCP or complex I inhibitor rotenone, TRPV4 activation generated large propagating, multicellular, Ca2+ waves in the absence of external Ca2+ . The ATP synthase inhibitor oligomycin did not potentiate TRPV4-mediated Ca2+ signals. GSK-evoked Ca2+ waves, when mitochondria were depolarised, were blocked by the TRPV4 channel blocker HC067047, the SERCA inhibitor cyclopiazonic acid, the PLC blocker U73122 and the inositol trisphosphate receptor blocker caffeine. The Ca2+ waves were also inhibited by the extracellular ATP blockers suramin and apyrase and the pannexin blocker probenecid. CONCLUSION AND IMPLICATIONS These results highlight a previously unknown role of mitochondria in shaping TRPV4-mediated Ca2+ signalling by facilitating ATP release. When mitochondria are depolarised, TRPV4-mediated release of ATP via pannexin channels activates plasma membrane purinergic receptors to trigger IP3 -evoked Ca2+ release.
Collapse
Affiliation(s)
- Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
8
|
Ding Z, Guo S, Luo L, Zheng Y, Gan S, Kang X, Wu X, Zhu S. Emerging Roles of Microglia in Neuro-vascular Unit: Implications of Microglia-Neurons Interactions. Front Cell Neurosci 2021; 15:706025. [PMID: 34712121 PMCID: PMC8546170 DOI: 10.3389/fncel.2021.706025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/10/2021] [Indexed: 12/20/2022] Open
Abstract
Microglia, which serve as the defensive interface of the nervous system, are activated in many neurological diseases. Their role as immune responding cells has been extensively studied in the past few years. Recent studies have demonstrated that neuronal feedback can be shaped by the molecular signals received and sent by microglia. Altered neuronal activity or synaptic plasticity leads to the release of various communication messages from neurons, which in turn exert effects on microglia. Research on microglia-neuron communication has thus expanded from focusing only on neurons to the neurovascular unit (NVU). This approach can be used to explore the potential mechanism of neurovascular coupling across sophisticated receptor systems and signaling cascades in health and disease. However, it remains unclear how microglia-neuron communication happens in the brain. Here, we discuss the functional contribution of microglia to synapses, neuroimmune communication, and neuronal activity. Moreover, the current state of knowledge of bidirectional control mechanisms regarding interactions between neurons and microglia are reviewed, with a focus on purinergic regulatory systems including ATP-P2RY12R signaling, ATP-adenosine-A1Rs/A2ARs, and the ATP-pannexin 1 hemichannel. This review aims to organize recent studies to highlight the multifunctional roles of microglia within the neural communication network in health and disease.
Collapse
Affiliation(s)
- Zhe Ding
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shaohui Guo
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lihui Luo
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yueying Zheng
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuyuan Gan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianhui Kang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaomin Wu
- Department of Anesthesiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Connexin Expression Is Altered in Liver Development of Yotari ( dab1 -/-) Mice. Int J Mol Sci 2021; 22:ijms221910712. [PMID: 34639052 PMCID: PMC8509723 DOI: 10.3390/ijms221910712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/26/2021] [Accepted: 09/29/2021] [Indexed: 01/05/2023] Open
Abstract
Disabled-1 (Dab1) protein is an intracellular adaptor of reelin signaling required for prenatal neuronal migration, as well as postnatal neurotransmission, memory formation and synaptic plasticity. Yotari, an autosomal recessive mutant of the mouse Dab1 gene is recognizable by its premature death, unstable gait and tremor. Previous findings are mostly based on neuronal abnormalities caused by Dab1 deficiency, but the role of the reelin signaling pathway in nonneuronal tissues and organs has not been studied until recently. Hepatocytes, the most abundant cells in the liver, communicate via gap junctions (GJ) are composed of connexins. Cell communication disruption in yotari mice was examined by analyzing the expression of connexins (Cxs): Cx26, Cx32, Cx37, Cx40, Cx43 and Cx45 during liver development at 13.5 and 15.5 gestation days (E13.5 and E15.5). Analyses were performed using immunohistochemistry and fluorescent microscopy, followed by quantification of area percentage covered by positive signal. Data are expressed as a mean ± SD and analyzed by one-way ANOVA. All Cxs examined displayed a significant decrease in yotari compared to wild type (wt) individuals at E13.5. Looking at E15.5 we have similar results with exception of Cx37 showing negligible expression in wt. Channels formation triggered by pathological stimuli, as well as propensity to apoptosis, was studied by measuring the expression of Pannexin1 (Panx1) and Apoptosis-inducing factor (AIF) through developmental stages mentioned above. An increase in Panx1 expression of E15.5 yotari mice, as well as a strong jump of AIF in both phases suggesting that yotari mice are more prone to apoptosis. Our results emphasize the importance of gap junction intercellular communication (GJIC) during liver development and their possible involvement in liver pathology and diagnostics where they can serve as potential biomarkers and drug targets.
Collapse
|
10
|
Dossi E, Rouach N. Pannexin 1 channels and ATP release in epilepsy: two sides of the same coin : The contribution of pannexin-1, connexins, and CALHM ATP-release channels to purinergic signaling. Purinergic Signal 2021; 17:533-548. [PMID: 34495463 DOI: 10.1007/s11302-021-09818-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/08/2021] [Indexed: 11/29/2022] Open
Abstract
Purinergic signaling mediated by ATP and its metabolites contributes to various brain physiological processes as well as to several pathological conditions, including neurodegenerative and neurological disorders, such as epilepsy. Among the different ATP release pathways, pannexin 1 channels represent one of the major conduits being primarily activated in pathological contexts. Investigations on in vitro and in vivo models of epileptiform activity and seizures in mice and human tissues revealed pannexin 1 involvement in aberrant network activity and epilepsy, and highlighted that pannexin 1 exerts a complex role. Pannexin 1 can indeed either sustain seizures through release of ATP that can directly activate purinergic receptors, or tune down epileptic activity via ATP-derived adenosine that decreases neuronal excitability. Interestingly, in-depth analysis of the literature unveils that this dichotomy is only apparent, as it depends on the model of seizure induction and the type of evoked epileptiform activity, two factors that can differentially activate pannexin 1 channels and trigger distinct intracellular signaling cascades. Here, we review the general properties and ATP permeability of pannexin 1 channels, and discuss their impact on acute epileptiform activity and chronic epilepsy according to the regime of activity and disease state. These data pave the way for the development of new antiepileptic strategies selectively targeting pannexin 1 channels in a context-dependent manner.
Collapse
Affiliation(s)
- Elena Dossi
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Centre National de la Recherche Scientifique UMR 7241, Institut National de la Santé Et de la Recherche Médicale U1050, Collège de France, Labex Memolife, Université PSL, Paris, France.
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Centre National de la Recherche Scientifique UMR 7241, Institut National de la Santé Et de la Recherche Médicale U1050, Collège de France, Labex Memolife, Université PSL, Paris, France.
| |
Collapse
|
11
|
Cárcaba L, García-Piqueras J, García-Mesa Y, Cobo R, García-Suárez O, Feito J, Vega JA. Human digital merkel cells display pannexin1 immunoreactivity. Ann Anat 2021; 239:151813. [PMID: 34384856 DOI: 10.1016/j.aanat.2021.151813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022]
Abstract
Pannexins are channel proteins displaying functional similarities to gap junctions in vertebrates and are regarded as transmembrane ATP-releasing channels. A member of this family, denominate pannexin1, has been detected in the epidermis and cutaneous adnexal structures. Here we used immunohistochemistry to investigate whether human digital Merkel cells express this protein since ATP is postulated as a neurotransmitter in the Merkel cell-axon complexes low-threshold mecahoreceptors. Pannexin1 immunoreactivity was found in cytokeratine 20-, chromogranin A- and synaptophysin-positive cells placed at the basal layer of the epidermis. Cell displaying pannexin1 immunoreactivities were thus identified as Merkel cells and showed close contact with nerve profiles. Light pannexin1 immunoreactivity in dermal blood vessels was also verified. Present results demonstrate for the first time the expression of pannexin1 in human digital Merkel cells supporting the idea that ATP can be involved directly or indirectly in the mechanotransductional process at Merkel-axon complexes.
Collapse
Affiliation(s)
- Lucia Cárcaba
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain
| | - Jorge García-Piqueras
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain
| | - Yolanda García-Mesa
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain
| | - Ramón Cobo
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain
| | - Olivia García-Suárez
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain
| | - Jorge Feito
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain; Servicio de Anatomía Patológica, Complejo Hospitalario Universitario de Salamanca, Salamanca, Spain; Departamento de Anatomía e Histología Humanas, Universidad de Salamanca, Salamanca, Spain
| | - José A Vega
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain; Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago de Chile, Chile.
| |
Collapse
|
12
|
Mim C, Perkins G, Dahl G. Structure versus function: Are new conformations of pannexin 1 yet to be resolved? J Gen Physiol 2021; 153:e202012754. [PMID: 33835130 PMCID: PMC8042604 DOI: 10.1085/jgp.202012754] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pannexin 1 (Panx1) plays a decisive role in multiple physiological and pathological settings, including oxygen delivery to tissues, mucociliary clearance in airways, sepsis, neuropathic pain, and epilepsy. It is widely accepted that Panx1 exerts its role in the context of purinergic signaling by providing a transmembrane pathway for ATP. However, under certain conditions, Panx1 can also act as a highly selective membrane channel for chloride ions without ATP permeability. A recent flurry of publications has provided structural information about the Panx1 channel. However, while these structures are consistent with a chloride selective channel, none show a conformation with strong support for the ATP release function of Panx1. In this Viewpoint, we critically assess the existing evidence for the function and structure of the Panx1 channel and conclude that the structure corresponding to the ATP permeation pathway is yet to be determined. We also list a set of additional topics needing attention and propose ways to attain the large-pore, ATP-permeable conformation of the Panx1 channel.
Collapse
Affiliation(s)
- Carsten Mim
- Department of Biomedical Engineering and Health Systems Royal Institute of Technology, Huddinge, Sweden
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California, San Diego School of Medicine, La Jolla, CA
| | - Gerhard Dahl
- Department of Physiology, University of Miami School of Medicine, Miami, FL
| |
Collapse
|
13
|
Moroz LL, Romanova DY, Kohn AB. Neural versus alternative integrative systems: molecular insights into origins of neurotransmitters. Philos Trans R Soc Lond B Biol Sci 2021; 376:20190762. [PMID: 33550949 PMCID: PMC7935107 DOI: 10.1098/rstb.2019.0762] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Transmitter signalling is the universal chemical language of any nervous system, but little is known about its early evolution. Here, we summarize data about the distribution and functions of neurotransmitter systems in basal metazoans as well as outline hypotheses of their origins. We explore the scenario that neurons arose from genetically different populations of secretory cells capable of volume chemical transmission and integration of behaviours without canonical synapses. The closest representation of this primordial organization is currently found in Placozoa, disk-like animals with the simplest known cell composition but complex behaviours. We propose that injury-related signalling was the evolutionary predecessor for integrative functions of early transmitters such as nitric oxide, ATP, protons, glutamate and small peptides. By contrast, acetylcholine, dopamine, noradrenaline, octopamine, serotonin and histamine were recruited as canonical neurotransmitters relatively later in animal evolution, only in bilaterians. Ligand-gated ion channels often preceded the establishment of novel neurotransmitter systems. Moreover, lineage-specific diversification of neurotransmitter receptors occurred in parallel within Cnidaria and several bilaterian lineages, including acoels. In summary, ancestral diversification of secretory signal molecules provides unique chemical microenvironments for behaviour-driven innovations that pave the way to complex brain functions and elementary cognition. This article is part of the theme issue 'Basal cognition: multicellularity, neurons and the cognitive lens'.
Collapse
Affiliation(s)
- Leonid L. Moroz
- Department of Neuroscience, McKnight Brain Institute and Whitney laboratory, University of Florida, 9505 Ocean shore Blvd, St Augustine, FL 32080, USA
| | - Daria Y. Romanova
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of RAS, 5A Butlerova Street, Moscow 117485, Russia
| | - Andrea B. Kohn
- Department of Neuroscience, McKnight Brain Institute and Whitney laboratory, University of Florida, 9505 Ocean shore Blvd, St Augustine, FL 32080, USA
| |
Collapse
|
14
|
Sarcoma Family Kinase-Dependent Pannexin-1 Activation after Cortical Spreading Depression is Mediated by NR2A-Containing Receptors. Int J Mol Sci 2020; 21:ijms21041269. [PMID: 32070042 PMCID: PMC7072958 DOI: 10.3390/ijms21041269] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/09/2020] [Accepted: 02/11/2020] [Indexed: 01/11/2023] Open
Abstract
Cortical spreading depression (CSD) is a propagating wave of depolarization followed by depression of cortical activity. CSD triggers neuroinflammation via the pannexin-1 (Panx1) channel opening, which may eventually cause migraine headaches. However, the regulatory mechanism of Panx1 is unknown. This study investigates whether sarcoma family kinases (SFK) are involved in transmitting CSD-induced Panx1 activation, which is mediated by the NR2A-containing N-methyl-D-aspartate receptor. CSD was induced by topical application of K+ to cerebral cortices of rats and mouse brain slices. SFK inhibitor, PP2, or NR2A–receptor antagonist, NVP–AAM077, was perfused into contralateral cerebral ventricles (i.c.v.) of rats prior to CSD induction. Co-immunoprecipitation and Western blot were used for detecting protein interactions, and histofluorescence for addressing Panx1 activation. The results demonstrated that PP2 attenuated CSD-induced Panx1 activation in rat ipsilateral cortices. Cortical susceptibility to CSD was reduced by PP2 in rats and by TAT-Panx308 that disrupts SFK–Panx1 interaction in mouse brain slices. Furthermore, CSD promoted activated SFK coupling with Panx1 in rat ipsilateral cortices. Moreover, inhibition of NR2A by NVP–AAM077 reduced elevation of ipsilateral SFK–Panx1 interaction, Panx1 activation induced by CSD and cortical susceptibility to CSD in rats. These data suggest NR2A-regulated, SFK-dependent Panx1 activity plays an important role in migraine aura pathogenesis.
Collapse
|
15
|
Jalaleddine N, El-Hajjar L, Dakik H, Shaito A, Saliba J, Safi R, Zibara K, El-Sabban M. Pannexin1 Is Associated with Enhanced Epithelial-To-Mesenchymal Transition in Human Patient Breast Cancer Tissues and in Breast Cancer Cell Lines. Cancers (Basel) 2019; 11:cancers11121967. [PMID: 31817827 PMCID: PMC6966616 DOI: 10.3390/cancers11121967] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Loss of connexin-mediated cell-cell communication is a hallmark of breast cancer progression. Pannexin1 (PANX1), a glycoprotein that shares structural and functional features with connexins and engages in cell communication with its environment, is highly expressed in breast cancer metastatic foci; however, PANX1 contribution to metastatic progression is still obscure. Here we report elevated expression of PANX1 in different breast cancer (BRCA) subtypes using RNA-seq data from The Cancer Genome Atlas (TCGA). The elevated PANX1 expression correlated with poorer outcomes in TCGA BRCA patients. In addition, gene set enrichment analysis (GSEA) revealed that epithelial-to-mesenchymal transition (EMT) pathway genes correlated positively with PANX1 expression. Pharmacological inhibition of PANX1, in MDA-MB-231 and MCF-7 breast cancer cells, or genetic ablation of PANX1, in MDA-MB-231 cells, reverted the EMT phenotype, as evidenced by decreased expression of EMT markers. In addition, PANX1 inhibition or genetic ablation decreased the invasiveness of MDA-MB-231 cells. Our results suggest PANX1 overexpression in breast cancer is associated with a shift towards an EMT phenotype, in silico and in vitro, attributing to it a tumor-promoting effect, with poorer clinical outcomes in breast cancer patients. This association offers a novel target for breast cancer therapy.
Collapse
Affiliation(s)
- Nour Jalaleddine
- Department of Biological and Environmental Sciences, Faculty of Science, Beirut Arab University, Beirut 1107-2809, Lebanon;
| | - Layal El-Hajjar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon;
| | - Hassan Dakik
- University of Tours, EA 7501 GICC, CNRS ERL 7001 LNOx, CEDEX 01, 37032 Tours, France;
| | - Abdullah Shaito
- Department of Biological and Chemical Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut 1105, Lebanon;
| | - Jessica Saliba
- Department of Biology, Faculty of Sciences, Lebanese University, Hadath, Beirut 1003, Lebanon;
| | - Rémi Safi
- Department of Dermatology, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon;
| | - Kazem Zibara
- ER045-Laboratory of Stem Cells, PRASE, Department of Biology, Faculty of Sciences, Lebanese University, Hadath, Beirut 1003, Lebanon;
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon;
- Correspondence: ; Tel.: +961-1-350000 (ext. 4765-4766)
| |
Collapse
|
16
|
Gap Junction Intercellular Communication in the Carcinogenesis Hallmarks: Is This a Phenomenon or Epiphenomenon? Cells 2019; 8:cells8080896. [PMID: 31416286 PMCID: PMC6721698 DOI: 10.3390/cells8080896] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/03/2019] [Accepted: 08/12/2019] [Indexed: 12/24/2022] Open
Abstract
If occupational tumors are excluded, cancer causes are largely unknown. Therefore, it appeared useful to work out a theory explaining the complexity of this disease. More than fifty years ago the first demonstration that cells communicate with each other by exchanging ions or small molecules through the participation of connexins (Cxs) forming Gap Junctions (GJs) occurred. Then the involvement of GJ Intercellular Communication (GJIC) in numerous physiological cellular functions, especially in proliferation control, was proven and accounts for the growing attention elicited in the field of carcinogenesis. The aim of the present paper is to verify and discuss the role of Cxs, GJs, and GJIC in cancer hallmarks, pointing on the different involved mechanisms in the context of the multi-step theory of carcinogenesis. Functional GJIC acts both as a tumor suppressor and as a tumor enhancer in the metastatic stage. On the contrary, lost or non-functional GJs allow the uncontrolled proliferation of stem/progenitor initiated cells. Thus, GJIC plays a key role in many biological phenomena or epiphenomena related to cancer. Depending on this complexity, GJIC can be considered a tumor suppressor in controlling cell proliferation or a cancer ally, with possible preventive or therapeutic implications in both cases.
Collapse
|
17
|
Beckmann A, Hainz N, Tschernig T, Meier C. Facets of Communication: Gap Junction Ultrastructure and Function in Cancer Stem Cells and Tumor Cells. Cancers (Basel) 2019; 11:cancers11030288. [PMID: 30823688 PMCID: PMC6468480 DOI: 10.3390/cancers11030288] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/28/2022] Open
Abstract
Gap junction proteins are expressed in cancer stem cells and non-stem cancer cells of many tumors. As the morphology and assembly of gap junction channels are crucial for their function in intercellular communication, one focus of our review is to outline the data on gap junction plaque morphology available for cancer cells. Electron microscopic studies and freeze-fracture analyses on gap junction ultrastructure in cancer are summarized. As the presence of gap junctions is relevant in solid tumors, we exemplarily outline their role in glioblastomas and in breast cancer. These were also shown to contain cancer stem cells, which are an essential cause of tumor onset and of tumor transmission into metastases. For these processes, gap junctional communication was shown to be important and thus we summarize, how the expression of gap junction proteins and the resulting communication between cancer stem cells and their surrounding cells contributes to the dissemination of cancer stem cells via blood or lymphatic vessels. Based on their importance for tumors and metastases, future cancer-specific therapies are expected to address gap junction proteins. In turn, gap junctions also seem to contribute to the unattainability of cancer stem cells by certain treatments and might thus contribute to therapeutic resistance.
Collapse
Affiliation(s)
- Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| | - Nadine Hainz
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| | - Thomas Tschernig
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| |
Collapse
|
18
|
Linden J, Koch-Nolte F, Dahl G. Purine Release, Metabolism, and Signaling in the Inflammatory Response. Annu Rev Immunol 2019; 37:325-347. [PMID: 30676821 DOI: 10.1146/annurev-immunol-051116-052406] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
ATP, NAD+, and nucleic acids are abundant purines that, in addition to having critical intracellular functions, have evolved extracellular roles as danger signals released in response to cell lysis, apoptosis, degranulation, or membrane pore formation. In general ATP and NAD+ have excitatory and adenosine has anti-inflammatory effects on immune cells. This review focuses on recent advances in our understanding of purine release mechanisms, ectoenzymes that metabolize purines (CD38, CD39, CD73, ENPP1, and ENPP2/autotaxin), and signaling by key P2 purinergic receptors (P2X7, P2Y2, and P2Y12). In addition to metabolizing ATP or NAD+, some purinergic ectoenzymes metabolize other inflammatory modulators, notably lysophosphatidic acid and cyclic GMP-AMP (cGAMP). Also discussed are extracellular signaling effects of NAD+ mediated by ADP-ribosylation, and epigenetic effects of intracellular adenosine mediated by modification of S-adenosylmethionine-dependent DNA methylation.
Collapse
Affiliation(s)
- Joel Linden
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, California 92037, USA; .,Department of Pharmacology, University of California, San Diego, La Jolla, California 92093, USA
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany;
| | - Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, Florida 33136, USA;
| |
Collapse
|
19
|
Faber DS, Pereda AE. Two Forms of Electrical Transmission Between Neurons. Front Mol Neurosci 2018; 11:427. [PMID: 30534051 PMCID: PMC6276723 DOI: 10.3389/fnmol.2018.00427] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 11/05/2018] [Indexed: 12/30/2022] Open
Abstract
Electrical signaling is a cardinal feature of the nervous system and endows it with the capability of quickly reacting to changes in the environment. Although synaptic communication between nerve cells is perceived to be mainly chemically mediated, electrical synaptic interactions also occur. Two different strategies are responsible for electrical communication between neurons. One is the consequence of low resistance intercellular pathways, called "gap junctions", for the spread of electrical currents between the interior of two cells. The second occurs in the absence of cell-to-cell contacts and is a consequence of the extracellular electrical fields generated by the electrical activity of neurons. Here, we place present notions about electrical transmission in a historical perspective and contrast the contributions of the two different forms of electrical communication to brain function.
Collapse
Affiliation(s)
- Donald S. Faber
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
- Marine Biological Laboratory, Woods Hole, MA, United States
| | - Alberto E. Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
- Marine Biological Laboratory, Woods Hole, MA, United States
| |
Collapse
|
20
|
Dahl G. The Pannexin1 membrane channel: distinct conformations and functions. FEBS Lett 2018; 592:3201-3209. [PMID: 29802622 DOI: 10.1002/1873-3468.13115] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 05/21/2018] [Indexed: 12/15/2022]
Abstract
The Pannexin1 (Panx1) membrane channel responds to different stimuli with distinct channel conformations. Most stimuli induce a large cation- and ATP-permeable conformation, hence Panx1 is involved in many physiological processes entailing purinergic signaling. For example, oxygen delivery in the peripheral circulatory system is regulated by ATP released from red blood cells and endothelial cells through Panx1 channels. The same membrane channel, however, when stimulated by positive membrane potential or by cleavage with caspase 3, is highly selective for the passage of chloride ions, excluding cations and ATP. Although biophysical data do not allow a distinction between the chloride-selective channels induced by voltage or by caspase cleavage, there must be other subtle differences in the structure, because overexpression of wtPanx1 is well tolerated by cells, while expression of the truncation mutant Panx1Δ378 results in slow cell death. Thus, in addition to the well-characterized two open conformations, there might be a third, more subtle conformational change involved in cell death.
Collapse
Affiliation(s)
- Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami School of Medicine, FL, USA
| |
Collapse
|
21
|
Wang J, Dahl G. Pannexin1: a multifunction and multiconductance and/or permeability membrane channel. Am J Physiol Cell Physiol 2018; 315:C290-C299. [PMID: 29719171 DOI: 10.1152/ajpcell.00302.2017] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Of the three pannexins in vertebrate proteomes, pannexin1 (Panx1) is the only one well characterized, and it is generally accepted that Panx1 functions as an ATP release channel for signaling to other cells. However, the ATP permeability of the channel is only observed with certain stimuli, including low oxygen, mechanical stress, and elevated extracellular potassium ion concentration. Otherwise, the Panx1 channel is selective for chloride ions and exhibits no ATP permeability when stimulated simply by depolarization to positive potentials. A third, irreversible activation of Panx1 follows cleavage of carboxyterminal amino acids by caspase 3. The selectivity/permeability properties of the caspase cleaved channel are unclear as it reportedly has features of both channel conformations. Here we describe the biophysical properties of the channel formed by the truncation mutant Panx1Δ378, which is identical to the caspase-cleaved protein. Consistent with previous findings for the caspase-activated channel, the Panx1Δ378 channel was constitutively active. However, like the voltage-gated channel, the Panx1Δ378 channel had high chloride selectivity, lacked cation permeability, and did not mediate ATP release unless stimulated by extracellular potassium ions. Thus, the caspase-cleaved Panx1 channel should be impermeable to ATP, contrary to previous claims.
Collapse
Affiliation(s)
- Junjie Wang
- Department of Physiology and Biophysics, University of Miami School of Medicine , Miami, Florida
| | - Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami School of Medicine , Miami, Florida
| |
Collapse
|
22
|
Wang J, Jackson DG, Dahl G. Cationic control of Panx1 channel function. Am J Physiol Cell Physiol 2018; 315:C279-C289. [PMID: 29719168 DOI: 10.1152/ajpcell.00303.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The sequence and predicted membrane topology of pannexin1 (Panx1) places it in the family of gap junction proteins. However, rather than forming gap junction channels, Panx1 forms channels in the nonjunctional membrane. Panx1 operates in two distinct open states, depending on the mode of stimulation. The exclusively voltage-gated channel has a small conductance (<100 pS) and is highly selective for the flux of chloride ions. The Panx1 channel activated by various physiological stimuli or by increased concentrations of extracellular potassium ions has a large conductance (~500 pS, however, with multiple, long-lasting subconductance states) and is nonselectively permeable to small molecules, including ATP. To test whether the two open conformations also differ pharmacologically, the effects of di-and trivalent cations on the two Panx1 channel conformations were investigated. The rationale for this venture was that, under certain experimental conditions, ATP release from cells can be inhibited by multivalent cations, yet the literature indicates that the ATP release channel Panx1 is not affected by these ions. Consistent with previous reports, the Panx1 channel was not activated by removal of extracellular Ca2+ and the currents through the voltage-activated channel were not altered by Ca2+, Zn2+, Ba2+, or Gd3+. In contrast, the Panx1 channel activated to the large channel conformation by extracellular K+, osmotic stress, or low oxygen was inhibited by the multivalent cations in a dose-dependent way. Thus, monovalent cations activated the Panx1 channel from the closed state to the "large" conformation, while di- and trivalent cations exclusively inhibited this large channel conformation.
Collapse
Affiliation(s)
- Junjie Wang
- Department of Physiology and Biophysics, University of Miami School of Medicine , Miami, Florida
| | - David George Jackson
- Department of Physiology and Biophysics, University of Miami School of Medicine , Miami, Florida
| | - Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami School of Medicine , Miami, Florida
| |
Collapse
|
23
|
Abstract
Adenosine triphosphate (ATP) has been well established as an important extracellular ligand of autocrine signaling, intercellular communication, and neurotransmission with numerous physiological and pathophysiological roles. In addition to the classical exocytosis, non-vesicular mechanisms of cellular ATP release have been demonstrated in many cell types. Although large and negatively charged ATP molecules cannot diffuse across the lipid bilayer of the plasma membrane, conductive ATP release from the cytosol into the extracellular space is possible through ATP-permeable channels. Such channels must possess two minimum qualifications for ATP permeation: anion permeability and a large ion-conducting pore. Currently, five groups of channels are acknowledged as ATP-release channels: connexin hemichannels, pannexin 1, calcium homeostasis modulator 1 (CALHM1), volume-regulated anion channels (VRACs, also known as volume-sensitive outwardly rectifying (VSOR) anion channels), and maxi-anion channels (MACs). Recently, major breakthroughs have been made in the field by molecular identification of CALHM1 as the action potential-dependent ATP-release channel in taste bud cells, LRRC8s as components of VRACs, and SLCO2A1 as a core subunit of MACs. Here, the function and physiological roles of these five groups of ATP-release channels are summarized, along with a discussion on the future implications of understanding these channels.
Collapse
|
24
|
Age-Related Macular Degeneration: New Paradigms for Treatment and Management of AMD. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8374647. [PMID: 29484106 PMCID: PMC5816845 DOI: 10.1155/2018/8374647] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 12/06/2017] [Indexed: 12/03/2022]
Abstract
Age-related macular degeneration (AMD) is a well-characterized and extensively studied disease. It is currently considered the leading cause of visual disability among patients over 60 years. The hallmark of early AMD is the formation of drusen, pigmentary changes at the macula, and mild to moderate vision loss. There are two forms of AMD: the “dry” and the “wet” form that is less frequent but is responsible for 90% of acute blindness due to AMD. Risk factors have been associated with AMD progression, and they are taking relevance to understand how AMD develops: (1) advanced age and the exposition to environmental factors inducing high levels of oxidative stress damaging the macula and (2) this damage, which causes inflammation inducing a vicious cycle, altogether causing central vision loss. There is neither a cure nor treatment to prevent AMD. However, there are some treatments available for the wet form of AMD. This article will review some molecular and cellular mechanisms associated with the onset of AMD focusing on feasible treatments for each related factor in the development of this pathology such as vascular endothelial growth factor, oxidative stress, failure of the clearance of proteins and organelles, and glial cell dysfunction in AMD.
Collapse
|
25
|
Nomura T, Taruno A, Shiraishi M, Nakahari T, Inui T, Sokabe M, Eaton DC, Marunaka Y. Current-direction/amplitude-dependent single channel gating kinetics of mouse pannexin 1 channel: a new concept for gating kinetics. Sci Rep 2017; 7:10512. [PMID: 28874774 PMCID: PMC5585217 DOI: 10.1038/s41598-017-10921-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/16/2017] [Indexed: 11/25/2022] Open
Abstract
The detailed single-channel gating kinetics of mouse pannexin 1 (mPanx1) remains unknown, although mPanx1 is reported to be a voltage-activated anion-selective channel. We investigated characteristics of single-channel conductances and opening and closing rates of mPanx1 using patch-clamp techniques. The unitary current of mPanx1 shows outward rectification with single-channel conductances of ~20 pS for inward currents and ~80 pS for outward currents. The channel open time for outward currents (Cl- influx) increases linearly as the amplitude of single channel currents increases, while the open time for inward currents (Cl- efflux) is constant irrespective of changes in the current amplitude, as if the direction and amplitude of the unitary current regulates the open time. This is supported by further observations that replacement of extracellular Cl- with gluconate- diminishes the inward tail current (Cl- efflux) at a membrane potential of -100 mV due to the lowered outward current (gluconate- influx) at membrane potential of 100 mV. These results suggest that the direction and rate of charge-carrier movement regulate the open time of mPanx1, and that the previously reported voltage-dependence of Panx1 channel gating is not directly mediated by the membrane potential but rather by the direction and amplitude of currents through the channel.
Collapse
Affiliation(s)
- Takeshi Nomura
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, 602-8566, Japan
- Department of Bio-Ionomics, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, 602-8566, Japan
- Department of Physical Therapy, Faculty of Rehabilitation, Kyushu Nutrition Welfare University, Kitakyushu, 800-0298, Japan
| | - Akiyuki Taruno
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, 602-8566, Japan
| | - Makoto Shiraishi
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, 602-8566, Japan
| | - Takashi Nakahari
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, 602-8566, Japan
- Department of Bio-Ionomics, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, 602-8566, Japan
- Japan Institute for Food Education and Health, St. Agnes' University, Kyoto, 602-8013, Japan
| | - Toshio Inui
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, 602-8566, Japan
- Department of Bio-Ionomics, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, 602-8566, Japan
- Saisei Mirai Clinics, Moriguchi, 570-0012, Japan
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Douglas C Eaton
- Center for Cell & Molecular Signaling, Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Yoshinori Marunaka
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, 602-8566, Japan.
- Department of Bio-Ionomics, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, 602-8566, Japan.
- Japan Institute for Food Education and Health, St. Agnes' University, Kyoto, 602-8013, Japan.
| |
Collapse
|
26
|
Richard M, Bauer R, Tavosanis G, Hoch M. The gap junction protein Innexin3 is required for eye disc growth in Drosophila. Dev Biol 2017; 425:191-207. [PMID: 28390801 DOI: 10.1016/j.ydbio.2017.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 03/30/2017] [Accepted: 04/03/2017] [Indexed: 12/23/2022]
Abstract
The Drosophila compound eye develops from a bilayered epithelial sac composed of an upper peripodial epithelium layer and a lower disc proper, the latter giving rise to the eye itself. During larval stages, complex signalling events between the layers contribute to the control of cell proliferation and differentiation in the disc. Previous work in our lab established the gap junction protein Innexin2 (Inx2) as crucial for early larval eye disc growth. By analysing the contribution of other Innexins to eye size control, we have identified Innexin3 (Inx3) as an important growth regulator. Depleting inx3 during larval eye development reduces eye size, while elevating inx3 levels increases eye size, thus phenocopying the inx2 loss- and gain-of-function situation. As demonstrated previously for inx2, inx3 regulates disc cell proliferation and interacts genetically with the Dpp pathway, being required for the proper activation of the Dpp pathway transducer Mad at the furrow and the expression of Dpp receptor Punt in the eye disc. At the developmental timepoint corresponding to eye disc growth, Inx3 colocalises with Inx2 in disc proper and peripodial epithelium cell membranes. In addition, we show that Inx3 protein levels critically depend on inx2 throughout eye development and that inx3 modulates Inx2 protein levels in the larval eye disc. Rescue experiments demonstrate that Inx3 and Inx2 cooperate functionally to enable eye disc growth in Drosophila. Finally, we demonstrate that expression of Inx3 and Inx2 is not only needed in the disc proper but also in the peripodial epithelium to regulate growth of the eye disc. Our data provide a functional demonstration that putative Inx2/Inx3 heteromeric channels regulate organ size.
Collapse
Affiliation(s)
- Mélisande Richard
- Life & Medical Sciences Institute (LIMES) Development, Genetics & Molecular Physiology Unit University of Bonn, Carl-Troll-Straße, 31 53115 Bonn, Germany
| | - Reinhard Bauer
- Life & Medical Sciences Institute (LIMES) Development, Genetics & Molecular Physiology Unit University of Bonn, Carl-Troll-Straße, 31 53115 Bonn, Germany
| | - Gaia Tavosanis
- German Center for Neurodegenerative Diseases (DZNE), Dendrite Differentiation Unit, Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Michael Hoch
- Life & Medical Sciences Institute (LIMES) Development, Genetics & Molecular Physiology Unit University of Bonn, Carl-Troll-Straße, 31 53115 Bonn, Germany.
| |
Collapse
|
27
|
Kim Y, Davidson JO, Green CR, Nicholson LFB, O'Carroll SJ, Zhang J. Connexins and Pannexins in cerebral ischemia. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:224-236. [PMID: 28347700 DOI: 10.1016/j.bbamem.2017.03.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/24/2017] [Accepted: 03/22/2017] [Indexed: 12/12/2022]
Abstract
A common cause of mortality and long-term adult disability, cerebral ischemia or brain ischemia imposes a significant health and financial burden on communities worldwide. Cerebral ischemia is a condition that arises from a sudden loss of blood flow and consequent failure to meet the high metabolic demands of the brain. The lack of blood flow initiates a sequelae of cell death mechanisms, including the activation of the inflammatory pathway, which can ultimately result in irreversible brain tissue damage. In particular, Connexins and Pannexins are non-selective channels with a large pore that have shown to play time-dependent roles in the perpetuation of ischaemic injury. This review highlights the roles of Connexin and Pannexin channels in cell death mechanisms as a promising therapeutic target in cerebral ischemia, and in particular connexin hemichannels which may contribute most of the ATP release as a result of ischemia as well as during reperfusion. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Yeri Kim
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland
| | - Joanne O Davidson
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland
| | - Colin R Green
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland
| | - Louise F B Nicholson
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland
| | - Simon J O'Carroll
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland
| | - Jie Zhang
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland.
| |
Collapse
|
28
|
Meda P. Gap junction proteins are key drivers of endocrine function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:124-140. [PMID: 28284720 DOI: 10.1016/j.bbamem.2017.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 01/07/2023]
Abstract
It has long been known that the main secretory cells of exocrine and endocrine glands are connected by gap junctions, made by a variety of connexin species that ensure their electrical and metabolic coupling. Experiments in culture systems and animal models have since provided increasing evidence that connexin signaling contributes to control the biosynthesis and release of secretory products, as well as to the life and death of secretory cells. More recently, genetic studies have further provided the first lines of evidence that connexins also control the function of human glands, which are central to the pathogenesis of major endocrine diseases. Here, we summarize the recent information gathered on connexin signaling in these systems, since the last reviews on the topic, with particular regard to the pancreatic beta cells which produce insulin, and the renal cells which produce renin. These cells are keys to the development of various forms of diabetes and hypertension, respectively, and combine to account for the exploding, worldwide prevalence of the metabolic syndrome. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Switzerland.
| |
Collapse
|
29
|
Crespo Yanguas S, Willebrords J, Johnstone SR, Maes M, Decrock E, De Bock M, Leybaert L, Cogliati B, Vinken M. Pannexin1 as mediator of inflammation and cell death. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:51-61. [PMID: 27741412 DOI: 10.1016/j.bbamcr.2016.10.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 10/07/2016] [Accepted: 10/08/2016] [Indexed: 02/06/2023]
Abstract
Pannexins form channels at the plasma membrane surface that establish a pathway for communication between the cytosol of individual cells and their extracellular environment. By doing so, pannexin signaling dictates several physiological functions, but equally underlies a number of pathological processes. Indeed, pannexin channels drive inflammation by assisting in the activation of inflammasomes, the release of pro-inflammatory cytokines, and the activation and migration of leukocytes. Furthermore, these cellular pores facilitate cell death, including apoptosis, pyroptosis and autophagy. The present paper reviews the roles of pannexin channels in inflammation and cell death. In a first part, a state-of-the-art overview of pannexin channel structure, regulation and function is provided. In a second part, the mechanisms behind their involvement in inflammation and cell death are discussed.
Collapse
Affiliation(s)
- Sara Crespo Yanguas
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Joost Willebrords
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Scott R Johnstone
- College of Medical, Veterinary and Life Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Michaël Maes
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke Decrock
- Department of Basic Medical Sciences, Physiology group, Ghent University, Gent, Belgium
| | - Marijke De Bock
- Department of Basic Medical Sciences, Physiology group, Ghent University, Gent, Belgium
| | - Luc Leybaert
- Department of Basic Medical Sciences, Physiology group, Ghent University, Gent, Belgium
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
30
|
Seref-Ferlengez Z, Suadicani SO, Thi MM. A new perspective on mechanisms governing skeletal complications in type 1 diabetes. Ann N Y Acad Sci 2016; 1383:67-79. [PMID: 27571221 DOI: 10.1111/nyas.13202] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/11/2016] [Accepted: 07/18/2016] [Indexed: 12/29/2022]
Abstract
This review focuses on bone mechanobiology in type 1 diabetes (T1D), an area of research on diabetes-associated skeletal complications that is still in its infancy. We first provide a brief overview of the deleterious effects of diabetes on the skeleton and of the knowledge gained from studies with rodent models of T1D. Second, we discuss two specific hallmarks of T1D, low insulin and high glucose, and address the extent to which they affect skeletal health. Third, we highlight the mechanosensitive nature of bone tissue and the importance of mechanical loading for bone health. We also summarize recent advances in bone mechanobiology that implicate osteocytes as the mechanosensors and major regulatory cells in the bone. Finally, we discuss recent evidence indicating that the diabetic bone is "deaf" to mechanical loading and that osteocytes are central players in mechanisms that lead to bone loss in T1D.
Collapse
Affiliation(s)
- Zeynep Seref-Ferlengez
- Department of Orthopaedic Surgery.,Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE)
| | - Sylvia O Suadicani
- Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE).,Department of Neuroscience.,Department of Urology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York
| | - Mia M Thi
- Department of Orthopaedic Surgery.,Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE).,Department of Neuroscience
| |
Collapse
|
31
|
Abstract
Communication among cells via direct cell-cell contact by connexin gap junctions, or between cell and extracellular environment via pannexin channels or connexin hemichannels, is a key factor in cell function and tissue homeostasis. Upon malignant transformation in different cancer types, the dysregulation of these connexin and pannexin channels and their effect in cellular communication, can either enhance or suppress tumorigenesis and metastasis. In this review, we will highlight the latest reports on the role of the well characterized connexin family and its ability to form gap junctions and hemichannels in cancer. We will also introduce the more recently discovered family of pannexin channels and our current knowledge about their involvement in cancer progression.
Collapse
Affiliation(s)
- Jean X Jiang
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, N6A5C1, Canada.
| |
Collapse
|
32
|
Seref-Ferlengez Z, Maung S, Schaffler MB, Spray DC, Suadicani SO, Thi MM. P2X7R-Panx1 Complex Impairs Bone Mechanosignaling under High Glucose Levels Associated with Type-1 Diabetes. PLoS One 2016; 11:e0155107. [PMID: 27159053 PMCID: PMC4861344 DOI: 10.1371/journal.pone.0155107] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/25/2016] [Indexed: 12/21/2022] Open
Abstract
Type 1 diabetes (T1D) causes a range of skeletal problems, including reduced bone density and increased risk for bone fractures. However, mechanisms underlying skeletal complications in diabetes are still not well understood. We hypothesize that high glucose levels in T1D alters expression and function of purinergic receptors (P2Rs) and pannexin 1 (Panx1) channels, and thereby impairs ATP signaling that is essential for proper bone response to mechanical loading and maintenance of skeletal integrity. We first established a key role for P2X7 receptor-Panx1 in osteocyte mechanosignaling by showing that these proteins are co-expressed to provide a major pathway for flow-induced ATP release. To simulate in vitro the glucose levels to which bone cells are exposed in healthy vs. diabetic bones, we cultured osteoblast and osteocyte cell lines for 10 days in medium containing 5.5 or 25 mM glucose. High glucose effects on expression and function of P2Rs and Panx1 channels were determined by Western Blot analysis, quantification of Ca2+ responses to P2R agonists and oscillatory fluid shear stress (± 10 dyne/cm2), and measurement of flow-induced ATP release. Diabetic C57BL/6J-Ins2Akita mice were used to evaluate in vivo effects of high glucose on P2R and Panx1. Western blotting indicated altered P2X7R, P2Y2R and P2Y4R expression in high glucose exposed bone cells, and in diabetic bone tissue. Moreover, high glucose blunted normal P2R- and flow-induced Ca2+ signaling and ATP release from osteocytes. These findings indicate that T1D impairs load-induced ATP signaling in osteocytes and affects osteoblast function, which are essential for maintaining bone health.
Collapse
Affiliation(s)
- Zeynep Seref-Ferlengez
- Departments of Orthopaedic Surgery, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
- Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE), Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
| | - Stephanie Maung
- Departments of Orthopaedic Surgery, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
- Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE), Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
| | - Mitchell B. Schaffler
- Department of Biomedical Engineering, City College of New York, New York, NY, United States of America
| | - David C. Spray
- Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
- Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE), Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
| | - Sylvia O. Suadicani
- Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
- Department of Urology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
- Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE), Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
| | - Mia M. Thi
- Departments of Orthopaedic Surgery, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
- Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
- Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE), Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States of America
- * E-mail:
| |
Collapse
|
33
|
Oshima A, Matsuzawa T, Murata K, Tani K, Fujiyoshi Y. Hexadecameric structure of an invertebrate gap junction channel. J Mol Biol 2016; 428:1227-1236. [DOI: 10.1016/j.jmb.2016.02.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/05/2016] [Accepted: 02/08/2016] [Indexed: 12/12/2022]
|
34
|
Dahl G. ATP release through pannexon channels. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0191. [PMID: 26009770 DOI: 10.1098/rstb.2014.0191] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Extracellular adenosine triphosphate (ATP) serves as a signal for diverse physiological functions, including spread of calcium waves between astrocytes, control of vascular oxygen supply and control of ciliary beat in the airways. ATP can be released from cells by various mechanisms. This review focuses on channel-mediated ATP release and its main enabler, Pannexin1 (Panx1). Six subunits of Panx1 form a plasma membrane channel termed 'pannexon'. Depending on the mode of stimulation, the pannexon has large conductance (500 pS) and unselective permeability to molecules less than 1.5 kD or is a small (50 pS), chloride-selective channel. Most physiological and pathological stimuli induce the large channel conformation, whereas the small conformation so far has only been observed with exclusive voltage activation of the channel. The interaction between pannexons and ATP is intimate. The pannexon is not only the conduit for ATP, permitting ATP efflux from cells down its concentration gradient, but the pannexon is also modulated by ATP. The channel can be activated by ATP through both ionotropic P2X as well as metabotropic P2Y purinergic receptors. In the absence of a control mechanism, this positive feedback loop would lead to cell death owing to the linkage of purinergic receptors with apoptotic processes. A control mechanism preventing excessive activation of the purinergic receptors is provided by ATP binding (with low affinity) to the Panx1 protein and gating the channel shut.
Collapse
Affiliation(s)
- Gerhard Dahl
- School of Medicine, University of Miami, 1600 NW 10th Avenue, Miami, FL 33136, USA
| |
Collapse
|
35
|
Talaverón R, Fernández P, Escamilla R, Pastor AM, Matarredona ER, Sáez JC. Neural progenitor cells isolated from the subventricular zone present hemichannel activity and form functional gap junctions with glial cells. Front Cell Neurosci 2015; 9:411. [PMID: 26528139 PMCID: PMC4602088 DOI: 10.3389/fncel.2015.00411] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 09/25/2015] [Indexed: 01/19/2023] Open
Abstract
The postnatal subventricular zone (SVZ) lining the walls of the lateral ventricles contains neural progenitor cells (NPCs) that generate new olfactory bulb interneurons. Communication via gap junctions between cells in the SVZ is involved in NPC proliferation and in neuroblast migration towards the olfactory bulb. SVZ NPCs can be expanded in vitro in the form of neurospheres that can be used for transplantation purposes after brain injury. We have previously reported that neurosphere-derived NPCs form heterocellular gap junctions with host glial cells when they are implanted after mechanical injury. To analyze functionality of NPC-glial cell gap junctions we performed dye coupling experiments in co-cultures of SVZ NPCs with astrocytes or microglia. Neurosphere-derived cells expressed mRNA for at least the hemichannel/gap junction channel proteins connexin 26 (Cx26), Cx43, Cx45 and pannexin 1 (Panx1). Dye coupling experiments revealed that gap junctional communication occurred among neurosphere cells (incidence of coupling: 100%). Moreover, hemichannel activity was also detected in neurosphere cells as evaluated in time-lapse measurements of ethidium bromide uptake. Heterocellular coupling between NPCs and glial cells was evidenced in co-cultures of neurospheres with astrocytes (incidence of coupling: 91.0 ± 4.7%) or with microglia (incidence of coupling: 71.9 ± 6.7%). Dye coupling in neurospheres and in co-cultures was inhibited by octanol, a gap junction blocker. Altogether, these results suggest the existence of functional hemichannels and gap junction channels in postnatal SVZ neurospheres. In addition, they demonstrate that SVZ-derived NPCs can establish functional gap junctions with astrocytes or microglia. Therefore, cell-cell communication via gap junctions and hemichannels with host glial cells might subserve a role in the functional integration of NPCs after implantation in the damaged brain.
Collapse
Affiliation(s)
- Rocío Talaverón
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla Sevilla, Spain
| | - Paola Fernández
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile and Instituto Milenio, Centro Interdisciplinario de Neurociencias de Valparaíso Chile
| | - Rosalba Escamilla
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile and Instituto Milenio, Centro Interdisciplinario de Neurociencias de Valparaíso Chile
| | - Angel M Pastor
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla Sevilla, Spain
| | | | - Juan C Sáez
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile and Instituto Milenio, Centro Interdisciplinario de Neurociencias de Valparaíso Chile
| |
Collapse
|
36
|
Richard M, Hoch M. Drosophila eye size is determined by Innexin 2-dependent Decapentaplegic signalling. Dev Biol 2015; 408:26-40. [PMID: 26455410 DOI: 10.1016/j.ydbio.2015.10.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/23/2015] [Accepted: 10/06/2015] [Indexed: 12/21/2022]
Abstract
Organogenesis relies on specific genetic and molecular programmes, which orchestrate growth and cellular differentiation over developmental time. This is particularly important during Drosophila eye development in which cell-cell inductive events and long-range signalling have to be integrated to regulate proper cell proliferation, differentiation and morphogenesis. How these processes are coordinated is still not very well understood. Here we identify the gap junction protein Innexin2 (Inx2) as an important regulator of eye development. Depleting inx2 during eye development reduces eye size whereas elevating inx2 levels increases eye size. Loss- and gain-of-function experiments demonstrate that inx2 is required functionally in larval eye disc cells where it localises apico-laterally. inx2 regulates disc cell proliferation as well as morphogenetic furrow movement and as a result the amount of differentiated photoreceptors. inx2 interacts genetically with the Dpp pathway and we find that proper activation of the Dpp pathway transducer Mad at the furrow and expression of Dpp receptors Thickveins and Punt in the anterior disc compartment require inx2. We further show that inx2 is required for the transcriptional activation of dpp and punt in the eye disc. Our results highlight the crucial role of gap junction proteins in regulating morphogen-dependent organ size determination.
Collapse
Affiliation(s)
- Mélisande Richard
- Life & Medical Sciences Institute (LIMES) Development, Genetics & Molecular Physiology Unit, University of Bonn, Carl-Troll-Straße, 31, D-53115 Bonn, Germany.
| | - Michael Hoch
- Life & Medical Sciences Institute (LIMES) Development, Genetics & Molecular Physiology Unit, University of Bonn, Carl-Troll-Straße, 31, D-53115 Bonn, Germany.
| |
Collapse
|
37
|
Beckmann A, Grissmer A, Krause E, Tschernig T, Meier C. Pannexin-1 channels show distinct morphology and no gap junction characteristics in mammalian cells. Cell Tissue Res 2015; 363:751-63. [PMID: 26386583 DOI: 10.1007/s00441-015-2281-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/24/2015] [Indexed: 10/23/2022]
Abstract
Pannexins (Panx) are proteins with a similar membrane topology to connexins, the integral membrane protein of gap junctions. Panx1 channels are generally of major importance in a large number of system and cellular processes and their function has been thoroughly characterized. In contrast, little is known about channel structure and subcellular distribution. We therefore determine the subcellular localization of Panx1 channels in cultured cells and aim at the identification of channel morphology in vitro. Using freeze-fracture replica immunolabeling on EYFP-Panx1-overexpressing HEK 293 cells, large particles were identified in plasma membranes, which were immunogold-labeled using either GFP or Panx1 antibodies. There was no labeling or particles in the nuclear membranes of these cells, pointing to plasma membrane localization of Panx1-EYFP channels. The assembly of particles was irregular, this being in contrast to the regular pattern of gap junctions. The fact that no counterparts were identified on apposing cells, which would have been indicative of intercellular signaling, supported the idea of Panx1 channels within one membrane. Control cells (transfected with EYFP only, non-transfected) were devoid of both particles and immunogold labeling. Altogether, this study provides the first demonstration of Panx1 channel morphology and assembly in intact cells. The identification of Panx1 channels as large particles within the plasma membrane provides the knowledge required to enable recognition of Panx1 channels in tissues in future studies. Thus, these results open up new avenues for the detailed analysis of the subcellular localization of Panx1 and of its nearest neighbors such as purinergic receptors in vivo.
Collapse
Affiliation(s)
- Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66421, Homburg/Saar, Germany
| | - Alexander Grissmer
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66421, Homburg/Saar, Germany
| | - Elmar Krause
- Department of Physiology, Saarland University, 66421, Homburg/Saar, Germany
| | - Thomas Tschernig
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66421, Homburg/Saar, Germany
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66421, Homburg/Saar, Germany.
| |
Collapse
|
38
|
Kringelbach TM, Aslan D, Novak I, Ellegaard M, Syberg S, Andersen CKB, Kristiansen KA, Vang O, Schwarz P, Jørgensen NR. Fine-tuned ATP signals are acute mediators in osteocyte mechanotransduction. Cell Signal 2015; 27:2401-9. [PMID: 26327582 DOI: 10.1016/j.cellsig.2015.08.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 12/12/2022]
Abstract
Osteocytes are considered the primary mechanosensors of bone, but the signaling pathways they apply in mechanotransduction are still incompletely investigated and characterized. A growing body of data strongly indicates that P2 receptor signaling among osteoblasts and osteoclasts has regulatory effects on bone remodeling. Therefore, we hypothesized that ATP signaling is also applied by osteocytes in mechanotransduction. We applied a short fluid pulse on MLO-Y4 osteocyte-like cells during real-time detection of ATP and demonstrated that mechanical stimulation activates the acute release of ATP and that these acute ATP signals are fine-tuned according to the magnitude of loading. ATP release was then challenged by pharmacological inhibitors, which indicated a vesicular release pathway for acute ATP signals. Finally, we showed that osteocytes express functional P2X2 and P2X7 receptors and respond to even low concentrations of nucleotides by increasing intracellular calcium concentration. These results indicate that in osteocytes, vesicular ATP release is an acute mediator of mechanical signals and the magnitude of loading. These and previous results, therefore, implicate purinergic signaling as an early signaling pathway in osteocyte mechanotransduction.
Collapse
Affiliation(s)
- Tina M Kringelbach
- Research Center for Ageing and Osteoporosis, Department of Diagnostics, Copenhagen University Hospital Glostrup, Glostrup, Denmark; Research Center for Ageing and Osteoporosis, Department of Medicine, Copenhagen University Hospital Glostrup, Glostrup, Denmark; The Osteoporosis and Bone Metabolic Unit, Dept. of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; The Osteoporosis and Bone Metabolic Unit, Dept. of Clinical Biochemistry, Copenhagen, University Hospital Hvidovre, Hvidovre, Denmark
| | - Derya Aslan
- Research Center for Ageing and Osteoporosis, Department of Diagnostics, Copenhagen University Hospital Glostrup, Glostrup, Denmark; Research Center for Ageing and Osteoporosis, Department of Medicine, Copenhagen University Hospital Glostrup, Glostrup, Denmark
| | - Ivana Novak
- Department of Biology, University of Copenhagen, Copenhagen Ø, Denmark
| | - Maria Ellegaard
- Research Center for Ageing and Osteoporosis, Department of Diagnostics, Copenhagen University Hospital Glostrup, Glostrup, Denmark; Research Center for Ageing and Osteoporosis, Department of Medicine, Copenhagen University Hospital Glostrup, Glostrup, Denmark
| | - Susanne Syberg
- Research Center for Ageing and Osteoporosis, Department of Diagnostics, Copenhagen University Hospital Glostrup, Glostrup, Denmark; Research Center for Ageing and Osteoporosis, Department of Medicine, Copenhagen University Hospital Glostrup, Glostrup, Denmark
| | - Christina K B Andersen
- Research Center for Ageing and Osteoporosis, Department of Diagnostics, Copenhagen University Hospital Glostrup, Glostrup, Denmark; Research Center for Ageing and Osteoporosis, Department of Medicine, Copenhagen University Hospital Glostrup, Glostrup, Denmark
| | - Kim A Kristiansen
- Department of Clinical Research, Copenhagen University Hospital Glostrup, Glostrup, Denmark
| | - Ole Vang
- Department of Science, Systems and Models, Roskilde University, Roskilde, Denmark
| | - Peter Schwarz
- Research Center for Ageing and Osteoporosis, Department of Diagnostics, Copenhagen University Hospital Glostrup, Glostrup, Denmark; Research Center for Ageing and Osteoporosis, Department of Medicine, Copenhagen University Hospital Glostrup, Glostrup, Denmark; Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Niklas R Jørgensen
- Research Center for Ageing and Osteoporosis, Department of Diagnostics, Copenhagen University Hospital Glostrup, Glostrup, Denmark; Research Center for Ageing and Osteoporosis, Department of Medicine, Copenhagen University Hospital Glostrup, Glostrup, Denmark; The Osteoporosis and Bone Metabolic Unit, Dept. of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; The Osteoporosis and Bone Metabolic Unit, Dept. of Clinical Biochemistry, Copenhagen, University Hospital Hvidovre, Hvidovre, Denmark.
| |
Collapse
|
39
|
Di Cesare Mannelli L, Marcoli M, Micheli L, Zanardelli M, Maura G, Ghelardini C, Cervetto C. Oxaliplatin evokes P2X7-dependent glutamate release in the cerebral cortex: A pain mechanism mediated by Pannexin 1. Neuropharmacology 2015; 97:133-41. [PMID: 26071109 DOI: 10.1016/j.neuropharm.2015.05.037] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/11/2015] [Accepted: 05/27/2015] [Indexed: 01/29/2023]
Abstract
Anticancer therapy based on the repeated administration of oxaliplatin is limited by the development of a neuropathic syndrome difficult to treat. Oxaliplatin neurotoxicity is based on complex nervous mechanisms, the comprehension of the role of single neurotransmitters and the knowledge of the signal flow among cells is matter of importance to improve therapeutic chances. In a rat model of oxaliplatin-induced neuropathy, we report increased P2X7-evoked glutamate release from cerebrocortical synaptosomes. The release was abolished by the P2X7 receptor (P2X7R) antagonists Brilliant-Blue-G (BBG) and A-438079, and significantly reduced by Carbenoxolone and the Pannexin 1 (Panx1) selective inhibitors Erioglaucine and (10)Panx suggesting the recruitment of Panx1. Aimed to evaluate the significance of P2X7R-Panx1 system activation in pain generated by oxaliplatin, pharmacological modulators were spinally infused by intrathecal catheter in oxaliplatin-treated animals. BBG, Erioglaucine and (10)Panx reverted oxaliplatin-dependent pain. Finally, the influence of the P2X7R-Panx1 system blockade on oxaliplatin anticancer activity was evaluated on the human colon cancer cell line HT-29. Prevention of HT-29 apoptosis and mortality was dependent by kind and concentration of P2X7R antagonists. On the contrary, the inhibition of Panx1 did not alter oxaliplatin lethality in tumor cells. It is concluded that glutamate release dependent on P2X7R is increased in cerebrocortical nerve terminals from oxaliplatin-treated rats; the increase is mediated by functional recruitment of Panx1; P2X7R antagonists and Panx1 inhibitors revert oxaliplatin-induced neuropathic pain; Panx1 inhibitors do not alter the oxaliplatin-induced mortality of cancer cells HT-29. The inhibition of Panx1 channel is suggested as a new and safe pharmacological target.
Collapse
Affiliation(s)
- Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy.
| | - Manuela Marcoli
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genova, Genova, Italy.
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Matteo Zanardelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Guido Maura
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genova, Genova, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Chiara Cervetto
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genova, Genova, Italy
| |
Collapse
|
40
|
Higgins G, Ringholz F, Buchanan P, McNally P, Urbach V. Physiological impact of abnormal lipoxin A₄ production on cystic fibrosis airway epithelium and therapeutic potential. BIOMED RESEARCH INTERNATIONAL 2015; 2015:781087. [PMID: 25866809 PMCID: PMC4383482 DOI: 10.1155/2015/781087] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 12/15/2022]
Abstract
Lipoxin A4 has been described as a major signal for the resolution of inflammation and is abnormally produced in the lungs of patients with cystic fibrosis (CF). In CF, the loss of chloride transport caused by the mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel gene results in dehydration, mucus plugging, and reduction of the airway surface liquid layer (ASL) height which favour chronic lung infection and neutrophil based inflammation leading to progressive lung destruction and early death of people with CF. This review highlights the unique ability of LXA4 to restore airway surface hydration, to stimulate airway epithelial repair, and to antagonise the proinflammatory program of the CF airway, circumventing some of the most difficult aspects of CF pathophysiology. The report points out novel aspects of the cellular mechanism involved in the physiological response to LXA4, including release of ATP from airway epithelial cell via pannexin channel and subsequent activation of and P2Y11 purinoreceptor. Therefore, inadequate endogenous LXA4 biosynthesis reported in CF exacerbates the ion transport abnormality and defective mucociliary clearance, in addition to impairing the resolution of inflammation, thus amplifying the vicious circle of airway dehydration, chronic infection, and inflammation.
Collapse
Affiliation(s)
- Gerard Higgins
- National Children's Research Centre, Crumlin, Dublin 12, Ireland
- Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Fiona Ringholz
- National Children's Research Centre, Crumlin, Dublin 12, Ireland
- Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Paul Buchanan
- National Children's Research Centre, Crumlin, Dublin 12, Ireland
| | - Paul McNally
- National Children's Research Centre, Crumlin, Dublin 12, Ireland
| | - Valérie Urbach
- National Children's Research Centre, Crumlin, Dublin 12, Ireland
- Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
- Institut National de la Santé et de la Recherche Médicale, U845, Faculté de Médecine Paris Descartes, Site Necker, 156 rue Vaugirard, 75015 Paris, France
| |
Collapse
|
41
|
Effects on channel properties and induction of cell death induced by c-terminal truncations of pannexin1 depend on domain length. J Membr Biol 2015; 248:285-94. [PMID: 25567359 DOI: 10.1007/s00232-014-9767-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/22/2014] [Indexed: 10/24/2022]
Abstract
Pannexin1 (Panx1) is an integral membrane protein and known to form multifunctional hexameric channels. Recently, Panx1 was identified to be responsible for the release of ATP and UTP from apoptotic cells after site-specific proteolysis by caspases 3/7. Cleavage at the carboxy-terminal (CT) position aa 376-379 irreversibly opens human Panx1 channels and leads to the release of the respective nucleotides resulting in recruitment of macrophages and in subsequent activation of the immunologic response. The fact that cleavage of the CT at this particular residues terminates in a permanently open channel raised the issue of functional relevance of the CT of Panx1 for regulating channel properties. To analyze the impact of the CT on channel gating, we generated 14 truncated versions of rat Panx1 cleaved at different positions in the C-terminus. This allowed elaboration of the influence of defined residues on channel formation, voltage-dependent gating, execution of cell mortality, and susceptibility to the Panx1 inhibitor carbenoxolone. We demonstrate that expression of Panx1 proteins, which were truncated to lengths between 370 and 393 residues, induces differential effects after expression in Xenopus laevis oocytes as well as in Neuro2A cells with strongest impact downstream the caspase 3/7 cleavage site.
Collapse
|
42
|
Palacios-Prado N, Huetteroth W, Pereda AE. Hemichannel composition and electrical synaptic transmission: molecular diversity and its implications for electrical rectification. Front Cell Neurosci 2014; 8:324. [PMID: 25360082 PMCID: PMC4197764 DOI: 10.3389/fncel.2014.00324] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/26/2014] [Indexed: 11/29/2022] Open
Abstract
Unapposed hemichannels (HCs) formed by hexamers of gap junction proteins are now known to be involved in various cellular processes under both physiological and pathological conditions. On the other hand, less is known regarding how differences in the molecular composition of HCs impact electrical synaptic transmission between neurons when they form intercellular heterotypic gap junctions (GJs). Here we review data indicating that molecular differences between apposed HCs at electrical synapses are generally associated with rectification of electrical transmission. Furthermore, this association has been observed at both innexin and connexin (Cx) based electrical synapses. We discuss the possible molecular mechanisms underlying electrical rectification, as well as the potential contribution of intracellular soluble factors to this phenomenon. We conclude that asymmetries in molecular composition and sensitivity to cellular factors of each contributing hemichannel can profoundly influence the transmission of electrical signals, endowing electrical synapses with more complex functional properties.
Collapse
Affiliation(s)
- Nicolás Palacios-Prado
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine Bronx, NY, USA ; Marine Biological Laboratory, Woods Hole Massachusetts, MA, USA
| | - Wolf Huetteroth
- Marine Biological Laboratory, Woods Hole Massachusetts, MA, USA ; Department of Neurobiology, University of Konstanz Konstanz, Germany
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine Bronx, NY, USA ; Marine Biological Laboratory, Woods Hole Massachusetts, MA, USA
| |
Collapse
|
43
|
Higgins G, Buchanan P, Perriere M, Al-Alawi M, Costello RW, Verriere V, McNally P, Harvey BJ, Urbach V. Activation of P2RY11 and ATP release by lipoxin A4 restores the airway surface liquid layer and epithelial repair in cystic fibrosis. Am J Respir Cell Mol Biol 2014; 51:178-90. [PMID: 24588705 DOI: 10.1165/rcmb.2012-0424oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In cystic fibrosis (CF), the airway surface liquid (ASL) height is reduced as a result of impaired ion transport, which favors bacterial colonization and inflammation of the airway and leads to progressive lung destruction. Lipoxin (LX)A4, which promotes resolution of inflammation, is inadequately produced in the airways of patients with CF. We previously demonstrated that LXA4 stimulates an ASL height increase and epithelial repair. Here we report the molecular mechanisms involved in these processes. We found that LXA4 (1 nM) induced an apical ATP release from non-CF (NuLi-1) and CF (CuFi-1) airway epithelial cell lines and CF primary cultures. The ATP release induced by LXA4 was completely inhibited by antagonists of the ALX/FPR2 receptor and Pannexin-1 channels. LXA4 induced an increase in intracellular cAMP and calcium, which were abolished by the selective inhibition of the P2RY11 purinoreceptor. Pannexin-1 and ATP hydrolysis inhibition and P2RY11 purinoreceptor knockdown all abolished the increase of ASL height induced by LXA4. Inhibition of the A2b adenosine receptor did not affect the ASL height increase induced by LXA4, whereas the PKA inhibitor partially inhibited this response. The stimulation of NuLi-1 and CuFi-1 cell proliferation, migration, and wound repair by LXA4 was inhibited by the antagonists of Pannexin-1 channel and P2RY11 purinoreceptor. Taken together, our results provide evidence for a novel role of LXA4 in stimulating apical ATP secretion via Pannexin-1 channels and P2RY11 purinoreceptors activation leading to an ASL height increase and epithelial repair.
Collapse
|
44
|
Sangaletti R, Dahl G, Bianchi L. Mechanosensitive unpaired innexin channels in C. elegans touch neurons. Am J Physiol Cell Physiol 2014; 307:C966-77. [PMID: 25252948 DOI: 10.1152/ajpcell.00246.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Invertebrate innexin proteins share sequence homology with vertebrate pannexins and general membrane topology with both pannexins and connexins. While connexins form gap junctions that mediate intercellular communication, pannexins are thought to function exclusively as plasma membrane channels permeable to both ions and small molecules. Undoubtedly, certain innexins function as gap junction proteins. However, due to sequence similarity to pannexins, it was postulated that innexins also function as plasma membrane channels. Indeed, some of the leech innexins were found to mediate ATP release as unpaired membrane channels with shared pharmacology to pannexin channels. We show here that Caenorhabditis elegans touch-sensing neurons express a mechanically gated innexin channel with a conductance of ∼1 nS and voltage-dependent and K(+)-selective subconductance state. We also show that C. elegans touch neurons take up ethidium bromide through a mechanism that is activated and blocked by innexin activating stimuli and inhibitors, respectively. Finally, we present evidence that touch neurons' innexins are required for cell death induced by chemical ischemia. Our work demonstrates that innexins function as plasma membrane channels in native C. elegans neurons, where they may play a role in pathological cell death.
Collapse
Affiliation(s)
- Rachele Sangaletti
- Department of Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, Florida
| | - Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, Florida
| | - Laura Bianchi
- Department of Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, Florida
| |
Collapse
|
45
|
Momboisse F, Olivares MJ, Báez-Matus X, Guerra MJ, Flores-Muñoz C, Sáez JC, Martínez AD, Cárdenas AM. Pannexin 1 channels: new actors in the regulation of catecholamine release from adrenal chromaffin cells. Front Cell Neurosci 2014; 8:270. [PMID: 25237296 PMCID: PMC4154466 DOI: 10.3389/fncel.2014.00270] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 08/20/2014] [Indexed: 11/13/2022] Open
Abstract
Chromaffin cells of the adrenal gland medulla synthesize and store hormones and peptides, which are released into the blood circulation in response to stress. Among them, adrenaline is critical for the fight-or-flight response. This neurosecretory process is highly regulated and depends on cytosolic [Ca2+]. By forming channels at the plasma membrane, pannexin-1 (Panx1) is a protein involved in many physiological and pathological processes amplifying ATP release and/or Ca2+ signals. Here, we show that Panx1 is expressed in the adrenal gland where it plays a role by regulating the release of catecholamines. In fact, inhibitors of Panx1 channels, such as carbenoxolone (Cbx) and probenecid, reduced the secretory activity induced with the nicotinic agonist 1,1-dimethyl-4-phenyl-piperazinium (DMPP, 50 μM) in whole adrenal glands. A similar inhibitory effect was observed in single chromaffin cells using Cbx or 10Panx1 peptide, another Panx1 channel inhibitors. Given that the secretory response depends on cytosolic [Ca2+] and Panx1 channels are permeable to Ca2+, we studied the possible implication of Panx1 channels in the Ca2+ signaling occurring during the secretory process. In support of this possibility, Panx1 channel inhibitors significantly reduced the Ca2+ signals evoked by DMPP in single chromaffin cells. However, the Ca2+ signals induced by caffeine in the absence of extracellular Ca2+ was not affected by Panx1 channel inhibitors, suggesting that this mechanism does not involve Ca2+ release from the endoplasmic reticulum. Conversely, Panx1 inhibitors significantly blocked the DMPP-induce dye uptake, supporting the idea that Panx1 forms functional channels at the plasma membrane. These findings indicate that Panx1 channels participate in the control the Ca2+ signal that triggers the secretory response of adrenal chromaffin cells. This mechanism could have physiological implications during the response to stress.
Collapse
Affiliation(s)
- Fanny Momboisse
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - María José Olivares
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Ximena Báez-Matus
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - María José Guerra
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Juan C Sáez
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso Valparaíso, Chile ; Departamento de Fisiología, Pontifícia Universidad Católica de Chile Santiago, Chile
| | - Agustín D Martínez
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Ana M Cárdenas
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| |
Collapse
|
46
|
Takeuchi H, Suzumura A. Gap junctions and hemichannels composed of connexins: potential therapeutic targets for neurodegenerative diseases. Front Cell Neurosci 2014; 8:189. [PMID: 25228858 PMCID: PMC4151093 DOI: 10.3389/fncel.2014.00189] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/19/2014] [Indexed: 12/03/2022] Open
Abstract
Microglia are macrophage-like resident immune cells that contribute to the maintenance of homeostasis in the central nervous system (CNS). Abnormal activation of microglia can cause damage in the CNS, and accumulation of activated microglia is a characteristic pathological observation in neurologic conditions such as trauma, stroke, inflammation, epilepsy, and neurodegenerative diseases. Activated microglia secrete high levels of glutamate, which damages CNS cells and has been implicated as a major cause of neurodegeneration in these conditions. Glutamate-receptor blockers and microglia inhibitors (e.g., minocycline) have been examined as therapeutic candidates for several neurodegenerative diseases; however, these compounds exerted little therapeutic benefit because they either perturbed physiological glutamate signals or suppressed the actions of protective microglia. The ideal therapeutic approach would hamper the deleterious roles of activated microglia without diminishing their protective effects. We recently found that abnormally activated microglia secrete glutamate via gap-junction hemichannels on the cell surface. Moreover, administration of gap-junction inhibitors significantly suppressed excessive microglial glutamate release and improved disease symptoms in animal models of neurologic conditions such as stroke, multiple sclerosis, amyotrophic lateral sclerosis, and Alzheimer's disease. Recent evidence also suggests that neuronal and glial communication via gap junctions amplifies neuroinflammation and neurodegeneration. Elucidation of the precise pathologic roles of gap junctions and hemichannels may lead to a novel therapeutic strategies that can slow and halt the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University Nagoya, Japan
| | - Akio Suzumura
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University Nagoya, Japan
| |
Collapse
|
47
|
Wang J, Ambrosi C, Qiu F, Jackson DG, Sosinsky G, Dahl G. The membrane protein Pannexin1 forms two open-channel conformations depending on the mode of activation. Sci Signal 2014; 7:ra69. [PMID: 25056878 DOI: 10.1126/scisignal.2005431] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pannexin1 (Panx1) participates in several signaling events that involve adenosine triphosphate (ATP) release, including the innate immune response, ciliary beat in airway epithelia, and oxygen supply in the vasculature. The view that Panx1 forms a large ATP release channel has been challenged by the association of a low-conductance, small anion-selective channel with the presence of Panx1. We showed that Panx1 membrane channels can function in two distinct modes with different conductances and permeabilities when heterologously expressed in Xenopus oocytes. When stimulated by potassium ions (K(+)), Panx1 formed a high-conductance channel of ~500 pS that was permeable to ATP. Various physiological stimuli can induce this ATP-permeable conformation of the channel in several cell types. In contrast, the channel had a low conductance (~50 pS) with no detectable ATP permeability when activated by voltage in the absence of K(+). The two channel states were associated with different reactivities of the terminal cysteine of Panx1 to thiol reagents, suggesting different conformations. Single-particle electron microscopic analysis revealed that K(+) stimulated the formation of channels with a larger pore diameter than those formed in the absence of K(+). These data suggest that different stimuli lead to distinct channel structures with distinct biophysical properties.
Collapse
Affiliation(s)
- Junjie Wang
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, FL 33136, USA
| | - Cinzia Ambrosi
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California, San Diego, La Jolla, CA 92093-06083, USA
| | - Feng Qiu
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, FL 33136, USA
| | - David G Jackson
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, FL 33136, USA
| | - Gina Sosinsky
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California, San Diego, La Jolla, CA 92093-06083, USA. Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093-06083, USA
| | - Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
48
|
Gaynullina D, Tarasova OS, Kiryukhina OO, Shestopalov VI, Panchin Y. Endothelial function is impaired in conduit arteries of pannexin1 knockout mice. Biol Direct 2014; 9:8. [PMID: 24885326 PMCID: PMC4046076 DOI: 10.1186/1745-6150-9-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 05/14/2014] [Indexed: 11/21/2022] Open
Abstract
Pannexin1 is ubiquitously expressed in vertebrate tissues, but the role it plays in vascular tone regulation remains unclear. We found that Pannexin1 expression level is much higher in the endothelium relative to smooth muscle of saphenous artery. The ability of endothelium-intact arteries for dilation was significantly impaired whereas contractile responses were considerably increased in mice with genetic ablation of Pannexin1. No such increased contractile responses were detected in the endothelium-denuded arteries. Combined, our findings suggest a new function of Pannexin1 as an important player in normal endothelium-dependent regulation of arterial tone, where it facilitates vessel dilation and attenuates constriction. Reviewed by Dr. Armen Mulkidjanian and Dr. Alexander Lobkovsky.
Collapse
Affiliation(s)
- Dina Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M,V, Lomonosov Moscow State University, Leninskie Gory 1-12, 119234 Moscow, Russia.
| | | | | | | | | |
Collapse
|
49
|
Billaud M, Lohman AW, Johnstone SR, Biwer LA, Mutchler S, Isakson BE. Regulation of cellular communication by signaling microdomains in the blood vessel wall. Pharmacol Rev 2014; 66:513-69. [PMID: 24671377 DOI: 10.1124/pr.112.007351] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It has become increasingly clear that the accumulation of proteins in specific regions of the plasma membrane can facilitate cellular communication. These regions, termed signaling microdomains, are found throughout the blood vessel wall where cellular communication, both within and between cell types, must be tightly regulated to maintain proper vascular function. We will define a cellular signaling microdomain and apply this definition to the plethora of means by which cellular communication has been hypothesized to occur in the blood vessel wall. To that end, we make a case for three broad areas of cellular communication where signaling microdomains could play an important role: 1) paracrine release of free radicals and gaseous molecules such as nitric oxide and reactive oxygen species; 2) role of ion channels including gap junctions and potassium channels, especially those associated with the endothelium-derived hyperpolarization mediated signaling, and lastly, 3) mechanism of exocytosis that has considerable oversight by signaling microdomains, especially those associated with the release of von Willebrand factor. When summed, we believe that it is clear that the organization and regulation of signaling microdomains is an essential component to vessel wall function.
Collapse
Affiliation(s)
- Marie Billaud
- Dept. of Molecular Physiology and Biophysics, University of Virginia School of Medicine, PO Box 801394, Charlottesville, VA 22902.
| | | | | | | | | | | |
Collapse
|
50
|
Innexin and pannexin channels and their signaling. FEBS Lett 2014; 588:1396-402. [PMID: 24632288 DOI: 10.1016/j.febslet.2014.03.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 03/06/2014] [Indexed: 01/24/2023]
Abstract
Innexins are bifunctional membrane proteins in invertebrates, forming gap junctions as well as non-junctional membrane channels (innexons). Their vertebrate analogues, the pannexins, have not only lost the ability to form gap junctions but are also prevented from it by glycosylation. Pannexins appear to form only non-junctional membrane channels (pannexons). The membrane channels formed by pannexins and innexins are similar in their biophysical and pharmacological properties. Innexons and pannexons are permeable to ATP, are present in glial cells, and are involved in activation of microglia by calcium waves in glia. Directional movement and accumulation of microglia following nerve injury, which has been studied in the leech which has unusually large glial cells, involves at least 3 signals: ATP is the "go" signal, NO is the "where" signal and arachidonic acid is a "stop" signal.
Collapse
|