1
|
Paul A, Chumbale SS, Lakra A, Kumar V, Alhat DS, Singh S. Insights into Leishmania donovani potassium channel family and their biological functions. 3 Biotech 2023; 13:266. [PMID: 37425093 PMCID: PMC10326225 DOI: 10.1007/s13205-023-03692-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 06/26/2023] [Indexed: 07/11/2023] Open
Abstract
Leishmania donovani is the causative organism for visceral leishmaniasis. Although this parasite was discovered over a century ago, nothing is known about role of potassium channels in L. donovani. Potassium channels are known for their crucial roles in cellular functions in other organisms. Recently the presence of a calcium-activated potassium channel in L. donovani was reported which prompted us to look for other proteins which could be potassium channels and to investigate their possible physiological roles. Twenty sequences were identified in L. donovani genome and subjected to estimation of physio-chemical properties, motif analysis, localization prediction and transmembrane domain analysis. Structural predictions were also done. The channels were majorly α-helical and predominantly localized in cell membrane and lysosomes. The signature selectivity filter of potassium channel was present in all the sequences. In addition to the conventional potassium channel activity, they were associated with gene ontology terms for mitotic cell cycle, cell death, modulation by virus of host process, cell motility etc. The entire study indicates the presence of potassium channel families in L. donovani which may have involvement in several cellular pathways. Further investigations on these putative potassium channels are needed to elucidate their roles in Leishmania. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03692-y.
Collapse
Affiliation(s)
- Anindita Paul
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| | - Shubham Sunil Chumbale
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| | - Anjana Lakra
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| | - Vijay Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| | - Dhanashri Sudam Alhat
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| | - Sushma Singh
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| |
Collapse
|
2
|
Choudhary S, Kaur R, Waziri A, Garg A, Kadian R, Alam MS. N-type calcium channel blockers: a new approach towards the treatment of chronic neuropathic pain. EXPLORATION OF MEDICINE 2023. [DOI: 10.37349/emed.2023.00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Neuropathic pain (NP) remains maltreated for a wide number of patients by the currently available treatments and little research has been done in finding new drugs for treating NP. Ziconotide (PrialtTM) had been developed as the new drug, which belongs to the class of ω-conotoxin MVIIA. It inhibits N-type calcium channels. Ziconotide is under the last phase of the clinical trial, a new non-narcotic drug for the management of NP. Synthetically it has shown the similarities with ω-conotoxin MVIIA, a constituent of poison found in fish hunting snails (Conus magus). Ziconotide acts by selectively blocking neural N-type voltage-sensitized Ca2+ channels (NVSCCs). Certain herbal drugs also have been studied but no clinical result is there and the study is only limited to preclinical data. This review emphasizes the N-type calcium channel inhibitors, and their mechanisms for blocking calcium channels with their remedial prospects for treating chronic NP.
Collapse
Affiliation(s)
- Shikha Choudhary
- School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram 122103, Haryana, India
| | - Raminderjit Kaur
- School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram 122103, Haryana, India
| | - Aafrin Waziri
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Delhi, 110078, India
| | - Arun Garg
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University, Jaipur 303121, Rajasthan, India
| | - Renu Kadian
- Ram Gopal College of Pharmacy, Gurugram 122506, Haryana, India
| | - Md Sabir Alam
- SGT College of Pharmacy, SGT University, Gurugram 122505, Haryana, India
| |
Collapse
|
3
|
Harding EK, Souza IA, Gandini MA, Gadotti VM, Ali MY, Huang S, Antunes FTT, Trang T, Zamponi GW. Differential regulation of Ca v 3.2 and Ca v 2.2 calcium channels by CB 1 receptors and cannabidiol. Br J Pharmacol 2023; 180:1616-1633. [PMID: 36647671 DOI: 10.1111/bph.16035] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/02/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND PURPOSE Cannabinoids are a promising therapeutic avenue for chronic pain. However, clinical trials often fail to report analgesic efficacy of cannabinoids. Inhibition of voltage gate calcium (Cav ) channels is one mechanism through which cannabinoids may produce analgesia. We hypothesized that cannabinoids and cannabinoid receptor agonists target different types of Cav channels through distinct mechanisms. EXPERIMENTAL APPROACH Electrophysiological recordings from tsA-201 cells expressing either Cav 3.2 or Cav 2.2 were used to assess inhibition by HU-210 or cannabidiol (CBD) in the absence and presence of the CB1 receptor. Homology modelling assessed potential interaction sites for CBD in both Cav 2.2 and Cav 3.2. Analgesic effects of CBD were assessed in mouse models of inflammatory and neuropathic pain. KEY RESULTS HU-210 (1 μM) inhibited Cav 2.2 function in the presence of CB1 receptor but had no effect on Cav 3.2 regardless of co-expression of CB1 receptor. By contrast, CBD (3 μM) produced no inhibition of Cav 2.2 and instead inhibited Cav 3.2 independently of CB1 receptors. Homology modelling supported these findings, indicating that CBD binds to and occludes the pore of Cav 3.2, but not Cav 2.2. Intrathecal CBD alleviated thermal and mechanical hypersensitivity in both male and female mice, and this effect was absent in Cav 3.2 null mice. CONCLUSION AND IMPLICATIONS Our findings reveal differential modulation of Cav 2.2 and Cav 3.2 channels by CB1 receptors and CBD. This advances our understanding of how different cannabinoids produce analgesia through action at different voltage-gated calcium channels and could influence the development of novel cannabinoid-based therapeutics for treatment of chronic pain.
Collapse
Affiliation(s)
- Erika K Harding
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Clinical Neurosciences, and Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Ivana A Souza
- Department of Clinical Neurosciences, and Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Maria A Gandini
- Department of Clinical Neurosciences, and Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Vinícius M Gadotti
- Department of Clinical Neurosciences, and Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Zymedyne Therapeutics, Calgary, AB, Canada
| | - Md Yousof Ali
- Department of Clinical Neurosciences, and Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Zymedyne Therapeutics, Calgary, AB, Canada
| | - Sun Huang
- Department of Clinical Neurosciences, and Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Flavia T T Antunes
- Department of Clinical Neurosciences, and Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Tuan Trang
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Clinical Neurosciences, and Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, and Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
4
|
Wołyniak M, Małecka-Wojciesko E, Zielińska M, Fabisiak A. A Crosstalk between the Cannabinoid Receptors and Nociceptin Receptors in Colitis-Clinical Implications. J Clin Med 2022; 11:jcm11226675. [PMID: 36431153 PMCID: PMC9696262 DOI: 10.3390/jcm11226675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammatory bowel diseases (IBD) refer to a group of gastrointestinal (GI) disorders with complex pathogenesis characterized by chronic intestinal inflammation with a variety of symptoms. Cannabinoid and nociceptin opioid receptors (NOPs) and their ligands are widely distributed in the GI tract. The nociceptin opioid receptor is a newly discovered member of the opioid receptor family with unique characteristics. Both cannabinoid and NOP systems exhibit antinociceptive and anti-inflammatory activity and contribute to maintaining proper motility, secretion and absorption in the GI tract. Furthermore, they influence high and low voltage calcium channels, which play a crucial role in the processing of pain, and share at least two kinases mediating their action. Among them there is NF-κB, a key factor in the regulation of inflammatory processes. Therefore, based on functional similarities between cannabinoid and nociceptin receptors and the anti-inflammatory effects exerted by their ligands, there is a high likelihood that there is an interaction between cannabinoid receptors 1 and 2 and the nociceptin receptor in colitis. In this review, we discuss potential overlaps between these two systems on a molecular and functional level in intestinal inflammation to create the basis for novel treatments of IBD.
Collapse
Affiliation(s)
- Maria Wołyniak
- Department of Digestive Tract Diseases, Faculty of Medicine, Medical University of Lodz, 90-153 Lodz, Poland
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, 92-215 Lodz, Poland
| | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Faculty of Medicine, Medical University of Lodz, 90-153 Lodz, Poland
| | - Marta Zielińska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, 92-215 Lodz, Poland
| | - Adam Fabisiak
- Department of Digestive Tract Diseases, Faculty of Medicine, Medical University of Lodz, 90-153 Lodz, Poland
- Correspondence: ; Tel.: +48-42-677-66-64
| |
Collapse
|
5
|
Denomme MM, Haywood ME, Parks JC, Schoolcraft WB, Katz‐Jaffe MG. The inherited methylome landscape is directly altered with paternal aging and associated with offspring neurodevelopmental disorders. Aging Cell 2020; 19:e13178. [PMID: 32610362 PMCID: PMC7431824 DOI: 10.1111/acel.13178] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/22/2020] [Accepted: 05/27/2020] [Indexed: 12/11/2022] Open
Abstract
Paternal aging and the prevalence of neurodevelopmental disorders in offspring are well documented. Yet, the underlying mechanism and the mode of inheritance have not been conclusively established. Advancing paternal age is a subtle and varying phenotype. As such, it is likely that a threshold for cumulative risk may exist that, if surpassed, culminates in a predisposition to disease and ultimately an observed phenotype in offspring. Epigenetic regulation provides a plausible explanation for the nongenetic paternal transmission of disease susceptibility. With the use of whole‐genome methylation sequencing, the data described herein substantiate an increasingly compromised DNA methylation profile as sperm ages and, for the first time, also demonstrate a generational correlation in sperm and blastocyst of an altered methylome associated with advanced paternal age. Methylation alterations are not randomly distributed across the genome, but appear clustered at certain chromosomal locations, and significantly colocalize with regions of nucleosome retention. Genes associated with autism spectrum disorder, schizophrenia, and bipolar disorder are significantly enriched with causative methylation aberrations in both sperm and embryos from aged fathers. The long‐term health burden and societal economic impact of these conditions are substantial and will continue with increasingly prevalent diagnosis. This work provides a mechanistic link between the paternal age effect and offspring neurodevelopmental disorders leading to a better understanding of causation and investigation into potential future therapy.
Collapse
Affiliation(s)
| | | | | | | | - Mandy G. Katz‐Jaffe
- Fertility Labs of Colorado Lone Tree CO USA
- Fertility Genetics Lone Tree CO USA
- Colorado Center for Reproductive Medicine Lone Tree CO USA
| |
Collapse
|
6
|
Haberberger RV, Barry C, Matusica D. Immortalized Dorsal Root Ganglion Neuron Cell Lines. Front Cell Neurosci 2020; 14:184. [PMID: 32636736 PMCID: PMC7319018 DOI: 10.3389/fncel.2020.00184] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Pain is one of the most significant causes of suffering and disability world-wide, and arguably the most burdensome global health challenge. The growing number of patients suffering from chronic pain conditions such as fibromyalgia, complex regional pain syndrome, migraine and irritable bowel syndrome, not only reflect the complexity and heterogeneity of pain types, but also our lack of understanding of the underlying mechanisms. Sensory neurons within the dorsal root ganglia (DRG) have emerged as viable targets for effective chronic pain therapy. However, DRG's contain different classes of primary sensory neurons including pain-associated nociceptive neurons, non-nociceptive temperature sensing, mechanosensory and chemoreceptive neurons, as well as multiple types of immune and endothelial cells. This cell-population heterogeneity makes investigations of individual subgroups of DRG neurons, such as nociceptors, difficult. In attempts to overcome some of these difficulties, a limited number of immortalized DRG-derived cell lines have been generated over the past few decades. In vitro experiments using DRG-derived cell lines have been useful in understanding sensory neuron function. In addition to retaining phenotypic similarities to primary cultured DRG neurons, these cells offer greater suitability for high throughput assays due to ease of culture, maintenance, growth efficiency and cost-effectiveness. For accurate interpretation and translation of results it is critical, however, that phenotypic similarities and differences of DRG-derived cells lines are methodically compared to native neurons. Published reports to date show notable variability in how these DRG-derived cells are maintained and differentiated. Understanding the cellular and molecular differences stemming from different culture methods, is essential to validate past and future experiments, and enable these cells to be used to their full potential. This review describes currently available DRG-derived cell lines, their known sensory and nociceptor specific molecular profiles, and summarize their morphological features related to differentiation and neurite outgrowth.
Collapse
Affiliation(s)
- Rainer Viktor Haberberger
- Anatomy & Histology, College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Christine Barry
- Anatomy & Histology, College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Dusan Matusica
- Anatomy & Histology, College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
7
|
|
8
|
Gi-DREADD Expression in Peripheral Nerves Produces Ligand-Dependent Analgesia, as well as Ligand-Independent Functional Changes in Sensory Neurons. J Neurosci 2017; 36:10769-10781. [PMID: 27798132 DOI: 10.1523/jneurosci.3480-15.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 08/23/2016] [Indexed: 12/22/2022] Open
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) are an advanced experimental tool that could potentially provide a novel approach to pain management. In particular, expression of an inhibitory (Gi-coupled) DREADD in nociceptors might enable ligand-dependent analgesia. To test this possibility, TRPV1-cre mice were used to restrict expression of Gi-DREADDs to predominantly C-fibers. Whereas baseline heat thresholds in both male and female mice expressing Gi-DREADD were normal, 1 mg/kg clozapine-N-oxide (CNO) produced a significant 3 h increase in heat threshold that returned to baseline by 5 h after injection. Consistent with these behavioral results, CNO decreased action potential firing in isolated sensory neurons from Gi-DREADD mice. Unexpectedly, however, the expression of Gi-DREADD in sensory neurons caused significant changes in voltage-gated Ca2+ and Na+ currents in the absence of CNO, as well as an increase in Na+ channel (NaV1.7) expression. Furthermore, CNO-independent excitatory and inhibitory second-messenger signaling was also altered in these mice, which was associated with a decrease in the analgesic effect of endogenous inhibitory G-protein-coupled receptor activation. These results highlight the potential of this exciting technology, but also its limitations, and that it is essential to identify the underlying mechanisms for any observed behavioral phenotypes. SIGNIFICANCE STATEMENT DREADD technology is a powerful tool enabling manipulation of activity and/or transmitter release from targeted cell populations. The purpose of this study was to determine whether inhibitory DREADDs in nociceptive afferents could be used to produce analgesia, and if so, how. DREADD activation produced a ligand-dependent analgesia to heat in vivo and a decrease in neuronal firing at the single-cell level. However, we observed that expression of Gi-DREADD also causes ligand-independent changes in ion channel activity and second-messenger signaling. These findings highlight both the potential and the limitations of this exciting technology as well as the necessity to identify the mechanisms underlying any observed phenotype.
Collapse
|
9
|
Sandoval A, Duran P, Gandini MA, Andrade A, Almanza A, Kaja S, Felix R. Regulation of L-type Ca V1.3 channel activity and insulin secretion by the cGMP-PKG signaling pathway. Cell Calcium 2017; 66:1-9. [PMID: 28807144 DOI: 10.1016/j.ceca.2017.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/03/2017] [Accepted: 05/12/2017] [Indexed: 12/28/2022]
Abstract
cGMP is a second messenger widely used in the nervous system and other tissues. One of the major effectors for cGMP is the serine/threonine protein kinase, cGMP-dependent protein kinase (PKG), which catalyzes the phosphorylation of a variety of proteins including ion channels. Previously, it has been shown that the cGMP-PKG signaling pathway inhibits Ca2+ currents in rat vestibular hair cells and chromaffin cells. This current allegedly flow through voltage-gated CaV1.3L-type Ca2+ channels, and is important for controlling vestibular hair cell sensory function and catecholamine secretion, respectively. Here, we show that native L-type channels in the insulin-secreting RIN-m5F cell line, and recombinant CaV1.3 channels heterologously expressed in HEK-293 cells, are regulatory targets of the cGMP-PKG signaling cascade. Our results indicate that the CaVα1 ion-conducting subunit of the CaV1.3 channels is highly expressed in RIN-m5F cells and that the application of 8-Br-cGMP, a membrane-permeable analogue of cGMP, significantly inhibits Ca2+ macroscopic currents and impair insulin release stimulated with high K+. In addition, KT-5823, a specific inhibitor of PKG, prevents the current inhibition generated by 8-Br-cGMP in the heterologous expression system. Interestingly, mutating the putative phosphorylation sites to residues resistant to phosphorylation showed that the relevant PKG sites for CaV1.3 L-type channel regulation centers on two amino acid residues, Ser793 and Ser860, located in the intracellular loop connecting the II and III repeats of the CaVα1 pore-forming subunit of the channel. These findings unveil a novel mechanism for how the cGMP-PKG signaling pathway may regulate CaV1.3 channels and contribute to regulate insulin secretion.
Collapse
Affiliation(s)
| | - Paz Duran
- Departamento de Biología Celular, Cinvestav-IPN, Ciudad de México, Mexico
| | - María A Gandini
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Arturo Andrade
- Department of Biological Sciences, University of New Hampshire, Durham, NH, USA
| | - Angélica Almanza
- Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría, Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Simon Kaja
- Department of Ophtalmology and Molecular Pharmacology & Therapeutics, Loyola University, Chicago, Strich School of Medicine, Maywood, IL, USA
| | - Ricardo Felix
- Departamento de Biología Celular, Cinvestav-IPN, Ciudad de México, Mexico.
| |
Collapse
|
10
|
Agosti F, Cordisco Gonzalez S, Martinez Damonte V, Tolosa MJ, Di Siervi N, Schioth HB, Davio C, Perello M, Raingo J. Melanocortin 4 receptor constitutive activity inhibits L-type voltage-gated calcium channels in neurons. Neuroscience 2017; 346:102-112. [PMID: 28093215 DOI: 10.1016/j.neuroscience.2017.01.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/16/2016] [Accepted: 01/05/2017] [Indexed: 11/19/2022]
Abstract
The melanocortin 4 receptor (MC4R) is a G protein-coupled receptor (GPCR) that is expressed in several brain nuclei playing a crucial role in the regulation of energy balance controlling the homeostasis of the organism. It displays both agonist-evoked and constitutive activity, and moreover, it can couple to different G proteins. Most of the research on MC4R has been focused on agonist-induced activity, while the molecular and cellular basis of MC4R constitutive activity remains scarcely studied. We have previously shown that neuronal N-type voltage-gated calcium channels (CaV2.2) are inhibited by MC4R agonist-dependent activation, while the CaV subtypes that carry L- and P/Q-type current are not. Here, we tested the hypothesis that MC4R constitutive activity can affect CaV, with focus on the channel subtypes that can control transcriptional activity coupled to depolarization (L-type, CaV1.2/1.3) and neurotransmitter release (N- and P/Q-type, CaV2.2 and CaV2.1). We found that MC4R constitutive activity inhibits specifically CaV1.2/1.3 and CaV2.1 subtypes of CaV. We also explored the signaling pathways mediating this inhibition, and thus propose that agonist-dependent and basal MC4R activation modes signal differentially through Gs and Gi/o pathways to impact on different CaV subtypes. In addition, we found that chronic incubation with MC4R endogenous inverse agonist, agouti and agouti-related peptide (AgRP), occludes CaV inhibition in a cell line and in amygdaloid complex cultured neurons as well. Thus, we define new mechanisms of control of the main mediators of depolarization-induced calcium entry into neurons by a GPCR that displays constitutive activity.
Collapse
Affiliation(s)
- F Agosti
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| | - S Cordisco Gonzalez
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| | - V Martinez Damonte
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| | - M J Tolosa
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| | - N Di Siervi
- Instituto de Investigaciones Farmacológicas, ININFA, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
| | - H B Schioth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - C Davio
- Instituto de Investigaciones Farmacológicas, ININFA, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
| | - M Perello
- Neurophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| | - J Raingo
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| |
Collapse
|
11
|
François A, Scherrer G. Delta Opioid Receptor Expression and Function in Primary Afferent Somatosensory Neurons. Handb Exp Pharmacol 2017; 247:87-114. [PMID: 28993838 DOI: 10.1007/164_2017_58] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The functional diversity of primary afferent neurons of the dorsal root ganglia (DRG) generates a variety of qualitatively and quantitatively distinct somatosensory experiences, from shooting pain to pleasant touch. In recent years, the identification of dozens of genetic markers specifically expressed by subpopulations of DRG neurons has dramatically improved our understanding of this diversity and provided the tools to manipulate their activity and uncover their molecular identity and function. Opioid receptors have long been known to be expressed by discrete populations of DRG neurons, in which they regulate cell excitability and neurotransmitter release. We review recent insights into the identity of the DRG neurons that express the delta opioid receptor (DOR) and the ion channel mechanisms that DOR engages in these cells to regulate sensory input. We highlight recent findings derived from DORGFP reporter mice and from in situ hybridization and RNA sequencing studies in wild-type mice that revealed DOR presence in cutaneous mechanosensory afferents eliciting touch and implicated in tactile allodynia. Mechanistically, we describe how DOR modulates opening of voltage-gated calcium channels (VGCCs) to control glutamatergic neurotransmission between somatosensory neurons and postsynaptic neurons in the spinal cord dorsal horn. We additionally discuss other potential signaling mechanisms, including those involving potassium channels, which DOR may engage to fine tune somatosensation. We conclude by discussing how this knowledge may explain the analgesic properties of DOR agonists against mechanical pain and uncovers an unanticipated specialized function for DOR in cutaneous mechanosensation.
Collapse
Affiliation(s)
- Amaury François
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.,Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.,Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA. .,Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA. .,Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
12
|
G-protein-gated Inwardly Rectifying Potassium Channels Modulate Respiratory Depression by Opioids. Anesthesiology 2016; 124:641-50. [PMID: 26675532 DOI: 10.1097/aln.0000000000000984] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Drugs acting on μ-opioid receptors (MORs) are widely used as analgesics but present side effects including life-threatening respiratory depression. MORs are G-protein-coupled receptors inhibiting neuronal activity through calcium channels, adenylyl cyclase, and/or G-protein-gated inwardly rectifying potassium (GIRK) channels. The pathways underlying MOR-dependent inhibition of rhythmic breathing are unknown. METHODS By using a combination of genetic, pharmacological, and physiological tools in rodents in vivo, the authors aimed to identify the role of GIRK channels in MOR-mediated inhibition of respiratory circuits. RESULTS GIRK channels were expressed in the ventrolateral medulla, a neuronal population regulating rhythmic breathing, and GIRK channel activation with flupirtine reduced respiratory rate in rats (percentage of baseline rate in mean ± SD: 79.4 ± 7.4%, n = 7), wild-type mice (82.6 ± 3.8%, n = 3), but not in mice lacking the GIRK2 subunit, an integral subunit of neuronal GIRK channels (GIRK2, 101.0 ± 1.9%, n = 3). Application of the MOR agonist [D-Ala, N-MePhe, Gly-ol]-enkephalin (DAMGO) to the ventrolateral medulla depressed respiratory rate, an effect partially reversed by the GIRK channel blocker Tertiapin-Q (baseline: 42.1 ± 7.4 breath/min, DAMGO: 26.1 ± 13.4 breath/min, Tertiapin-Q + DAMGO: 33.9 ± 9.8 breath/min, n = 4). Importantly, DAMGO applied to the ventrolateral medulla failed to reduce rhythmic breathing in GIRK2 mice (percentage of baseline rate: 103.2 ± 12.1%, n = 4), whereas it considerably reduced rate in wild-type mice (62.5 ± 17.7% of baseline, n = 4). Respiratory rate depression by systemic injection of the opioid analgesic fentanyl was markedly reduced in GIRK2 (percentage of baseline: 12.8 ± 15.8%, n = 5) compared with wild-type mice (72.9 ± 27.3%). CONCLUSIONS Overall, these results identify that GIRK channels contribute to respiratory inhibition by MOR, an essential step toward understanding respiratory depression by opioids.
Collapse
|
13
|
Arias-Calderón M, Almarza G, Díaz-Vegas A, Contreras-Ferrat A, Valladares D, Casas M, Toledo H, Jaimovich E, Buvinic S. Characterization of a multiprotein complex involved in excitation-transcription coupling of skeletal muscle. Skelet Muscle 2016; 6:15. [PMID: 27069569 PMCID: PMC4827232 DOI: 10.1186/s13395-016-0087-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/19/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Electrical activity regulates the expression of skeletal muscle genes by a process known as "excitation-transcription" (E-T) coupling. We have demonstrated that release of adenosine 5'-triphosphate (ATP) during depolarization activates membrane P2X/P2Y receptors, being the fundamental mediators between electrical stimulation, slow intracellular calcium transients, and gene expression. We propose that this signaling pathway would require the proper coordination between the voltage sensor (dihydropyridine receptor, DHPR), pannexin 1 channels (Panx1, ATP release conduit), nucleotide receptors, and other signaling molecules. The goal of this study was to assess protein-protein interactions within the E-T machinery and to look for novel constituents in order to characterize the signaling complex. METHODS Newborn derived myotubes, adult fibers, or triad fractions from rat or mouse skeletal muscles were used. Co-immunoprecipitation, 2D blue native SDS/PAGE, confocal microscopy z-axis reconstruction, and proximity ligation assays were combined to assess the physical proximity of the putative complex interactors. An L6 cell line overexpressing Panx1 (L6-Panx1) was developed to study the influence of some of the complex interactors in modulation of gene expression. RESULTS Panx1, DHPR, P2Y2 receptor (P2Y2R), and dystrophin co-immunoprecipitated in the different preparations assessed. 2D blue native SDS/PAGE showed that DHPR, Panx1, P2Y2R and caveolin-3 (Cav3) belong to the same multiprotein complex. We observed co-localization and protein-protein proximity between DHPR, Panx1, P2Y2R, and Cav3 in adult fibers and in the L6-Panx1 cell line. We found a very restricted location of Panx1 and Cav3 in a putative T-tubule zone near the sarcolemma, while DHPR was highly expressed all along the transverse (T)-tubule. By Panx1 overexpression, extracellular ATP levels were increased both at rest and after electrical stimulation. Basal mRNA levels of the early gene cfos and the oxidative metabolism markers citrate synthase and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) were significantly increased by Panx1 overexpression. Interleukin 6 expression evoked by 20-Hz electrical stimulation (270 pulses, 0.3 ms each) was also significantly upregulated in L6-Panx1 cells. CONCLUSIONS We propose the existence of a relevant multiprotein complex that coordinates events involved in E-T coupling. Unveiling the molecular actors involved in the regulation of gene expression will contribute to the understanding and treatment of skeletal muscle disorders due to wrong-expressed proteins, as well as to improve skeletal muscle performance.
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Animals
- Animals, Newborn
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Caveolin 3/genetics
- Caveolin 3/metabolism
- Cell Line
- Connexins/genetics
- Connexins/metabolism
- Dystrophin/genetics
- Dystrophin/metabolism
- Electric Stimulation
- Gene Expression Regulation
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Multiprotein Complexes
- Muscle Contraction
- Muscle Fibers, Skeletal/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Protein Binding
- Rats, Wistar
- Receptors, Purinergic P2Y2/genetics
- Receptors, Purinergic P2Y2/metabolism
- Transcription, Genetic
- Transcriptional Activation
- Transfection
Collapse
Affiliation(s)
- Manuel Arias-Calderón
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
- />Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492 Santiago, Chile
| | - Gonzalo Almarza
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Alexis Díaz-Vegas
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Ariel Contreras-Ferrat
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Denisse Valladares
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Mariana Casas
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Héctor Toledo
- />Programa de Biología Molecular y Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Enrique Jaimovich
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
- />Programa de Biología Molecular y Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Sonja Buvinic
- />Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492 Santiago, Chile
| |
Collapse
|
14
|
Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW. Calcium-permeable ion channels in pain signaling. Physiol Rev 2014; 94:81-140. [PMID: 24382884 DOI: 10.1152/physrev.00023.2013] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The detection and processing of painful stimuli in afferent sensory neurons is critically dependent on a wide range of different types of voltage- and ligand-gated ion channels, including sodium, calcium, and TRP channels, to name a few. The functions of these channels include the detection of mechanical and chemical insults, the generation of action potentials and regulation of neuronal firing patterns, the initiation of neurotransmitter release at dorsal horn synapses, and the ensuing activation of spinal cord neurons that project to pain centers in the brain. Long-term changes in ion channel expression and function are thought to contribute to chronic pain states. Many of the channels involved in the afferent pain pathway are permeable to calcium ions, suggesting a role in cell signaling beyond the mere generation of electrical activity. In this article, we provide a broad overview of different calcium-permeable ion channels in the afferent pain pathway and their role in pain pathophysiology.
Collapse
|
15
|
Mallimo EM, Kusnecov AW. The role of orphanin FQ/nociceptin in neuroplasticity: relationship to stress, anxiety and neuroinflammation. Front Cell Neurosci 2013; 7:173. [PMID: 24155687 PMCID: PMC3792366 DOI: 10.3389/fncel.2013.00173] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 09/14/2013] [Indexed: 01/23/2023] Open
Abstract
The neuropeptide, orphanin FQ/nociceptin (OFQ/N or simply, nociceptin), is expressed in both neuronal and non-neuronal tissue, including the immune system. In the brain, OFQ/N has been investigated in relation to stress, anxiety, learning and memory, and addiction. More recently, it has also been found that OFQ/N influences glial cell functions, including oligodendrocytes, astrocytes, and microglial cells. However, this latter research is relatively small, but potentially important, when observations regarding the relationship of OFQ/N to stress and emotional functions is taken into consideration and integrated with the growing evidence for its involvement in cells that mediate inflammatory events. This review will first provide an overview and understanding of how OFQ/N has been implicated in the HPA axis response to stress, followed by an understanding of its influence on natural and learned anxiety-like behavior. What emerges from an examination of the literature is a neuropeptide that appears to counteract anxiogenic influences, but paradoxically, without attenuating HPA axis responses generated in response to stress. Studies utilized both central administration of OFQ/N, which was shown to activate the HPA axis, as well as antagonism of NOP-R, the OFQ/N receptor. In contrast, antagonist or transgenic OFQ/N or NOP-R knockout studies, showed augmentation of HPA axis responses to stress, suggesting that OFQ/N may be needed to control the magnitude of the HPA axis response to stress. Investigations of behavior in standard exploratory tests of anxiogenic behavior (eg., elevated plus maze) or learned fear responses have suggested that OFQ/N is needed to attenuate fear or anxiety-like behavior. However, some discrepant observations, in particular, those that involve appetitive behaviors, suggest a failure of NOP-R deletion to increase anxiety. However, it is also suggested that OFQ/N may operate in an anxiolytic manner when initial anxiogenic triggers (eg., the neuropeptide CRH) are initiated. Finally, the regulatory functions of OFQ/N in relation to emotion-related behaviors may serve to counteract potential neuroinflammatory events in the brain. This appears to be evident within the glial cell environment of the brain, since OFQ/N has been shown to reduce the production of proinflammatory cellular and cytokine events. Given that both OFQ/N and glial cells are activated in response to stress, it is possible that there is a possible convergence of these two systems that has important repercussions for behavior and neuroplasticity.
Collapse
Affiliation(s)
- Elyse M Mallimo
- Behavioral and Systems Neuroscience Program, Department of Psychology, Rutgers University New Brunswick, NJ, USA
| | | |
Collapse
|
16
|
The κ-opioid receptor agonist U-50488 blocks Ca2+ channels in a voltage- and G protein-independent manner in sensory neurons. Reg Anesth Pain Med 2013; 38:21-7. [PMID: 23222359 DOI: 10.1097/aap.0b013e318274a8a1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND OBJECTIVES κ-Opioid receptor (κ-OR) activation is known to play a role in analgesia and central sedation. The purpose of the present study was to examine the effect of the κ-OR agonist, U-50488 (an arylacetamide), on Ca channel currents and the signaling proteins involved in acutely isolated rat dorsal root ganglion (DRG) neurons expressing the putative promoter region of the tetrodotoxin-resistant Na channel (NaV 1.8) that is known to be involved in pain transmission. METHODS Acutely isolated rat DRG neurons were transfected with cDNA coding for enhanced green fluorescent protein (EGFP), whose expression is driven by the NaV 1.8 promoter region. Thereafter, the whole-cell variant of the patch-clamp technique was used to record Ca channel currents in neurons expressing EGFP. RESULTS Exposure of EGFP-expressing DRG neurons to U-50488 (0.3-40 μM) led to voltage-independent inhibition of the Ca channel currents. The modulation of the Ca currents did not appear to be mediated by the Gα protein subfamilies: Gαi/o, Gαs, Gαq/11, Gα14, and Gαz. Furthermore, dialysis of the hydrolysis-resistant GDP analog, GDP-β-S (1 mM), did not affect the U-50488-mediated blocking effect, ruling out involvement of other G protein subunits. Finally, U-50488 (20 μM) blocked Ca channels heterologously expressed in HeLa cells that do not express κ-OR. CONCLUSION These results suggest that the antinociceptive actions mediated by U-50488 are likely due to both a direct block of Ca channels in sensory neurons as well as G protein modulation of Ca currents via κ-OR-expressing neurons.
Collapse
|
17
|
Ponterio G, Tassone A, Sciamanna G, Riahi E, Vanni V, Bonsi P, Pisani A. Powerful inhibitory action of mu opioid receptors (MOR) on cholinergic interneuron excitability in the dorsal striatum. Neuropharmacology 2013; 75:78-85. [PMID: 23891638 DOI: 10.1016/j.neuropharm.2013.07.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/12/2013] [Accepted: 07/03/2013] [Indexed: 11/18/2022]
Abstract
Cholinergic interneurons (ChIs) of dorsal striatum play a key role in motor control and in behavioural learning. Neuropeptides regulate cholinergic transmission and mu opioid receptor (MOR) activation modulates striatal acetylcholine release. However, the mechanisms underlying this effect are yet uncharacterized. Here, we examined the electrophysiological responses of ChIs to the selective MOR agonist, DAMGO {[D-Ala2-MePhe4-Gly(ol)5] enkephalin}. We observed a robust, dose-dependent inhibition of spontaneous firing activity (0.06-3 μM) which was reversible upon drug washout and blocked by the selective antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP) (1 μM). Voltage-clamp analysis of the reversal potential of the DAMGO effect did not provide univocal results, indicating the involvement of multiple membrane conductances. The MOR-dependent effect persisted in the presence of GABAA and ionotropic glutamate receptor antagonists, ruling out an indirect effect. Additionally, it depended upon G-protein activation, as it was prevented by intrapipette GDP-β-S. Because D2 dopamine receptors (D2R) and MOR share a common post-receptor signalling pathway, occlusion experiments were performed with maximal doses of both D2R and MOR agonists. The D2R agonist quinpirole decreased spike discharge, which was further reduced by adding DAMGO. Then, D2R or MOR antagonists were used to challenge the response to the respective agonists, DAMGO or quinpirole. No cross-effect was observed, suggesting that the two receptors act independently. Our findings demonstrate a postsynaptic inhibitory modulation by MOR on ChIs excitability. Such opioidergic regulation of cholinergic transmission might contribute to shape information processing in basal ganglia circuits, and represent a potential target for pharmacological intervention.
Collapse
Affiliation(s)
- G Ponterio
- Department of System Medicine, University of Rome "Tor Vergata", via Montpellier, Rome 00133, Italy; Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - A Tassone
- Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - G Sciamanna
- Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - E Riahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; (d)Physiology Department, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - V Vanni
- Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - P Bonsi
- Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - A Pisani
- Department of System Medicine, University of Rome "Tor Vergata", via Montpellier, Rome 00133, Italy; Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia, IRCCS, Rome, Italy.
| |
Collapse
|
18
|
Voisin AN, Mouginot D, Drolet G. Multiple episodes of sodium depletion in the rat: a remodeling of the electrical properties of median preoptic nucleus neurons. Eur J Neurosci 2013; 38:2730-41. [DOI: 10.1111/ejn.12273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Aurore N. Voisin
- Axe Neurosciences du Centre de recherche du CHU and Université Laval; P-09800, 2705 Laurier; Québec; QC; G1V4G2; Canada
| | - Didier Mouginot
- Axe Neurosciences du Centre de recherche du CHU and Université Laval; P-09800, 2705 Laurier; Québec; QC; G1V4G2; Canada
| | - Guy Drolet
- Axe Neurosciences du Centre de recherche du CHU and Université Laval; P-09800, 2705 Laurier; Québec; QC; G1V4G2; Canada
| |
Collapse
|
19
|
Adams DJ, Berecki G. Mechanisms of conotoxin inhibition of N-type (Ca(v)2.2) calcium channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:1619-28. [PMID: 23380425 DOI: 10.1016/j.bbamem.2013.01.019] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/16/2013] [Accepted: 01/19/2013] [Indexed: 12/27/2022]
Abstract
N-type (Ca(v)2.2) voltage-gated calcium channels (VGCC) transduce electrical activity into other cellular functions, regulate calcium homeostasis and play a major role in processing pain information. Although the distribution and function of these channels vary widely among different classes of neurons, they are predominantly expressed in nerve terminals, where they control neurotransmitter release. To date, genetic and pharmacological studies have identified that high-threshold, N-type VGCCs are important for pain sensation in disease models. This suggests that N-type VGCC inhibitors or modulators could be developed into useful drugs to treat neuropathic pain. This review discusses the role of N-type (Ca(v)2.2) VGCCs in nociception and pain transmission through primary sensory dorsal root ganglion (DRG) neurons (nociceptors). It also outlines the potent and selective inhibition of N-type VGCCs by conotoxins, small disulfide-rich peptides isolated from the venom of marine cone snails. Of these conotoxins, ω-conotoxins are selective N-type VGCC antagonists that preferentially block nociception in inflammatory pain models, and allodynia and/or hyperalgesia in neuropathic pain models. Another conotoxin family, α-conotoxins, were initially proposed as competitive antagonists of muscle and neuronal nicotinic acetylcholine receptors (nAChR). Surprisingly, however, α-conotoxins Vc1.1 and RgIA, also potently inhibit N-type VGCC currents in the sensory DRG neurons of rodents and α9 nAChR knockout mice, via intracellular signaling mediated by G protein-coupled GABAB receptors. Understanding how conotoxins inhibit VGCCs is critical for developing these peptides into analgesics and may result in better pain management. This article is part of a Special Issue entitled: Calcium channels.
Collapse
Affiliation(s)
- David J Adams
- Health Innovations Research Institute, RMIT University, Melbourne, Victoria, Australia.
| | | |
Collapse
|
20
|
|
21
|
Compartmentalization of the GABAB receptor signaling complex is required for presynaptic inhibition at hippocampal synapses. J Neurosci 2011; 31:12523-32. [PMID: 21880914 DOI: 10.1523/jneurosci.1527-11.2011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Presynaptic inhibition via G-protein-coupled receptors (GPCRs) and voltage-gated Ca(2+) channels constitutes a widespread regulatory mechanism of synaptic strength. Yet, the mechanism of intermolecular coupling underlying GPCR-mediated signaling at central synapses remains unresolved. Using FRET spectroscopy, we provide evidence for formation of spatially restricted (<100 Å) complexes between GABA(B) receptors composed of GB(1a)/GB(2) subunits, Gα(o)β(1)γ(2) G-protein heterotrimer, and Ca(V)2.2 channels in hippocampal boutons. GABA release was not required for the assembly but for structural reorganization of the precoupled complex. Unexpectedly, GB(1a) deletion disrupted intermolecular associations within the complex. The GB(1a) proximal C-terminal domain was essential for association of the receptor, Ca(V)2.2 and Gβγ, but was dispensable for agonist-induced receptor activation and cAMP inhibition. Functionally, boutons lacking this complex-formation domain displayed impaired presynaptic inhibition of Ca(2+) transients and synaptic vesicle release. Thus, compartmentalization of the GABA(B1a) receptor, Gβγ, and Ca(V)2.2 channel in a signaling complex is required for presynaptic inhibition at hippocampal synapses.
Collapse
|
22
|
Turner RW, Anderson D, Zamponi GW. Signaling complexes of voltage-gated calcium channels. Channels (Austin) 2011; 5:440-8. [PMID: 21832880 DOI: 10.4161/chan.5.5.16473] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Voltage gated calcium channels are key mediators of depolarization induced calcium entry into electrically excitable cells. There is increasing evidence that voltage gated calcium channels, like many other types of ionic channels, do not operate in isolation, but instead forms signaling complexes with signaling molecules, G protein coupled receptors, and other types of ion channels. Furthermore, there appears to be bidirectional signaling within these protein complexes, thus allowing not only for efficient translation of calcium signals into cellular responses, but also for tight control of calcium entry per se. In this review, we will focus predominantly on signaling complexes between G protein-coupled receptors and high voltage activated calcium channels, and on complexes of voltage-gated calcium channels and members of the potassium channel superfamily.
Collapse
Affiliation(s)
- Ray W Turner
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| | | | | |
Collapse
|
23
|
Sobota JA, Mohler WA, Cowan AE, Eipper BA, Mains RE. Dynamics of peptidergic secretory granule transport are regulated by neuronal stimulation. BMC Neurosci 2010; 11:32. [PMID: 20202202 PMCID: PMC2838897 DOI: 10.1186/1471-2202-11-32] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 03/04/2010] [Indexed: 12/03/2022] Open
Abstract
Background Peptidergic neurons store and secrete the contents of large dense core vesicles (LDCVs) from axon terminals and from dendrites. Secretion of peptides requires a highly regulated exocytotic mechanism, plus coordinated synthesis and transport of LDCVs to their sites of release. Although these trafficking events are critical to function, little is known regarding the dynamic behavior of LDCVs and the mechanisms by which their transport is regulated. Sensory neurons also package opiate receptors in peptide-containing LDCVs, which is thought to be important in pain sensation. Since peptide granules cannot be refilled locally after their contents are secreted, it is particularly important to understand how neurons support regulated release of peptides. Results A vector encoding soluble peptidylglycine α-hydroxylating monooxygenase fused to green fluorescent protein was constructed to address these questions in cultured primary peptidergic neurons of the trigeminal ganglion using time lapse confocal microscopy. The time course of release differs with secretagogue; the secretory response to depolarization with K+ is rapid and terminates within 15 minutes, while phorbol ester stimulation of secretion is maintained over a longer period. The data demonstrate fundamental differences between LDCV dynamics in axons and growth cones under basal conditions. Conclusions Under basal conditions, LDCVs move faster away from the soma than toward the soma, but fewer LDCVs travel anterograde than retrograde. Stimulation decreased average anterograde velocity and increases granule pausing. Data from antibody uptake, quantification of enzyme secretion and appearance of pHluorin fluorescence demonstrate distributed release of peptides all along the axon, not just at terminals.
Collapse
Affiliation(s)
- Jacqueline A Sobota
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Ave, Farmington, CT 06030-3401, USA
| | | | | | | | | |
Collapse
|
24
|
Acid-evoked Ca2+ signalling in rat sensory neurones: effects of anoxia and aglycaemia. Pflugers Arch 2010; 459:159-81. [PMID: 19806360 PMCID: PMC2765625 DOI: 10.1007/s00424-009-0715-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 07/30/2009] [Accepted: 08/14/2009] [Indexed: 12/11/2022]
Abstract
Ischaemia excites sensory neurones (generating pain) and promotes calcitonin gene-related peptide release from nerve endings. Acidosis is thought to play a key role in mediating excitation via the activation of proton-sensitive cation channels. In this study, we investigated the effects of acidosis upon Ca2+ signalling in sensory neurones from rat dorsal root ganglia. Both hypercapnic (pHo 6.8) and metabolic–hypercapnic (pHo 6.2) acidosis caused a biphasic increase in cytosolic calcium concentration ([Ca2+]i). This comprised a brief Ca2+ transient (half-time approximately 30 s) caused by Ca2+ influx followed by a sustained rise in [Ca2+]i due to Ca2+ release from caffeine and cyclopiazonic acid-sensitive internal stores. Acid-evoked Ca2+ influx was unaffected by voltage-gated Ca2+-channel inhibition with nickel and acid sensing ion channel (ASIC) inhibition with amiloride but was blocked by inhibition of transient receptor potential vanilloid receptors (TRPV1) with (E)-3-(4-t-butylphenyl)-N-(2,3-dihydrobenzo[b][1,4] dioxin-6-yl)acrylamide (AMG 9810; 1 μM) and N-(4-tertiarybutylphenyl)-4-(3-cholorphyridin-2-yl) tetrahydropryazine-1(2H)-carbox-amide (BCTC; 1 μM). Combining acidosis with anoxia and aglycaemia increased the amplitude of both phases of Ca2+ elevation and prolonged the Ca2+ transient. The Ca2+ transient evoked by combined acidosis, aglycaemia and anoxia was also substantially blocked by AMG 9810 and BCTC and, to a lesser extent, by amiloride. In summary, the principle mechanisms mediating increase in [Ca2+]i in response to acidosis are a brief Ca2+ influx through TRPV1 followed by sustained Ca2+ release from internal stores. These effects are potentiated by anoxia and aglycaemia, conditions also prevalent in ischaemia. The effects of anoxia and aglycaemia are suggested to be largely due to the inhibition of Ca2+-clearance mechanisms and possible increase in the role of ASICs.
Collapse
|
25
|
Brittain JM, Piekarz AD, Wang Y, Kondo T, Cummins TR, Khanna R. An atypical role for collapsin response mediator protein 2 (CRMP-2) in neurotransmitter release via interaction with presynaptic voltage-gated calcium channels. J Biol Chem 2009; 284:31375-90. [PMID: 19755421 PMCID: PMC2781534 DOI: 10.1074/jbc.m109.009951] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 08/11/2009] [Indexed: 11/06/2022] Open
Abstract
Collapsin response mediator proteins (CRMPs) specify axon/dendrite fate and axonal growth of neurons through protein-protein interactions. Their functions in presynaptic biology remain unknown. Here, we identify the presynaptic N-type Ca(2+) channel (CaV2.2) as a CRMP-2-interacting protein. CRMP-2 binds directly to CaV2.2 in two regions: the channel domain I-II intracellular loop and the distal C terminus. Both proteins co-localize within presynaptic sites in hippocampal neurons. Overexpression in hippocampal neurons of a CRMP-2 protein fused to enhanced green fluorescent protein caused a significant increase in Ca(2+) channel current density, whereas lentivirus-mediated CRMP-2 knockdown abolished this effect. Interestingly, the increase in Ca(2+) current density was not due to a change in channel gating. Rather, cell surface biotinylation studies showed an increased number of CaV2.2 at the cell surface in CRMP-2-overexpressing neurons. These neurons also exhibited a significant increase in vesicular release in response to a depolarizing stimulus. Depolarization of CRMP-2-enhanced green fluorescent protein-overexpressing neurons elicited a significant increase in release of glutamate compared with control neurons. Toxin block of Ca(2+) entry via CaV2.2 abolished this stimulated release. Thus, the CRMP-2-Ca(2+) channel interaction represents a novel mechanism for modulation of Ca(2+) influx into nerve terminals and, hence, of synaptic strength.
Collapse
Affiliation(s)
- Joel M. Brittain
- From the Paul and Carole Stark Neurosciences Research Institute and
| | - Andrew D. Piekarz
- From the Paul and Carole Stark Neurosciences Research Institute and
- Departments of Pharmacology and Toxicologyand
| | - Yuying Wang
- From the Paul and Carole Stark Neurosciences Research Institute and
| | - Takako Kondo
- Otolaryngology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Theodore R. Cummins
- From the Paul and Carole Stark Neurosciences Research Institute and
- Departments of Pharmacology and Toxicologyand
| | - Rajesh Khanna
- From the Paul and Carole Stark Neurosciences Research Institute and
- Departments of Pharmacology and Toxicologyand
| |
Collapse
|
26
|
Marini P, Moriello AS, Cristino L, Palmery M, De Petrocellis L, Di Marzo V. Cannabinoid CB1 receptor elevation of intracellular calcium in neuroblastoma SH-SY5Y cells: Interactions with muscarinic and δ-opioid receptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1289-303. [DOI: 10.1016/j.bbamcr.2009.05.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 04/28/2009] [Accepted: 05/05/2009] [Indexed: 01/07/2023]
|
27
|
Grey CL, Chang JP. Ghrelin-induced growth hormone release from goldfish pituitary cells involves voltage-sensitive calcium channels. Gen Comp Endocrinol 2009; 160:148-57. [PMID: 19038258 DOI: 10.1016/j.ygcen.2008.11.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 10/15/2008] [Accepted: 11/01/2008] [Indexed: 11/23/2022]
Abstract
Ghrelin (GRL) is a stimulator of growth hormone (GH) release in many organisms, including goldfish. As a first study to examine the signalling mechanisms mediating GRL action on GH release in goldfish, we tested the hypothesis that GLR induces GH release from goldfish pituitary cells by enhancing Ca(2+) entry through L-type voltage-sensitive Ca(2+) channels (LVSCCs) using perifusion GH release and fura-2/AM Ca(2+)-imaging experiments. Goldfish (g)GRL(19) at 1 nM elicited reversible and repeatable GH responses from dispersed goldfish mixed pituitary cultures. However, the lack of a dose-response relationship in sequential treatments with decreasing concentrations of gGRL(19) (ranging from 10 to 0.01 nM) implicated rapid desensitization of the GH response. Sequential applications of gGRL(19) (1 nM) and salmon GnRH (100 nM), a known Ca(2+)-dependent stimulator of GH release, increased intracellular free Ca(2+) levels ([Ca(2+)](i)) from the same identified somatotropes, suggesting co-expression of GRL and GnRH receptors on single cells. In contrast, 1 nM gGRL(19) failed to elicit GH release and elevation in [Ca(2+)](i) when the cells are incubated with nominally Ca(2+)-free media. When GH release and [Ca(2+)](i) increases were already stimulated by the LVSCC agonist Bay K8644 (10 microM), addition of 1 nM gGRL(19) did not further elevate these responses. Finally, the LVSCC inhibitors nifedipine (1 microM) and verapamil (1 microM) abolished 1nM gGRL(19)-induced GH release responses while nifedipine eliminated gGRL(19)-induced [Ca(2+)](i) increase. Taken together, the results of this study provide evidence that entry of extracellular Ca(2+) through LVSCCs is a key component of the GRL signalling pathway leading to GH release in the goldfish pituitary.
Collapse
Affiliation(s)
- Caleb L Grey
- Department of Biological Sciences, University of Alberta, Edmonton, Alta., Canada T6G 2E9
| | | |
Collapse
|