1
|
Shokr MM, Badawi GA, Elshazly SM, Zaki HF, Mohamed AF. Sigma 1 Receptor and Its Pivotal Role in Neurological Disorders. ACS Pharmacol Transl Sci 2025; 8:47-65. [PMID: 39816800 PMCID: PMC11729429 DOI: 10.1021/acsptsci.4c00564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
Sigma 1 receptor (S1R) is a multifunctional, ligand-activated protein located in the membranes of the endoplasmic reticulum (ER). It mediates a variety of neurological disorders, including epilepsy, amyotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease. The wide neuroprotective effects of S1R agonists are achieved by a variety of pro-survival and antiapoptotic S1R-mediated signaling functions. Nonetheless, relatively little is known about the specific molecular mechanisms underlying S1R activity. Many studies on S1R protein have highlighted the importance of maintaining normal cellular homeostasis through its control of calcium and lipid exchange between the ER and mitochondria, ER-stress response, and many other mechanisms. In this review, we will discuss S1R different cellular localization and explain S1R-associated biological activity, such as its localization in the ER-plasma membrane and Mitochondrion-Associated ER Membrane interfaces. While outlining the cellular mechanisms and important binding partners involved in these processes, we also explained how the dysregulation of these pathways contributes to neurodegenerative disorders.
Collapse
Affiliation(s)
- Mustafa M. Shokr
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University−Arish Branch, Arish, 45511, Egypt
| | - Ghada A. Badawi
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University−Arish Branch, Arish, 45511, Egypt
| | - Shimaa M. Elshazly
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Hala F. Zaki
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed F. Mohamed
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Faculty
of Pharmacy, King Salman International University
(KSIU), South Sinai 46612, Egypt
| |
Collapse
|
2
|
Verma K, Prasanth MI, Tencomnao T, Brimson JM. Ligand docking in the sigma-1 receptor compared to the sigma-1 receptor-BiP complex and the effects of agonists and antagonists on C. elegans lifespans. Biomed Pharmacother 2025; 182:117783. [PMID: 39729653 DOI: 10.1016/j.biopha.2024.117783] [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: 10/08/2024] [Revised: 12/15/2024] [Accepted: 12/20/2024] [Indexed: 12/29/2024] Open
Abstract
Model organisms are commonly used to study human diseases; we set out to understand the relevance of several model organisms with relation to the σ1R protein. The study explored the interactions of σ1R with various agonists, antagonists across different species. Ligand and protein-protein (σ1R-BiP) docking approaches were used to understand the significance of σ1R in modulating neuroprotective mechanisms and its potential role in Alzheimer's. Ligand docking revealed that common σ1R antagonists generally exhibited stronger σ1R binding than commonly used agonists. Human σ1R showed high binding affinity for S1RA and NE100. Orthologs in yeast, slime mold, and C. elegans displayed varied binding affinities, indicating evolutionary adaptation in their binding pockets. We evaluated the relevance of σ1R-ligand interactions in C. elegans, measuring life-spans showing the impact of ligands on lifespan depends on genetic background and amyloid-beta pathology. Haloperidol (5-10 mM) extended wild-type worms' lifespan, but this effect was absent in the σ1R-KO, suggesting at least a partial role for the σ1R. Fluoxetine (5-10 mM) also promoted a small increase in longevity in wild-type worms but was not seen in the σ1R-KO strain. BD1047 (5 & 10 mM) reduced the lifespan of amyloid-beta-expressing transgenic worms, whereas dipentylamine (DPA) (5 mM) significantly increased the lifespan in a σ1R antagonist-sensitive manner. These findings highlight the importance of the σ1R in neurodegeneration and suggest that ligand interactions are modulated by BiP. Further research using in-vitro and in-vivo models is needed to clarify σ1R's therapeutic potential in neurodegenerative diseases, where modulating σ1R could provide neuroprotective effects.
Collapse
Affiliation(s)
- Kanika Verma
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Mani Iyer Prasanth
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - James Michael Brimson
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing, Chulalongkorn University, Bangkok 10330, Thailand; Research, Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
3
|
Prasanth MI, Verma K, Brimson S, Tencomnao T, Brimson JM. Simple ammonium salt and sigma-1 receptor ligand dipentylammonium provides neuroprotective effects in cell culture and Caenorhabditis elegans models of Alzheimer's disease. Biomed Pharmacother 2024; 173:116455. [PMID: 38503234 DOI: 10.1016/j.biopha.2024.116455] [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: 01/02/2024] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
The sigma-1 receptor (σ-1R), a chaperone protein located at the mitochondria-associated membrane (MAM) of the endoplasmic reticulum, can interact with and modify the signaling pathways of various proteins, thereby modulating many disease pathologies, including Alzheimer's disease (AD). The σ-1R ligand dipentylammonium (DPA) was analyzed for its anti-AD properties using PC12 cells (in vitro) and Caenorhabditis elegans (in vivo) models along with molecular docking (in silico) analysis. DPA at 1 and 10 µM concentrations was able to significantly potentiate NGF-induced neurite growth length by 137.7 ± 12.0 and 187.8 ± 16.4, respectively, when compared to the control 76.9 ± 7.4. DPA also regulated neurite damage caused by Aβ(25-35) treatment in differentiated PC12 cells by improving cell viability and neurite length. In C. elegans, DPA could significantly extend the median and maximum lifespan of Aβ transgenic strain CL2006 without impacting wild-type nematodes. Additionally, it could significantly reduce the paralysis phenotype of another Aβ transgenic strain, CL4176, thereby improving the overall health in AD pathogenesis. This effect depended on σ-1R, as DPA could not modulate the lifespan of σ-1R mutant TM3443. This was further confirmed using agonist PRE084 and antagonist BD1047, wherein the agonist alone could extend the lifespan of CL2006, while the antagonist suppressed the effect of DPA in CL2006. Interestingly, neither had an TM3443. Further, molecular docking analysis showed that DPA had a similar binding affinity as that of PRE084, BD1047 and pentazocine against the σ-1R receptor in humans and C. elegans, which collectively suggests the anti-AD properties of DPA.
Collapse
Affiliation(s)
- Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kanika Verma
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Molecular Epidemiology, ICMR-National Institute of Malaria Research (NIMR), New Delhi 110077, India
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - James Michael Brimson
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Research Unit for Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
4
|
Malar DS, Thitilertdecha P, Ruckvongacheep KS, Brimson S, Tencomnao T, Brimson JM. Targeting Sigma Receptors for the Treatment of Neurodegenerative and Neurodevelopmental Disorders. CNS Drugs 2023; 37:399-440. [PMID: 37166702 PMCID: PMC10173947 DOI: 10.1007/s40263-023-01007-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 05/12/2023]
Abstract
The sigma-1 receptor is a 223 amino acid-long protein with a recently identified structure. The sigma-2 receptor is a genetically unrelated protein with a similarly shaped binding pocket and acts to influence cellular activities similar to the sigma-1 receptor. Both proteins are highly expressed in neuronal tissues. As such, they have become targets for treating neurological diseases, including Alzheimer's disease (AD), Huntington's disease (HD), Parkinson's disease (PD), multiple sclerosis (MS), Rett syndrome (RS), developmental and epileptic encephalopathies (DEE), and motor neuron disease/amyotrophic lateral sclerosis (MND/ALS). In recent years, there have been many pre-clinical and clinical studies of sigma receptor (1 and 2) ligands for treating neurological disease. Drugs such as blarcamesine, dextromethorphan and pridopidine, which have sigma-1 receptor activity as part of their pharmacological profile, are effective in treating multiple aspects of several neurological diseases. Furthermore, several sigma-2 receptor ligands are under investigation, including CT1812, rivastigmine and SAS0132. This review aims to provide a current and up-to-date analysis of the current clinical and pre-clinical data of drugs with sigma receptor activities for treating neurological disease.
Collapse
Affiliation(s)
- Dicson S Malar
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanokphorn S Ruckvongacheep
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - James M Brimson
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Research, Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Room 409, ChulaPat-1 Building, 154 Rama 1 Road, Bangkok, 10330, Thailand.
| |
Collapse
|
5
|
Vavers E, Zvejniece L, Dambrova M. Sigma-1 receptor and seizures. Pharmacol Res 2023; 191:106771. [PMID: 37068533 PMCID: PMC10176040 DOI: 10.1016/j.phrs.2023.106771] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 04/19/2023]
Abstract
Over the last decade, sigma-1 receptor (Sig1R) has been recognized as a valid target for the treatment of seizure disorders and seizure-related comorbidities. Clinical trials with Sig1R ligands are underway testing therapies for the treatment of drug-resistant seizures, developmental and epileptic encephalopathies, and photosensitive epilepsy. However, the direct molecular mechanism by which Sig1R modulates seizures and the balance between excitatory and inhibitory pathways has not been fully elucidated. This review article aims to summarize existing knowledge of Sig1R and its involvement in seizures by focusing on the evidence obtained from Sig1R knockout animals and the anti-seizure effects of Sig1R ligands. In addition, this review article includes a discussion of the advantages and disadvantages of the use of existing compounds and describes the challenges and future perspectives on the use of Sig1R as a target for the treatment of seizure disorders.
Collapse
Affiliation(s)
- Edijs Vavers
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles 21, LV-1006, Riga, Latvia; University of Tartu, Faculty of Science and Technology, Institute of Chemistry, Ravila 14a, 50411, Tartu, Estonia.
| | - Liga Zvejniece
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles 21, LV-1006, Riga, Latvia
| | - Maija Dambrova
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles 21, LV-1006, Riga, Latvia; Riga Stradiņš University, Faculty of Pharmacy, Konsula 21, LV-1007, Riga, Latvia
| |
Collapse
|
6
|
Cai L, Liow JS, Morse CL, Telu S, Davies R, Manly LS, Zoghbi SS, Chin FT, Innis RB, Pike VW. Candidate 3-benzazepine-1-ol type GluN2B receptor radioligands ( 11C-NR2B-Me enantiomers) have high binding in cerebellum but not to σ1 receptors. EJNMMI Res 2023; 13:28. [PMID: 37017827 PMCID: PMC10076467 DOI: 10.1186/s13550-023-00975-6] [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: 11/09/2022] [Accepted: 03/12/2023] [Indexed: 04/06/2023] Open
Abstract
INTRODUCTION We recently reported 11C-NR2B-SMe ([S-methyl-11C](R,S)-7-thiomethoxy-3-(4-(4-methyl-phenyl)butyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-ol) and its enantiomers as candidate radioligands for imaging the GluN2B subunit within rat N-methyl-D-aspartate receptors. However, these radioligands gave unexpectedly high and displaceable binding in rat cerebellum, possibly due to cross-reactivity with sigma-1 (σ1) receptors. This study investigated 11C-labeled enantiomers of a close analogue (7-methoxy-3-(4-(p-tolyl)butyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-ol; NR2B-Me) of 11C-NR2B-SMe as new candidate GluN2B radioligands. PET was used to evaluate these radioligands in rats and to assess potential cross-reactivity to σ1 receptors. METHODS NR2B-Me was assayed for binding affinity and selectivity to GluN2B in vitro. 11C-NR2B-Me and its enantiomers were prepared by Pd-mediated treatment of boronic ester precursors with 11C-iodomethane. Brain PET scans were conducted after radioligand intravenous injection into rats. Various ligands for GluN2B receptors or σ1 receptors were administered at set doses in pre-blocking or displacement experiments to assess their impact on imaging data. 18F-FTC146 and enantiomers of 11C-NR2B-SMe were used for comparison. Radiometabolites from brain and plasma were measured ex vivo and in vitro. RESULTS NR2B-Me enantiomers showed high GluN2B affinity and selectivity in vitro. 11C-NR2B-Me enantiomers gave high early whole rat brain uptake of radioactivity, including high uptake in cerebellum, followed by slower decline. Radioactivity in brain at 30 min ex vivo was virtually all unchanged radioligand. Only less lipophilic radiometabolites appeared in plasma. When 11C-(R)-NR2B-Me was used, three high-affinity GluN2B ligands-NR2B-SMe, Ro25-6981, and CO101,244-showed increasing pre-block of whole brain radioactivity retention with increasing dose. Two σ1 receptor antagonists, FTC146 and BD1407, were ineffective pre-blocking agents. Together, these results strongly resemble those obtained with 11C-NR2B-SMe enantiomers, except that 11C-NR2B-Me enantiomers showed faster reversibility of binding. When 18F-FTC146 was used as a radioligand, FTC146 and BD1407 showed strong pre-blocking effects whereas GluN2B ligands showed only weak blocking effects. CONCLUSION 11C-NR2B-Me enantiomers showed specific binding to GluN2B receptors in rat brain in vivo. High unexpected specific binding in cerebellum was not due to σ1 receptors. Additional investigation is needed to identify the source of the high specific binding.
Collapse
Affiliation(s)
- Lisheng Cai
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA.
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Riley Davies
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Lester S Manly
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Frederick T Chin
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 1201 Welch Road, Rm. PS049, Stanford, CA, 94305-584, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| |
Collapse
|
7
|
Vera-Zambrano A, Baena-Nuevo M, Rinné S, Villegas-Esguevillas M, Barreira B, Telli G, de Benito-Bueno A, Blázquez JA, Climent B, Pérez-Vizcaino F, Valenzuela C, Decher N, Gonzalez T, Cogolludo A. Sigma-1 receptor modulation fine-tunes K V1.5 channels and impacts pulmonary vascular function. Pharmacol Res 2023; 189:106684. [PMID: 36740150 DOI: 10.1016/j.phrs.2023.106684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/23/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
KV1.5 channels are key players in the regulation of vascular tone and atrial excitability and their impairment is associated with cardiovascular diseases including pulmonary arterial hypertension (PAH) and atrial fibrillation (AF). Unfortunately, pharmacological strategies to improve KV1.5 channel function are missing. Herein, we aimed to study whether the chaperone sigma-1 receptor (S1R) is able to regulate these channels and represent a new strategy to enhance their function. By using different electrophysiological and molecular techniques in X. laevis oocytes and HEK293 cells, we demonstrate that S1R physically interacts with KV1.5 channels and regulate their expression and function. S1R induced a bimodal regulation of KV1.5 channel expression/activity, increasing it at low concentrations and decreasing it at high concentrations. Of note, S1R agonists (PRE084 and SKF10047) increased, whereas the S1R antagonist BD1047 decreased, KV1.5 expression and activity. Moreover, PRE084 markedly increased KV1.5 currents in pulmonary artery smooth muscle cells and attenuated vasoconstriction and proliferation in pulmonary arteries. We also show that both KV1.5 channels and S1R, at mRNA and protein levels, are clearly downregulated in samples from PAH and AF patients. Moreover, the expression of both genes showed a positive correlation. Finally, the ability of PRE084 to increase KV1.5 function was preserved under sustained hypoxic conditions, as an in vitro PAH model. Our study provides insight into the key role of S1R in modulating the expression and activity of KV1.5 channels and highlights the potential role of this chaperone as a novel pharmacological target for pathological conditions associated with KV1.5 channel dysfunction.
Collapse
Affiliation(s)
- Alba Vera-Zambrano
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain; Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain.
| | - Maria Baena-Nuevo
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain
| | - Susanne Rinné
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35043 Marburg, Germany
| | - Marta Villegas-Esguevillas
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Gokcen Telli
- Hacettepe University, Department of Pharmacology, Faculty of Pharmacy, Ankara, Turkey
| | | | | | - Belén Climent
- Department of Physiology, Faculty of Pharmacy, University Complutense of Madrid, Madrid, Spain
| | - Francisco Pérez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain; Spanish Network for Biomedical Research in Cardiovascular Research (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35043 Marburg, Germany
| | - Teresa Gonzalez
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain; Department of Physiology, Faculty of Pharmacy, University Complutense of Madrid, Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
| |
Collapse
|
8
|
Munguia-Galaviz FJ, Miranda-Diaz AG, Cardenas-Sosa MA, Echavarria R. Sigma-1 Receptor Signaling: In Search of New Therapeutic Alternatives for Cardiovascular and Renal Diseases. Int J Mol Sci 2023; 24:ijms24031997. [PMID: 36768323 PMCID: PMC9916216 DOI: 10.3390/ijms24031997] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Cardiovascular and renal diseases are among the leading causes of death worldwide, and regardless of current efforts, there is a demanding need for therapeutic alternatives to reduce their progression to advanced stages. The stress caused by diseases leads to the activation of protective mechanisms in the cell, including chaperone proteins. The Sigma-1 receptor (Sig-1R) is a ligand-operated chaperone protein that modulates signal transduction during cellular stress processes. Sig-1R interacts with various ligands and proteins to elicit distinct cellular responses, thus, making it a potential target for pharmacological modulation. Furthermore, Sig-1R ligands activate signaling pathways that promote cardioprotection, ameliorate ischemic injury, and drive myofibroblast activation and fibrosis. The role of Sig-1R in diseases has also made it a point of interest in developing clinical trials for pain, neurodegeneration, ischemic stroke, depression in patients with heart failure, and COVID-19. Sig-1R ligands in preclinical models have significantly beneficial effects associated with improved cardiac function, ventricular remodeling, hypertrophy reduction, and, in the kidney, reduced ischemic damage. These basic discoveries could inform clinical trials for heart failure (HF), myocardial hypertrophy, acute kidney injury (AKI), and chronic kidney disease (CKD). Here, we review Sig-1R signaling pathways and the evidence of Sig-1R modulation in preclinical cardiac and renal injury models to support the potential therapeutic use of Sig-1R agonists and antagonists in these diseases.
Collapse
Affiliation(s)
- Francisco Javier Munguia-Galaviz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Division de Ciencias de la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzman 49000, Jalisco, Mexico
| | - Alejandra Guillermina Miranda-Diaz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Miguel Alejandro Cardenas-Sosa
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Raquel Echavarria
- CONACYT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
- Correspondence:
| |
Collapse
|
9
|
Wang YM, Xia CY, Jia HM, He J, Lian WW, Yan Y, Wang WP, Zhang WK, Xu JK. Sigma-1 receptor: A potential target for the development of antidepressants. Neurochem Int 2022; 159:105390. [PMID: 35810915 DOI: 10.1016/j.neuint.2022.105390] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/10/2022] [Accepted: 07/05/2022] [Indexed: 10/17/2022]
Abstract
Though a great many of studies on the development of antidepressants for the therapy of major depression disorder (MDD) and the development of antidepressants have been carried out, there still lacks an efficient approach in clinical practice. The involvement of Sigma-1 receptor in the pathological process of MDD has been verified. In this review, recent research focusing on the role of Sigma-1 receptor in the etiology of MDD were summarized. Preclinical studies and clinical trials have found that stress induce the variation of Sigma-1 receptor in the blood, brain and heart. Dysfunction and absence of Sigma-1 receptor result in depressive-like behaviors in rodent animals. Agonists of Sigma-1 receptor show not only antidepressant-like activities but also therapeutical effects in complications of depression. The mechanisms underlying antidepressant-like effects of Sigma-1 receptor may include suppressing neuroinflammation, regulating neurotransmitters, ameliorating brain-derived neurotrophic factor and N-Methyl-D-Aspartate receptor, and alleviating the endoplasmic reticulum stress and mitochondria damage during stress. Therefore, Sigma-1 receptor represents a potential target for antidepressants development.
Collapse
Affiliation(s)
- Yu-Ming Wang
- School of Life Sciences & School of Chinese Medicine Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China; Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Cong-Yuan Xia
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Hong-Mei Jia
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, PR China
| | - Jun He
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wen-Wen Lian
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Yu Yan
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wen-Ping Wang
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wei-Ku Zhang
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China.
| | - Jie-Kun Xu
- School of Life Sciences & School of Chinese Medicine Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| |
Collapse
|
10
|
Fallica AN, Ciaffaglione V, Modica MN, Pittalà V, Salerno L, Amata E, Marrazzo A, Romeo G, Intagliata S. Structure-activity relationships of mixed σ1R/σ2R ligands with antiproliferative and anticancer effects. Bioorg Med Chem 2022; 73:117032. [DOI: 10.1016/j.bmc.2022.117032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
|
11
|
Sałaciak K, Pytka K. Revisiting the sigma-1 receptor as a biological target to treat affective and cognitive disorders. Neurosci Biobehav Rev 2022; 132:1114-1136. [PMID: 34736882 PMCID: PMC8559442 DOI: 10.1016/j.neubiorev.2021.10.037] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022]
Abstract
Depression and cognitive disorders are diseases with complex and not-fully understood etiology. Unfortunately, the COVID-19 pandemic dramatically increased the prevalence of both conditions. Since the current treatments are inadequate in many patients, there is a constant need for discovering new compounds, which will be more effective in ameliorating depressive symptoms and treating cognitive decline. Proteins attracting much attention as potential targets for drugs treating these conditions are sigma-1 receptors. Sigma-1 receptors are multi-functional proteins localized in endoplasmic reticulum membranes, which play a crucial role in cellular signal transduction by interacting with receptors, ion channels, lipids, and kinases. Changes in their functions and expression may lead to various diseases, including depression or memory impairments. Thus, sigma-1 receptor modulation might be useful in treating these central nervous system diseases. Importantly, two sigma-1 receptor ligands entered clinical trials, showing that this compound group possesses therapeutic potential. Therefore, based on preclinical studies, this review discusses whether the sigma-1 receptor could be a promising target for drugs treating affective and cognitive disorders.
Collapse
Affiliation(s)
- Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland.
| |
Collapse
|
12
|
Aishwarya R, Abdullah CS, Morshed M, Remex NS, Bhuiyan MS. Sigmar1's Molecular, Cellular, and Biological Functions in Regulating Cellular Pathophysiology. Front Physiol 2021; 12:705575. [PMID: 34305655 PMCID: PMC8293995 DOI: 10.3389/fphys.2021.705575] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The Sigma 1 receptor (Sigmar1) is a ubiquitously expressed multifunctional inter-organelle signaling chaperone protein playing a diverse role in cellular survival. Recessive mutation in Sigmar1 have been identified as a causative gene for neuronal and neuromuscular disorder. Since the discovery over 40 years ago, Sigmar1 has been shown to contribute to numerous cellular functions, including ion channel regulation, protein quality control, endoplasmic reticulum-mitochondrial communication, lipid metabolism, mitochondrial function, autophagy activation, and involved in cellular survival. Alterations in Sigmar1’s subcellular localization, expression, and signaling has been implicated in the progression of a wide range of diseases, such as neurodegenerative diseases, ischemic brain injury, cardiovascular diseases, diabetic retinopathy, cancer, and drug addiction. The goal of this review is to summarize the current knowledge of Sigmar1 biology focusing the recent discoveries on Sigmar1’s molecular, cellular, pathophysiological, and biological functions.
Collapse
Affiliation(s)
- Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Md Shenuarin Bhuiyan
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States.,Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| |
Collapse
|
13
|
Quadir SG, Tanino SM, Sami YN, Minnig MA, Iyer MR, Rice KC, Cottone P, Sabino V. Antagonism of Sigma-1 receptor blocks heavy alcohol drinking and associated hyperalgesia in male mice. Alcohol Clin Exp Res 2021; 45:1398-1407. [PMID: 34060104 PMCID: PMC8295198 DOI: 10.1111/acer.14635] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/16/2021] [Accepted: 05/10/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a complex psychiatric disease characterized by high alcohol intake as well as hyperkatifeia and hyperalgesia during withdrawal. A role for Sigma-1 receptors (Sig-1Rs) in the rewarding and reinforcing effects of alcohol has started to emerge in recent years, as rat studies have indicated that Sig-1R hyperactivity may result in excessive alcohol drinking. Sig-1R studies in mice are very scarce, and its potential role in alcohol-induced hyperalgesia is also unknown. METHODS In this study, we investigated the role of Sig-1R in alcohol drinking and associated hyperalgesia in male mice, using an intermittent access 2-bottle choice model of heavy drinking. RESULTS The Sig-1R antagonist BD-1063 was found dose dependently to reduce both alcohol intake and preference, without affecting either water or sucrose intake, suggesting that the effects are specific for alcohol. Notably, the ability of BD-1063 to suppress ethanol intake correlated with the individual baseline levels of alcohol drinking, suggesting that the treatment was more efficacious in heavy drinking animals. In addition, BD-1063 reversed alcohol-induced hyperalgesia during withdrawal, assessed using an automatic Hargreaves test, without affecting thermal sensitivity in alcohol-naïve animals or locomotor activity in either group. CONCLUSIONS These data show that Sig-1R antagonism dose-dependently reduced ethanol consumption in heavy drinking mice as well as its efficacy in reducing alcohol-induced hyperalgesia. These findings provide a foundation for the development of novel treatments for AUD and associated pain states.
Collapse
Affiliation(s)
- Sema G. Quadir
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord St., Boston, MA, USA
| | - Sean M. Tanino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord St., Boston, MA, USA
| | - Yasmine N. Sami
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord St., Boston, MA, USA
| | - Margaret A. Minnig
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord St., Boston, MA, USA
| | - Malliga R. Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Kenner C. Rice
- Drug Design and Synthesis, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord St., Boston, MA, USA
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord St., Boston, MA, USA
| |
Collapse
|
14
|
Brimson JM, Prasanth MI, Malar DS, Brimson S, Thitilertdecha P, Tencomnao T. Drugs that offer the potential to reduce hospitalization and mortality from SARS-CoV-2 infection: The possible role of the sigma-1 receptor and autophagy. Expert Opin Ther Targets 2021; 25:435-449. [PMID: 34236922 PMCID: PMC8290373 DOI: 10.1080/14728222.2021.1952987] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
Introduction: Despite the availability of new vaccines for SARS-CoV-2, there has been slow uptake and problems with supply in some parts of the world. Hence, there is still a necessity for drugs that can prevent hospitalization of patients and reduce the strain on health care systems. Drugs with sigma affinity potentially provide protection against the most severe symptoms of SARS-COV-2 and could prevent mortality via interactions with the sigma-1 receptor.Areas covered: This review examines the role of the sigma-1 receptor and autophagy in SARS-CoV-2 infections and how they may be linked. The authors reveal how sigma ligands may reduce the symptoms, complications, and deaths resulting from SARS-CoV-2 and offer insights on those patient cohorts that may benefit most from these drugs.Expert opinion: Drugs with sigma affinity potentially offer protection against the most severe symptoms of SARS-CoV-2 via interactions with the sigma-1 receptor. Agonists of the sigma-1 receptor may provide protection of the mitochondria, activate mitophagy to remove damaged and leaking mitochondria, prevent ER stress, manage calcium ion transport, and induce autophagy to prevent cell death in response to infection.
Collapse
Affiliation(s)
- James Michael Brimson
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Dicson Sheeja Malar
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
15
|
Noyer L, Lemonnier L, Mariot P, Gkika D. Partners in Crime: Towards New Ways of Targeting Calcium Channels. Int J Mol Sci 2019; 20:ijms20246344. [PMID: 31888223 PMCID: PMC6940757 DOI: 10.3390/ijms20246344] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/05/2019] [Accepted: 12/13/2019] [Indexed: 12/16/2022] Open
Abstract
The characterization of calcium channel interactome in the last decades opened a new way of perceiving ion channel function and regulation. Partner proteins of ion channels can now be considered as major components of the calcium homeostatic mechanisms, while the reinforcement or disruption of their interaction with the channel units now represents an attractive target in research and therapeutics. In this review we will focus on the targeting of calcium channel partner proteins in order to act on the channel activity, and on its consequences for cell and organism physiology. Given the recent advances in the partner proteins’ identification, characterization, as well as in the resolution of their interaction domain structures, we will develop the latest findings on the interacting proteins of the following channels: voltage-dependent calcium channels, transient receptor potential and ORAI channels, and inositol 1,4,5-trisphosphate receptor.
Collapse
Affiliation(s)
- Lucile Noyer
- Univ. Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, F-59000 Lille, France; (L.N.); (L.L.); (P.M.)
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - Loic Lemonnier
- Univ. Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, F-59000 Lille, France; (L.N.); (L.L.); (P.M.)
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - Pascal Mariot
- Univ. Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, F-59000 Lille, France; (L.N.); (L.L.); (P.M.)
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - Dimitra Gkika
- Univ. Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, F-59000 Lille, France; (L.N.); (L.L.); (P.M.)
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, 59655 Villeneuve d’Ascq, France
- Correspondence: ; Tél.: +33-(0)3-2043-6838
| |
Collapse
|
16
|
Soriani O, Kourrich S. The Sigma-1 Receptor: When Adaptive Regulation of Cell Electrical Activity Contributes to Stimulant Addiction and Cancer. Front Neurosci 2019; 13:1186. [PMID: 31780884 PMCID: PMC6861184 DOI: 10.3389/fnins.2019.01186] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022] Open
Abstract
The sigma-1 receptor (σ1R) is an endoplasmic reticulum (ER)-resident chaperone protein that acts like an inter-organelle signaling modulator. Among its several functions such as ER lipid metabolisms/transports and indirect regulation of genes transcription, one of its most intriguing feature is the ability to regulate the function and trafficking of a variety of functional proteins. To date, and directly relevant to the present review, σ1R has been found to regulate both voltage-gated ion channels (VGICs) belonging to distinct superfamilies (i.e., sodium, Na+; potassium, K+; and calcium, Ca2+ channels) and non-voltage-gated ion channels. This regulatory function endows σ1R with a powerful capability to fine tune cells’ electrical activity and calcium homeostasis—a regulatory power that appears to favor cell survival in pathological contexts such as stroke or neurodegenerative diseases. In this review, we present the current state of knowledge on σ1R’s role in the regulation of cellular electrical activity, and how this seemingly adaptive function can shift cell homeostasis and contribute to the development of very distinct chronic pathologies such as psychostimulant abuse and tumor cell growth in cancers.
Collapse
Affiliation(s)
| | - Saïd Kourrich
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC, Canada.,Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
17
|
Schmidt HR, Kruse AC. The Molecular Function of σ Receptors: Past, Present, and Future. Trends Pharmacol Sci 2019; 40:636-654. [PMID: 31387763 PMCID: PMC6748033 DOI: 10.1016/j.tips.2019.07.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/03/2019] [Accepted: 07/10/2019] [Indexed: 10/26/2022]
Abstract
The σ1 and σ2 receptors are enigmatic proteins that have attracted attention for decades due to the chemical diversity and therapeutic potential of their ligands. However, despite ongoing clinical trials with σ receptor ligands for multiple conditions, relatively little is known regarding the molecular function of these receptors. In this review, we revisit past research on σ receptors and discuss the interpretation of these data in light of recent developments. We provide a synthesis of emerging structural and genetic data on the σ1 receptor and discuss the recent cloning of the σ2 receptor. Finally, we discuss the major questions that remain in the study of σ receptors.
Collapse
Affiliation(s)
- Hayden R Schmidt
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
18
|
Ryskamp DA, Korban S, Zhemkov V, Kraskovskaya N, Bezprozvanny I. Neuronal Sigma-1 Receptors: Signaling Functions and Protective Roles in Neurodegenerative Diseases. Front Neurosci 2019; 13:862. [PMID: 31551669 PMCID: PMC6736580 DOI: 10.3389/fnins.2019.00862] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
Abstract
Sigma-1 receptor (S1R) is a multi-functional, ligand-operated protein situated in endoplasmic reticulum (ER) membranes and changes in its function and/or expression have been associated with various neurological disorders including amyotrophic lateral sclerosis/frontotemporal dementia, Alzheimer's (AD) and Huntington's diseases (HD). S1R agonists are broadly neuroprotective and this is achieved through a diversity of S1R-mediated signaling functions that are generally pro-survival and anti-apoptotic; yet, relatively little is known regarding the exact mechanisms of receptor functioning at the molecular level. This review summarizes therapeutically relevant mechanisms by which S1R modulates neurophysiology and implements neuroprotective functions in neurodegenerative diseases. These mechanisms are diverse due to the fact that S1R can bind to and modulate a large range of client proteins, including many ion channels in both ER and plasma membranes. We summarize the effect of S1R on its interaction partners and consider some of the cell type- and disease-specific aspects of these actions. Besides direct protein interactions in the endoplasmic reticulum, S1R is likely to function at the cellular/interorganellar level by altering the activity of several plasmalemmal ion channels through control of trafficking, which may help to reduce excitotoxicity. Moreover, S1R is situated in lipid rafts where it binds cholesterol and regulates lipid and protein trafficking and calcium flux at the mitochondrial-associated membrane (MAM) domain. This may have important implications for MAM stability and function in neurodegenerative diseases as well as cellular bioenergetics. We also summarize the structural and biochemical features of S1R proposed to underlie its activity. In conclusion, S1R is incredibly versatile in its ability to foster neuronal homeostasis in the context of several neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniel A. Ryskamp
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Svetlana Korban
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - Vladimir Zhemkov
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Nina Kraskovskaya
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, United States
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| |
Collapse
|
19
|
Abraham MJ, Fleming KL, Raymond S, Wong AYC, Bergeron R. The sigma-1 receptor behaves as an atypical auxiliary subunit to modulate the functional characteristics of Kv1.2 channels expressed in HEK293 cells. Physiol Rep 2019; 7:e14147. [PMID: 31222975 PMCID: PMC6586770 DOI: 10.14814/phy2.14147] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/17/2019] [Accepted: 05/25/2019] [Indexed: 12/11/2022] Open
Abstract
Expression of Kv1.2 within Kv1.x potassium channel complexes is critical in maintaining appropriate neuronal excitability and determining the threshold for action potential firing. This is attributed to the interaction of Kv1.2 with a hitherto unidentified protein that confers bimodal channel activation gating, allowing neurons to adapt to repetitive trains of stimulation and protecting against hyperexcitability. One potential protein candidate is the sigma-1 receptor (Sig-1R), which regulates other members of the Kv1.x channel family; however, the biophysical nature of the interaction between Sig-1R and Kv1.2 has not been elucidated. We hypothesized that Sig-1R may regulate Kv1.2 and may further act as the unidentified modulator of Kv1.2 activation. In transiently transfected HEK293 cells, we found that ligand activation of the Sig-1R modulates Kv1.2 current amplitude. More importantly, Sig-1R interacts with Kv1.2 in baseline conditions to influence bimodal activation gating. These effects are abolished in the presence of the auxiliary subunit Kvβ2 and when the Sig-1R mutation underlying ALS16 (Sig-1R-E102Q), is expressed. These data suggest that Kvβ2 occludes the interaction of Sig-1R with Kv1.2, and that E102 may be a residue critical for Sig-1R modulation of Kv1.2. The results of this investigation describe an important new role for Sig-1R in the regulation of neuronal excitability and introduce a novel mechanism of pathophysiology in Sig-1R dysfunction.
Collapse
Affiliation(s)
- Madelyn J. Abraham
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Kayla L. Fleming
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Sophie Raymond
- NeuroscienceOttawa Hospital Research InstituteOttawaOntarioCanada
| | | | - Richard Bergeron
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- NeuroscienceOttawa Hospital Research InstituteOttawaOntarioCanada
| |
Collapse
|
20
|
Quadir SG, Cottone P, Sabino V. Role of Sigma Receptors in Alcohol Addiction. Front Pharmacol 2019; 10:687. [PMID: 31258483 PMCID: PMC6586921 DOI: 10.3389/fphar.2019.00687] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/27/2019] [Indexed: 12/13/2022] Open
Abstract
Pharmacological treatments for alcohol use disorder (AUD) are few in number and often ineffective, despite the significant research carried out so far to better comprehend the neurochemical underpinnings of the disease. Hence, research has been directed towards the discovery of novel therapeutic targets for the treatment of AUD. In the last decade, the sigma receptor system has been proposed as a potential mediator of alcohol reward and reinforcement. Preclinical studies have shown that the motivational effects of alcohol and excessive ethanol consumption involve the recruitment of the sigma receptor system. Furthermore, sigma receptor antagonism has been shown to be sufficient to inhibit many behaviors related to AUDs. This paper will review the most current evidence in support of this receptor system as a potential target for the development of pharmacological agents for the treatment of alcohol addiction.
Collapse
Affiliation(s)
- Sema G Quadir
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, United States
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, United States
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
21
|
Morales-Lázaro SL, González-Ramírez R, Rosenbaum T. Molecular Interplay Between the Sigma-1 Receptor, Steroids, and Ion Channels. Front Pharmacol 2019; 10:419. [PMID: 31068816 PMCID: PMC6491805 DOI: 10.3389/fphar.2019.00419] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 04/03/2019] [Indexed: 11/17/2022] Open
Abstract
Cell excitability is tightly regulated by the activity of ion channels that allow for the passage of ions across cell membranes. Ion channel activity is controlled by different mechanisms that change their gating properties, expression or abundance in the cell membrane. The latter can be achieved by forming complexes with a diversity of proteins like chaperones such as the Sigma-1 receptor (Sig-1R), which is one with unique features and exhibits a role as a ligand-operated chaperone. This molecule also displays high intracellular mobility according to its activation level since, depletion of internal Ca+2 stores or the presence of specific ligands, produce Sig-1R’s mobilization from the endoplasmic reticulum toward the plasma membrane or nuclear envelope. The function of the Sig-1R as a chaperone is regulated by synthetic and endogenous ligands, with some of these compounds being a steroids and acting as key endogenous modifiers of the actions of the Sig-1R. There are cases in the literature that exemplify the close relationship between the actions of steroids on the Sig-1R and the resulting negative or positive effects on ion channel function/abundance. Such interactions have been shown to importantly influence the physiology of mammalian cells leading to changes in their excitability. The present review focuses on describing how the Sig-1R regulates the functional properties and the expression of some sodium, calcium, potassium, and TRP ion channels in the presence of steroids and the physiological consequences of these interplays at the cellular level are also discussed.
Collapse
Affiliation(s)
- Sara L Morales-Lázaro
- Departamento de Neurociencia Cognitiva, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Ricardo González-Ramírez
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General Dr. Manuel Gea González, Secretaría de Salud, Ciudad de México, Mexico
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
22
|
Cerveró C, Blasco A, Tarabal O, Casanovas A, Piedrafita L, Navarro X, Esquerda JE, Calderó J. Glial Activation and Central Synapse Loss, but Not Motoneuron Degeneration, Are Prevented by the Sigma-1 Receptor Agonist PRE-084 in the Smn2B/- Mouse Model of Spinal Muscular Atrophy. J Neuropathol Exp Neurol 2018; 77:577-597. [PMID: 29767748 DOI: 10.1093/jnen/nly033] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Spinal muscular atrophy (SMA) is characterized by the loss of α-motoneurons (MNs) with concomitant muscle denervation. MN excitability and vulnerability to disease are particularly regulated by cholinergic synaptic afferents (C-boutons), in which Sigma-1 receptor (Sig1R) is concentrated. Alterations in Sig1R have been associated with MN degeneration. Here, we investigated whether a chronic treatment with the Sig1R agonist PRE-084 was able to exert beneficial effects on SMA. We used a model of intermediate SMA, the Smn2B/- mouse, in which we performed a detailed characterization of the histopathological changes that occur throughout the disease. We report that Smn2B/- mice exhibited qualitative differences in major alterations found in mouse models of severe SMA: Smn2B/- animals showed more prominent MN degeneration, early motor axon alterations, marked changes in sensory neurons, and later MN deafferentation that correlated with conspicuous reactive gliosis and altered neuroinflammatory M1/M2 microglial balance. PRE-084 attenuated reactive gliosis, mitigated M1/M2 imbalance, and prevented MN deafferentation in Smn2B/- mice. These effects were also observed in a severe SMA model, the SMNΔ7 mouse. However, the prevention of gliosis and MN deafferentation promoted by PRE-084 were not accompanied by any improvements in clinical outcome or other major pathological changes found in SMA mice.
Collapse
Affiliation(s)
- Clàudia Cerveró
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Xavier Navarro
- Group of Neuroplasticity and Regeneration, Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Catalonia, Spain
| | - Josep E Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| |
Collapse
|
23
|
Sambo DO, Lebowitz JJ, Khoshbouei H. The sigma-1 receptor as a regulator of dopamine neurotransmission: A potential therapeutic target for methamphetamine addiction. Pharmacol Ther 2018; 186:152-167. [PMID: 29360540 PMCID: PMC5962385 DOI: 10.1016/j.pharmthera.2018.01.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Methamphetamine (METH) abuse is a major public health issue around the world, yet there are currently no effective pharmacotherapies for the treatment of METH addiction. METH is a potent psychostimulant that increases extracellular dopamine levels by targeting the dopamine transporter (DAT) and alters neuronal activity in the reward centers of the brain. One promising therapeutic target for the treatment of METH addiction is the sigma-1 receptor (σ1R). The σ1R is an endoplasmic reticulum-localized chaperone protein that is activated by cellular stress, and, unique to this chaperone, its function can also be induced or inhibited by different ligands. Upon activation of this unique "chaperone receptor", the σ1R regulates a variety of cellular functions and possesses neuroprotective activity in the brain. Interestingly, a variety of σ1R ligands modulate dopamine neurotransmission and reduce the behavioral effects of METH in animal models of addictive behavior, suggesting that the σ1R may be a viable therapeutic target for the treatment of METH addiction. In this review, we provide background on METH and the σ1R as well as a literature review regarding the role of σ1Rs in modulating both dopamine neurotransmission and the effects of METH. We aim to highlight the complexities of σ1R pharmacology and function as well as the therapeutic potential of the σ1R as a target for the treatment of METH addiction.
Collapse
Affiliation(s)
- Danielle O Sambo
- University of Florida, College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Joseph J Lebowitz
- University of Florida, College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Habibeh Khoshbouei
- University of Florida, College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States.
| |
Collapse
|
24
|
Ortíz-Rentería M, Juárez-Contreras R, González-Ramírez R, Islas LD, Sierra-Ramírez F, Llorente I, Simon SA, Hiriart M, Rosenbaum T, Morales-Lázaro SL. TRPV1 channels and the progesterone receptor Sig-1R interact to regulate pain. Proc Natl Acad Sci U S A 2018; 115:E1657-E1666. [PMID: 29378958 PMCID: PMC5816171 DOI: 10.1073/pnas.1715972115] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Transient Receptor Potential Vanilloid 1 (TRPV1) ion channel is expressed in nociceptors where, when activated by chemical or thermal stimuli, it functions as an important transducer of painful and itch-related stimuli. Although the interaction of TRPV1 with proteins that regulate its function has been previously explored, their modulation by chaperones has not been elucidated, as is the case for other mammalian TRP channels. Here we show that TRPV1 physically interacts with the Sigma 1 Receptor (Sig-1R), a chaperone that binds progesterone, an antagonist of Sig-1R and an important neurosteroid associated to the modulation of pain. Antagonism of Sig-1R by progesterone results in the down-regulation of TRPV1 expression in the plasma membrane of sensory neurons and, consequently, a decrease in capsaicin-induced nociceptive responses. This is observed both in males treated with a synthetic antagonist of Sig-1R and in pregnant females where progesterone levels are elevated. This constitutes a previously undescribed mechanism by which TRPV1-dependent nociception and pain can be regulated.
Collapse
Affiliation(s)
- Miguel Ortíz-Rentería
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Coyoacán, Ciudad de México, México
| | - Rebeca Juárez-Contreras
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Coyoacán, Ciudad de México, México
| | - Ricardo González-Ramírez
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General Dr. Manuel Gea González, Secretaría de Salud, 14080 Tlalpan, Ciudad de México, México
| | - León D Islas
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Coyoacán, Ciudad de México, México
| | - Félix Sierra-Ramírez
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Coyoacán, Ciudad de México, México
| | - Itzel Llorente
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Coyoacán, Ciudad de México, México
| | - Sidney A Simon
- Department of Neurobiology, Duke University, Durham, NC 27710
| | - Marcia Hiriart
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Coyoacán, Ciudad de México, México
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Coyoacán, Ciudad de México, México
| | - Sara L Morales-Lázaro
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Coyoacán, Ciudad de México, México;
| |
Collapse
|
25
|
Ji LL, Peng JB, Fu CH, Tong L, Wang ZY. Sigma-1 receptor activation ameliorates anxiety-like behavior through NR2A-CREB-BDNF signaling pathway in a rat model submitted to single-prolonged stress. Mol Med Rep 2017; 16:4987-4993. [PMID: 28791385 DOI: 10.3892/mmr.2017.7185] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/11/2017] [Indexed: 11/05/2022] Open
Abstract
Accumulating evidence has demonstrated that the σ-1 receptor (σ‑1R) possesses neuroprotective effects and is a potential novel therapeutic target for certain psychiatric diseases, including post‑traumatic stress disorder (PTSD) accompanied with anxiety disorder. It has been reported that σ‑1R agonist treatment could be modulated by the brain‑derived neurotrophic factor (BDNF) signaling pathway. However, it remains unclear whether BDNF and its upstream regulator are mechanistically involved in the therapeutic effect of σ‑1R in PTSD. To address this question, rats were subjected to a single‑prolonged stress (SPS) paradigm and σ‑1R agonist administration. Open‑field and elevated plus maze tests were implemented to evaluate the effect of σ‑1R activation on the improvement of anxiety‑like behaviors. Furthermore, the expression levels of BDNF, phosphorylated cAMP responsive element‑binding protein (CREB) and glutamate receptor ionotropic N‑methyl D‑aspartate 2A (NMDAR2A) were investigated in the hippocampi of rats. It was revealed that the downregulation of BDNF, phosphorylated CREB and NMDAR2A induced by SPS were reversed by σ‑1R activation. Collectively, the results of the present study suggest that the NMDAR2A/CREB/BDNF signaling pathway is involved in the activation of σ‑1R resulting in therapeutic effects for PTSD.
Collapse
Affiliation(s)
- Li-Li Ji
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Jun-Bo Peng
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Chang-Hai Fu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Lei Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Zhen-Yu Wang
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| |
Collapse
|
26
|
Potential independent action of sigma receptor ligands through inhibition of the Kv2.1 channel. Oncotarget 2017; 8:59345-59358. [PMID: 28938641 PMCID: PMC5601737 DOI: 10.18632/oncotarget.19581] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/16/2017] [Indexed: 12/14/2022] Open
Abstract
The sigma-1 receptor (σ1-R) and sigma-2 receptor (σ2-R) are potential drug targets for treatment of cancer, pain, depression, retinal degeneration and other neuronal diseases. Previous reports show that sigma-1 receptor modulates the activities of multiple channels. We are interested in possible sigma receptor modulation of Kv2.1, a K+ channel abundant in retinal photoreceptors. We tested the effect of established sigma receptor ligands on Kv2.1 channels which were stably expressed in HEK293 cells. Surprisingly, σ1-R antagonists inhibited Kv2.1 currents in both wild type and σ1-R knockout HEK293 cells that we engineered using the CRISPR/Cas9 technology. Moreover, PB28, a σ1-R antagonist and also σ2-R agonist, inhibited Kv2.1 in σ1-R knockout cells, but this action was not blocked by the σ2-R antagonists that did not have an effect on Kv2.1. We also observed inhibition of electroretinogram by PB28 in wild type as well as σ1-R knockout mice. Thus, the results in this study indicate that the Kv2.1-inhibiting function of the sigma ligands is not sigma receptor dependent, suggesting a direct effect of these ligands on the Kv2.1 channel.
Collapse
|
27
|
Zhang K, Zhao Z, Lan L, Wei X, Wang L, Liu X, Yan H, Zheng J. Sigma-1 Receptor Plays a Negative Modulation on N-type Calcium Channel. Front Pharmacol 2017; 8:302. [PMID: 28603497 PMCID: PMC5445107 DOI: 10.3389/fphar.2017.00302] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/10/2017] [Indexed: 12/30/2022] Open
Abstract
The sigma-1 receptor is a 223 amino acids molecular chaperone with a single transmembrane domain. It is resident to eukaryotic mitochondrial-associated endoplasmic reticulum and plasma membranes. By chaperone-mediated interactions with ion channels, G-protein coupled receptors and cell-signaling molecules, the sigma-1 receptor performs broad physiological and pharmacological functions. Despite sigma-1 receptors have been confirmed to regulate various types of ion channels, the relationship between the sigma-1 receptor and N-type Ca2+ channel is still unclear. Considering both sigma-1 receptors and N-type Ca2+ channels are involved in intracellular calcium homeostasis and neurotransmission, we undertake studies to explore the possible interaction between these two proteins. In the experiment, we confirmed the expression of the sigma-1 receptors and the N-type calcium channels in the cholinergic interneurons (ChIs) in rat striatum by using single-cell reverse transcription-polymerase chain reaction (scRT-PCR) and immunofluorescence staining. N-type Ca2+ currents recorded from ChIs in the brain slice of rat striatum was depressed when sigma-1 receptor agonists (SKF-10047 and Pre-084) were administrated. The inhibition was completely abolished by sigma-1 receptor antagonist (BD-1063). Co-expression of the sigma-1 receptors and the N-type calcium channels in Xenopus oocytes presented a decrease of N-type Ca2+ current amplitude with an increase of sigma-1 receptor expression. SKF-10047 could further depress N-type Ca2+ currents recorded from oocytes. The fluorescence resonance energy transfer (FRET) assays and co-immunoprecipitation (Co-IP) demonstrated that sigma-1 receptors and N-type Ca2+ channels formed a protein complex when they were co-expressed in HEK-293T (Human Embryonic Kidney -293T) cells. Our results revealed that the sigma-1 receptors played a negative modulation on N-type Ca2+ channels. The mechanism for the inhibition of sigma-1 receptors on N-type Ca2+ channels probably involved a chaperone-mediated direct interaction and agonist-induced conformational changes in the receptor-channel complexes on the cell surface.
Collapse
Affiliation(s)
- Kang Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and ToxicologyBeijing, China
| | - Zhe Zhao
- Department of Neurobiology, Beijing Institute of Basic Medical SciencesBeijing, China
| | - Liting Lan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and ToxicologyBeijing, China
| | - Xiaoli Wei
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and ToxicologyBeijing, China
| | - Liyun Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and ToxicologyBeijing, China
| | - Xiaoyan Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and ToxicologyBeijing, China
| | - Haitao Yan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and ToxicologyBeijing, China
| | - Jianquan Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and ToxicologyBeijing, China
| |
Collapse
|
28
|
Xu Q, Ji XF, Chi TY, Liu P, Jin G, Chen L, Zou LB. Sigma-1 receptor in brain ischemia/reperfusion: Possible role in the NR2A-induced pathway to regulate brain-derived neurotrophic factor. J Neurol Sci 2017; 376:166-175. [PMID: 28431607 DOI: 10.1016/j.jns.2017.03.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 02/18/2017] [Accepted: 03/17/2017] [Indexed: 11/18/2022]
Abstract
Sigma-1 receptor (σ1r) activation could attenuate the learning and memory deficits in the AD model, ischemia model and others. In our previous study, the activation of σ1r increased the expression of brain-derived neurotrophic factor (BDNF), possibly through the NR2A-induced pathway, and σ1r agonists might function as neuroprotectant agents in vascular dementia. Here, we used σ1r knockout mice to confirm the role of σ1r. Furthermore, an antagonist of NR2A was first used to investigate whether the NR2A-induced pathway is the necessary link between σ1r and BDNF. The operation of brain ischemia/reperfusion was induced by bilateral common carotid artery occlusion for 20min in C57BL/6 and σ1r knockout mice as the ischemic group. A σ1r agonist, PRE084 (1mg/kg, i.p.), and NR2A antagonist, PEAQX (10mg/kg, i.p.), were administered once daily throughout the experiment. Behavioral tests were performed starting on day 8. On day 22 after brain ischemia/reperfusion, mice were sacrificed and brains were immediately collected and the injured and the hippocampus was isolated and stored at -80°C for western blot analysis. After ischemic operation, contrast with the σ1r knockout mice, PRE084 significantly ameliorated learning and memory impairments in the behavioral evaluation, and prevented the protein decline of BDNF, NR2A, CaMKIV and TORC1 expression in wild-type mice. However, the effects of PRE084 on CaMKIV-TORC1-CREB and BDNF, even for learning and memory impairment, were antagonized by the co-administration of PEAQX, an antagonist of NR2A. The activation of σ1r improves the impairment of learning and memory in the ischemia/reperfusion model, and the expression of BDNF, which may have been achieved through the NR2A-CaMKIV-TORC1 pathway.
Collapse
Affiliation(s)
- Qian Xu
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xue-Fei Ji
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tian-Yan Chi
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Peng Liu
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ge Jin
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ling Chen
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China.
| | - Li-Bo Zou
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
29
|
Dasargyri A, Kümin CD, Leroux JC. Targeting Nanocarriers with Anisamide: Fact or Artifact? ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1603451. [PMID: 27885719 DOI: 10.1002/adma.201603451] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/08/2016] [Indexed: 05/19/2023]
Abstract
Encapsulating chemotherapeutics in nanoparticles can reduce the side effects of intravenous administration and improve their antitumor efficacy. Additionally, surface decoration of the nanocarriers with tumor-targeting ligands may enhance their specificity for cancer cells overexpressing the corresponding ligand-binding counterpart. The focus here is on anisamide, a low-molecular-weight benzamide derivative used as a tumor-directing moiety in functionalized nanosystems, based on its alleged interaction with Sigma receptors. The scintigraphic agents that initially inspired the use of anisamide for tumor targeting are described, and the published anisamide-tethered nanocarrier formulations are reviewed, together with a critical overview of the ligand's tumor-targeting properties. Moreover, anisamide's putative but dubious cellular target, the Sigma-1 receptor, is discussed with regard to its subcellular localization and implications in cancer. Data from in vivo studies reveal that the effect of anisamide on the antitumor efficacy of the decorated nanosystems varies considerably among the published reports. Together with the evidence questioning the interaction of anisamide with the Sigma receptors, the variability of anisamide's effect on the tumor deposition and the antitumor efficacy of the decorated drug carriers calls into question the extent of the ligand's tumor-targeting effect. Further research is necessary to elucidate the ligand's utility in tumor targeting.
Collapse
Affiliation(s)
- Athanasia Dasargyri
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), Zurich, 8093, Switzerland
| | - Carole D Kümin
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), Zurich, 8093, Switzerland
| | - Jean-Christophe Leroux
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), Zurich, 8093, Switzerland
| |
Collapse
|
30
|
Mavlyutov TA, Baker EM, Losenegger TM, Kim JR, Torres B, Epstein ML, Ruoho AE. The Sigma-1 Receptor-A Therapeutic Target for the Treatment of ALS? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:255-265. [PMID: 28315276 DOI: 10.1007/978-3-319-50174-1_17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The membrane bound 223 amino acid Sigma-1 Receptor (S1R) serves as a molecular chaperone and functional regulator of many signaling proteins. Spinal cord motor neuron activation occurs, in part, via large ventral horn cholinergic synapses called C-boutons/C-terminals. Chronic excitation of motor neurons and alterations in C-terminals has been associated with Amyotrophic Lateral Sclerosis (ALS ). The S1R has an important role in regulating motor neuron function. High levels of the S1R are localized in postsynaptic endoplasmic reticulum (ER) subsurface cisternae within 10-20 nm of the plasma membrane that contain muscarinic type 2 acetylcholine receptors (M2AChR), calcium activated potassium channels (Kv2.1) and slow potassium (SK) channels. An increase in action potentials in the S1R KO mouse motor neurons indicates a critical role for the S1R as a "brake" on motor neuron function possibly via calcium dependent hyperpolarization mechanisms involving the aforementioned potassium channels. The longevity of SOD-1/S1R KO ALS mice is significantly reduced compared to SOD-1/WT ALS controls. The S1R colocalizes in C-terminals with Indole(ethyl)amine-N-methyl transferase (INMT ), the enzyme that produces the S1R agonist , N,N'- dimethyltryptamine (DMT). INMT methylation can additionally neutralize endogenous toxic sulfur and selenium derivatives thus providing functional synergism with DMT to reduce oxidative stress in motor neurons . Small molecule activation of the S1R and INMT thus provides a possible therapeutic strategy to treat ALS .
Collapse
Affiliation(s)
- Timur A Mavlyutov
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Erin M Baker
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Tasher M Losenegger
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Jaimie R Kim
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Brian Torres
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Miles L Epstein
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Arnold E Ruoho
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
31
|
Mancuso R, Navarro X. Sigma-1 Receptor in Motoneuron Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:235-254. [PMID: 28315275 DOI: 10.1007/978-3-319-50174-1_16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS ) is a neurodegenerative disease affecting spinal cord and brain motoneurons , leading to paralysis and early death. Multiple etiopathogenic mechanisms appear to contribute in the development of ALS , including glutamate excitotoxicity, oxidative stress , protein misfolding, mitochondrial defects, impaired axonal transport, inflammation and glial cell alterations. The Sigma-1 receptor is highly expressed in motoneurons of the spinal cord, particularly enriched in the endoplasmic reticulum (ER) at postsynaptic cisternae of cholinergic C-terminals. Several evidences point to participation of Sigma-1R alterations in motoneuron degeneration. Thus, mutations of the transmembrane domain of the Sigma-1R have been described in familial ALS cases. Interestingly, Sigma-1R KO mice display muscle weakness and motoneuron loss. On the other hand, Sigma-1R agonists promote neuroprotection and neurite elongation through activation of protein kinase C on motoneurons in vitro and in vivo after ventral root avulsion. Remarkably, treatment of SOD1 mice, the most usual animal model of ALS , with Sigma-1R agonists resulted in significantly enhanced motoneuron function and preservation, and increased animal survival. Sigma-1R activation also reduced microglial reactivity and increased the glial expression of neurotrophic factors. Two main interconnected mechanisms seem to underlie the effects of Sigma-1R manipulation on motoneurons: modulation of neuronal excitability and regulation of calcium homeostasis. In addition, Sigma-1R also contributes to regulating protein degradation, and reducing oxidative stress. Therefore, the multi-functional nature of the Sigma-1R represents an attractive target for treating aspects of ALS and other motoneuron diseases .
Collapse
Affiliation(s)
- Renzo Mancuso
- Center for Biological Sciences, University of Southampton, Southampton General Hospital, SO16 6YD, Southampton, UK
| | - Xavier Navarro
- Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain.
| |
Collapse
|
32
|
Soriani O, Rapetti-Mauss R. Sigma 1 Receptor and Ion Channel Dynamics in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:63-77. [PMID: 28315265 DOI: 10.1007/978-3-319-50174-1_6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SigmaR1 is a multitasking chaperone protein which has mainly been studied in CNS physiological and pathophysiological processes such as pain, memory, neurodegenerative diseases (amyotrophic lateral sclerosis , Parkinson's and Alzheimer's diseases, retinal neurodegeneration ), stroke and addiction . Strikingly, G-protein and ion channels are the main client protein fami lies of this atypical chaperone and the recent advances that have been performed for the last 10 years demonstrate that SigmaR1 is principally activated following tissue injury and disease development to promote cell survival. In this chapter, we synthesize the data enhancing our comprehension of the interaction between SigmaR1 and ion channels and the unexpected consequences of such functional coupling in cancer development. We also describe a model in which the pro-survival functions of SigmaR1 observed in CNS pathologies are hijacked by cancer cells to shape their electrical signature and behavior in response to the tumor microenvironment .
Collapse
Affiliation(s)
- Olivier Soriani
- University of Nice Sophia Antipolis, CNRS, Inserm, iBV, 06108, Nice, France.
- Bâtiment Sciences Naturelles; UFR Sciences, 06108, Nice, France.
| | | |
Collapse
|
33
|
Abstract
The sigma-1 receptor (Sig-1R), via interaction with various proteins, including voltage-gated and ligand-gated ion channels (VGICs and LGICs), is involved in a plethora of neuronal functions. This capability to regulate a variety of ion channel targets endows the Sig-1R with a powerful capability to fine tune neuronal excitability, and thereby the transmission of information within brain circuits. This versatility may also explain why the Sig-1R is associated to numerous diseases at both peripheral and central levels. To date, how the Sig-1R chooses its targets and how the combinations of target modulations alter overall neuronal excitability is one of the challenges in the field of Sig-1R-dependent regulation of neuronal activity. Here, we will describe and discuss the latest findings on Sig-1R-dependent modulation of VGICs and LGICs, and provide hypotheses that may explain the diverse excitability outcomes that have been reported so far.
Collapse
Affiliation(s)
- Saïd Kourrich
- Department of Psychiatry, University of Texas Southwestern Medical Center, 2201 Inwood Road, Dallas, TX, 75390-9070, USA.
| |
Collapse
|
34
|
Aydar E, Stratton D, Fraser SP, Djamgoz MBA, Palmer C. Sigma-1 receptors modulate neonatal Na v1.5 ion channels in breast cancer cell lines. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2016; 45:671-683. [PMID: 27160185 DOI: 10.1007/s00249-016-1135-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/05/2016] [Accepted: 04/20/2016] [Indexed: 01/07/2023]
Abstract
The main aim of this study was to investigate a possible functional connection between sigma-1 receptors and voltage-gated sodium channels (VGSCs) in human breast cancer cells. The hypothesis was that sigma-1 drugs could alter the metastatic properties of breast cancer cells via the VGSC. Evidence was found for expression of sigma-1 receptor and neonatal Nav1.5 (nNav1.5) expression in both MDA-MB-231 and MDA-MB-468 cells. Sigma-1 drugs (SKF10047 and dimethyltryptamine) did not affect cell proliferation or migration but significantly reduced adhesion to the substrate. Silencing sigma-1 receptor expression by siRNA similarly reduced the adhesion. Blocking nNav1.5 activity with a polyclonal antibody (NESOpAb) targeting an extracellular region of nNav1.5 also reduced the adhesion in both cell lines. Importantly, the results of combined treatments with NESOpAb and a sigma-1 drug or sigma-1 siRNA suggested that both treatments targeted the same mechanism. The possibility was tested, therefore, that the sigma-1 receptor and the nNav1.5 channel formed a physical, functional complex. This suggestion was supported by the results of co-immunoprecipitation experiments. Furthermore, application of sigma-1 drugs to the cells reduced the surface expression of nNav1.5 protein, which could explain how sigma-1 receptor activation could alter the metastatic behaviour of breast cancer cells. Overall, these results are consistent with the idea of a sigma-1 protein behaving like either a "chaperone" or a regulatory subunit associated with nNav1.5.
Collapse
Affiliation(s)
- Ebru Aydar
- Institute of Ophthalmology, University College London, 11/43 Bath Street, London, EC1V 9EL, UK
| | - Dan Stratton
- Faculty of Life Sciences, London Metropolitan University, London, N7 8DB, UK
| | - Scott P Fraser
- Neuroscience Solutions to Cancer Research Group, Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, London, SW7 2AZ, UK
| | - Mustafa B A Djamgoz
- Neuroscience Solutions to Cancer Research Group, Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, London, SW7 2AZ, UK
- Biotechnology Research Centre (BRC), Cyprus International University, Haspolat, Lefkosa, Cyprus
| | - Christopher Palmer
- Faculty of Life Sciences, London Metropolitan University, London, N7 8DB, UK.
| |
Collapse
|
35
|
Bustamante M, Standl M, Bassat Q, Vilor-Tejedor N, Medina-Gomez C, Bonilla C, Ahluwalia TS, Bacelis J, Bradfield JP, Tiesler CMT, Rivadeneira F, Ring S, Vissing NH, Fink NR, Jugessur A, Mentch FD, Ballester F, Kriebel J, Kiefte-de Jong JC, Wolsk HM, Llop S, Thiering E, Beth SA, Timpson NJ, Andersen J, Schulz H, Jaddoe VWV, Evans DM, Waage J, Hakonarson H, Grant SFA, Jacobsson B, Bønnelykke K, Bisgaard H, Davey Smith G, Moll HA, Heinrich J, Estivill X, Sunyer J. A genome-wide association meta-analysis of diarrhoeal disease in young children identifies FUT2 locus and provides plausible biological pathways. Hum Mol Genet 2016; 25:4127-4142. [PMID: 27559109 PMCID: PMC5291237 DOI: 10.1093/hmg/ddw264] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/21/2016] [Accepted: 07/28/2016] [Indexed: 02/02/2023] Open
Abstract
More than a million childhood diarrhoeal episodes occur worldwide each year, and in developed countries a considerable part of them are caused by viral infections. In this study, we aimed to search for genetic variants associated with diarrhoeal disease in young children by meta-analyzing genome-wide association studies, and to elucidate plausible biological mechanisms. The study was conducted in the context of the Early Genetics and Lifecourse Epidemiology (EAGLE) consortium. Data about diarrhoeal disease in two time windows (around 1 year of age and around 2 years of age) was obtained via parental questionnaires, doctor interviews or medical records. Standard quality control and statistical tests were applied to the 1000 Genomes imputed genotypic data. The meta-analysis (N = 5758) followed by replication (N = 3784) identified a genome-wide significant association between rs8111874 and diarrhoea at age 1 year. Conditional analysis suggested that the causal variant could be rs601338 (W154X) in the FUT2 gene. Children with the A allele, which results in a truncated FUT2 protein, had lower risk of diarrhoea. FUT2 participates in the production of histo-blood group antigens and has previously been implicated in the susceptibility to infections, including Rotavirus and Norovirus Gene-set enrichment analysis suggested pathways related to the histo-blood group antigen production, and the regulation of ion transport and blood pressure. Among others, the gastrointestinal tract, and the immune and neuro-secretory systems were detected as relevant organs. In summary, this genome-wide association meta-analysis suggests the implication of the FUT2 gene in diarrhoeal disease in young children from the general population.
Collapse
Affiliation(s)
- Mariona Bustamante
- ISGlobal, Center for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Pompeu Fabra University (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Marie Standl
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Quique Bassat
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Natalia Vilor-Tejedor
- ISGlobal, Center for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Pompeu Fabra University (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Carolina Medina-Gomez
- The Generation R Study Group, Erasmus MC, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Carolina Bonilla
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Tarunveer S Ahluwalia
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Bacelis
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jonathan P Bradfield
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Carla M T Tiesler
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Ludwig-Maximilians-University of Munich, Dr. von Hauner Children's Hospital, Division of Metabolic Diseases and Nutritional Medicine, Munich, Germany
| | - Fernando Rivadeneira
- The Generation R Study Group, Erasmus MC, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Susan Ring
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Nadja H Vissing
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nadia R Fink
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Astanand Jugessur
- Department of Genetics and Bioinformatics, Area of Health Data and Digitalisation, Institute of Public Health, Oslo, Norway
| | - Frank D Mentch
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ferran Ballester
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, València, Spain
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Jessica C Kiefte-de Jong
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands
- Leiden University College, The Hague, The Netherlands
| | - Helene M Wolsk
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Sabrina Llop
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, València, Spain
| | - Elisabeth Thiering
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Ludwig-Maximilians-University of Munich, Dr. von Hauner Children's Hospital, Division of Metabolic Diseases and Nutritional Medicine, Munich, Germany
| | - Systke A Beth
- The Generation R Study Group, Erasmus MC, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands
| | - Nicholas J Timpson
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Josefine Andersen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Holger Schulz
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands
| | - David M Evans
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Johannes Waage
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Struan F A Grant
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Endocrinology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Genetics and Bioinformatics, Area of Health Data and Digitalisation, Institute of Public Health, Oslo, Norway
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - George Davey Smith
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Henriette A Moll
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, Inner City Clinic, University Hospital of Munich (LMU), Munich, Germany
| | - Xavier Estivill
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Pompeu Fabra University (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Experimental Genetics, Sidra Medical and Research Centre, Doha, Qatar
| | - Jordi Sunyer
- ISGlobal, Center for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Pompeu Fabra University (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
36
|
Wong AYC, Hristova E, Ahlskog N, Tasse LA, Ngsee JK, Chudalayandi P, Bergeron R. Aberrant Subcellular Dynamics of Sigma-1 Receptor Mutants Underlying Neuromuscular Diseases. Mol Pharmacol 2016; 90:238-53. [PMID: 27418673 DOI: 10.1124/mol.116.104018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/11/2016] [Indexed: 12/16/2023] Open
Abstract
The sigma-1 receptor (σ-1R) is an endoplasmic reticulum resident chaperone protein involved in a plethora of cellular functions, and whose disruption has been implicated in a wide range of diseases. Genetic analysis has revealed two σ-1R mutants involved in neuromuscular disorders. A point mutation (E102Q) in the ligand-binding domain results in the juvenile form of amyotrophic lateral sclerosis (ALS16), and a 20 amino-acid deletion (Δ31-50) in the putative cytosolic domain leads to a form of distal hereditary motor neuropathy. We investigated the localization and functional properties of these mutants in cell lines using confocal imaging and electrophysiology. The σ-1R mutants exhibited a significant increase in mobility, aberrant localization, and enhanced block of the inwardly rectifying K(+) channel Kir2.1, compared with the wild-type σ-1R. Thus, these σ-1R mutants have different functional properties that could contribute to their disease phenotypes.
Collapse
Affiliation(s)
- Adrian Y C Wong
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Elitza Hristova
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Nina Ahlskog
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Louis-Alexandre Tasse
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Johnny K Ngsee
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Prakash Chudalayandi
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Richard Bergeron
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| |
Collapse
|
37
|
Su TP, Su TC, Nakamura Y, Tsai SY. The Sigma-1 Receptor as a Pluripotent Modulator in Living Systems. Trends Pharmacol Sci 2016; 37:262-278. [PMID: 26869505 PMCID: PMC4811735 DOI: 10.1016/j.tips.2016.01.003] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 01/21/2023]
Abstract
The sigma-1 receptor (Sig-1R) is an endoplasmic reticulum (ER) protein that resides specifically in the mitochondria-associated endoplasmic reticulum (ER) membrane (MAM), an interface between ER and mitochondria. In addition to being able to translocate to the plasma membrane (PM) to interact with ion channels and other receptors, Sig-1R also occurs at the nuclear envelope, where it recruits chromatin-remodeling factors to affect the transcription of genes. Sig-1Rs have also been reported to interact with other membranous or soluble proteins at other loci, including the cytosol, and to be involved in several central nervous system (CNS) diseases. Here, we propose that Sig-1R is a pluripotent modulator with resultant multiple functional manifestations in living systems.
Collapse
Affiliation(s)
- Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA.
| | - Tzu-Chieh Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA
| | - Yoki Nakamura
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA
| | - Shang-Yi Tsai
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA
| |
Collapse
|
38
|
Chu UB, Ruoho AE. Biochemical Pharmacology of the Sigma-1 Receptor. Mol Pharmacol 2016; 89:142-53. [PMID: 26560551 DOI: 10.1124/mol.115.101170] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/06/2015] [Indexed: 12/19/2022] Open
Abstract
The sigma-1 receptor (S1R) is a 223 amino acid two transmembrane (TM) pass protein. It is a non-ATP-binding nonglycosylated ligand-regulated molecular chaperone of unknown three-dimensional structure. The S1R is resident to eukaryotic mitochondrial-associated endoplasmic reticulum and plasma membranes with broad functions that regulate cellular calcium homeostasis and reduce oxidative stress. Several multitasking functions of the S1R are underwritten by chaperone-mediated direct (and indirect) interactions with ion channels, G-protein coupled receptors and cell-signaling molecules involved in the regulation of cell growth. The S1R is a promising drug target for the treatment of several neurodegenerative diseases related to cellular stress. In vitro and in vivo functional and molecular characteristics of the S1R and its interactions with endogenous and synthetic small molecules have been discovered by the use of pharmacologic, biochemical, biophysical, and molecular biology approaches. The S1R exists in monomer, dimer, tetramer, hexamer/octamer, and higher oligomeric forms that may be important determinants in defining the pharmacology and mechanism(s) of action of the S1R. A canonical GXXXG in putative TM2 is important for S1R oligomerization. The ligand-binding regions of S1R have been identified and include portions of TM2 and the TM proximal regions of the C terminus. Some client protein chaperone functions and interactions with the cochaperone 78-kDa glucose-regulated protein (binding immunoglobulin protein) involve the C terminus. Based on its biochemical features and mechanisms of chaperone action the possibility that the S1R is a member of the small heat shock protein family is discussed.
Collapse
Affiliation(s)
- Uyen B Chu
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Arnold E Ruoho
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
39
|
Mari Y, Katnik C, Cuevas J. σ-1 Receptor Inhibition of ASIC1a Channels is Dependent on a Pertussis Toxin-Sensitive G-Protein and an AKAP150/Calcineurin Complex. Neurochem Res 2015; 40:2055-67. [PMID: 24925261 DOI: 10.1007/s11064-014-1324-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/17/2014] [Accepted: 05/03/2014] [Indexed: 10/25/2022]
Abstract
ASIC1a channels play a major role in various pathophysiological conditions including depression, anxiety, epilepsy, and neurodegeneration following ischemic stroke. Sigma-1 (σ-1) receptor stimulation depresses the activity of ASIC1a channels in cortical neurons, but the mechanism(s) by which σ-1 receptors exert their influence on ASIC1a remains unknown. Experiments were undertaken to elucidate the signaling cascade linking σ-1 receptors to ASIC1a channels. Immunohistochemical studies showed that σ-1 receptors, ASIC1a and A-kinase anchoring peptide 150 colocalize in the plasma membrane of the cell body and processes of cortical neurons. Fluorometric Ca(2+) imaging experiments showed that disruption of the macromolecular complexes containing AKAP150 diminished the effects of the σ-1 on ASIC1a, as did application of the calcineurin inhibitors, cyclosporin A and FK-506. Moreover, whole-cell patch clamp experiments showed that σ-1 receptors were less effective at decreasing ASIC1a-mediated currents in the presence of the VIVIT peptide, which binds to calcineurin and prevents cellular effects dependent on AKAP150/calcineurin interaction. The coupling of σ-1 to ASIC1a was also disrupted by preincubation of the neurons in the G-protein inhibitor, pertussis toxin (PTX). Taken together, our data reveal that σ-1 receptor block of ASIC1a function is dependent on activation of a PTX-sensitive G-protein and stimulation of AKAP150 bound calcineurin.
Collapse
Affiliation(s)
- Yelenis Mari
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., MDC-9, Tampa, FL, 33612-4799, USA
| | - Christopher Katnik
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., MDC-9, Tampa, FL, 33612-4799, USA
| | - Javier Cuevas
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., MDC-9, Tampa, FL, 33612-4799, USA.
| |
Collapse
|
40
|
Abstract
INTRODUCTION Neuropathic pain is difficult to relieve with standard analgesics and tends to be resistant to opioid therapy. Sigma-1 receptors activated during neuropathic injury may sustain pain. Neuropathic injury activates sigma-1 receptors, which results in activation of various kinases, modulates the activity of multiple ion channels, ligand activated ion channels and voltage-gated ion channels; alters monoamine neurotransmission and dampens opioid receptors G-protein activation. Activation of sigma-1 receptors tonically inhibits opioid receptor G-protein activation and thus dampens analgesic responses. Therefore, sigma-1 receptor antagonists are potential analgesics for neuropathic and adjuvants to opioid therapy. AREAS COVERED This article reviews the importance of sigma-1 receptors as pain generators in multiple animal models in order to illustrate both the importance of these unique receptors in pathologic pain and the potential benefits to sigma-1 receptor antagonists as analgesics. EXPERT OPINION Sigma-1 receptor antagonists have a great potential as analgesics for acute neuropathic injury (herpes zoster, acute postoperative pain and chemotherapy induced neuropathy) and may, as an additional benefit, prevent the development of chronic neuropathic pain. Antagonists are potentially effective as adjuvants to opioid therapy when used early to prevent analgesic tolerance. Drug development is complicated by the complexity of sigma-1 receptor pharmacodynamics and its multiple targets, the lack of a specific sigma-1 receptor antagonist, and potential side effects due to on-target toxicities (cognitive impairment, depression).
Collapse
Affiliation(s)
- Mellar P Davis
- Case Western Reserve University, Taussig Cancer Institute, Cleveland Clinic Lerner School of Medicine, Palliative Medicine and Supportive Oncology Services, Division of Solid Tumor, The Cleveland Clinic , 9500 Euclid Ave, Cleveland, OH 44195 , USA
| |
Collapse
|
41
|
Mishra AK, Mavlyutov T, Singh DR, Biener G, Yang J, Oliver JA, Ruoho A, Raicu V. The sigma-1 receptors are present in monomeric and oligomeric forms in living cells in the presence and absence of ligands. Biochem J 2015; 466:263-271. [PMID: 25510962 PMCID: PMC4500508 DOI: 10.1042/bj20141321] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The sigma-1 receptor (S1R) is a 223-amino-acid membrane protein that resides in the endoplasmic reticulum and the plasma membrane of some mammalian cells. The S1R is regulated by various synthetic molecules including (+)-pentazocine, cocaine and haloperidol and endogenous molecules such as sphingosine, dimethyltryptamine and dehydroepiandrosterone. Ligand-regulated protein chaperone functions linked to oxidative stress and neurodegenerative disorders such as amyotrophic lateral sclerosis (ALS) and neuropathic pain have been attributed to the S1R. Several client proteins that interact with S1R have been identified including various types of ion channels and G-protein coupled receptors (GPCRs). When S1R constructs containing C-terminal monomeric GFP2 and YFP fusions were co-expressed in COS-7 cells and subjected to FRET spectrometry analysis, monomers, dimers and higher oligomeric forms of S1R were identified under non-liganded conditions. In the presence of the prototypic S1R agonist, (+)-pentazocine, however, monomers and dimers were the prevailing forms of S1R. The prototypic antagonist, haloperidol, on the other hand, favoured higher order S1R oligomers. These data, in sum, indicate that heterologously expressed S1Rs occur in vivo in COS-7 cells in multiple oligomeric forms and that S1R ligands alter these oligomeric structures. We suggest that the S1R oligomerization states may regulate its function(s).
Collapse
Affiliation(s)
- Ashish K. Mishra
- Physics Department, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, U.S.A
| | - Timur Mavlyutov
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53211, U.S.A
| | - Deo R. Singh
- Physics Department, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, U.S.A
| | - Gabriel Biener
- Physics Department, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, U.S.A
| | - Jay Yang
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI 53211, U.S.A
| | - Julie A. Oliver
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, U.S.A
| | - Arnold Ruoho
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53211, U.S.A
| | - Valerică Raicu
- Physics Department, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, U.S.A
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, U.S.A
| |
Collapse
|
42
|
Mavlyutov TA, Guo LW, Epstein ML, Ruoho AE. Role of the Sigma-1 receptor in Amyotrophic Lateral Sclerosis (ALS). J Pharmacol Sci 2015; 127:10-6. [PMID: 25704013 PMCID: PMC4489701 DOI: 10.1016/j.jphs.2014.12.013] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 12/18/2014] [Accepted: 12/21/2014] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease affecting spinal cord motoneurons (MN) with an associative connection to Frontotemporal Lobar Dementia (FTLD). The endoplasmic reticulum (ER) bound Sigma-1 Receptor (S1R) chaperone protein localizes to specialized ER cisternae within 10 nm of the plasma membrane in spinal cord ventral horn cholinergic post synaptic C-terminals. Removal of the S1R gene in the Superoxide Dismutase-1 (SOD-1) mouse model of ALS exacerbated the neurodegenerative condition and resulted in a significantly reduced longevity when compared to the SOD-1/S1R wild type (WT) mouse. The proposed amelioration of the ALS phenotype by the S1R is likely due to a "brake" on excitation of the MN as evidenced by a reduction in action potential generation in the MN of the WT when compared to the S1R KO mouse MN. Although the precise signal transduction pathway(s) regulated by the S1R in the MN has/have not been elucidated at present, it is likely that direct or indirect functional interactions occur between the S1R in the ER cisternae with voltage gated potassium channels and/or with muscarinic M2 receptor signaling in the post synaptic plasma membrane. Possible mechanisms for regulation of MN excitability by S1R are discussed.
Collapse
Affiliation(s)
- Timur A Mavlyutov
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, USA; Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, USA
| | - Lian-Wang Guo
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, USA
| | - Miles L Epstein
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, USA
| | - Arnold E Ruoho
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, USA.
| |
Collapse
|
43
|
Balasuriya D, D'Sa L, Talker R, Dupuis E, Maurin F, Martin P, Borgese F, Soriani O, Edwardson JM. A direct interaction between the sigma-1 receptor and the hERG voltage-gated K+ channel revealed by atomic force microscopy and homogeneous time-resolved fluorescence (HTRF®). J Biol Chem 2014; 289:32353-32363. [PMID: 25266722 PMCID: PMC4231707 DOI: 10.1074/jbc.m114.603506] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/17/2014] [Indexed: 01/29/2023] Open
Abstract
The sigma-1 receptor is an endoplasmic reticulum chaperone protein, widely expressed in central and peripheral tissues, which can translocate to the plasma membrane and modulate the function of various ion channels. The human ether-à-go-go-related gene encodes hERG, a cardiac voltage-gated K(+) channel that is abnormally expressed in many human cancers and is known to interact functionally with the sigma-1 receptor. Our aim was to investigate the nature of the interaction between the sigma-1 receptor and hERG. We show that the two proteins can be co-isolated from a detergent extract of stably transfected HEK-293 cells, consistent with a direct interaction between them. Atomic force microscopy imaging of the isolated protein confirmed the direct binding of the sigma-1 receptor to hERG monomers, dimers, and tetramers. hERG dimers and tetramers became both singly and doubly decorated by sigma-1 receptors; however, hERG monomers were only singly decorated. The distribution of angles between pairs of sigma-1 receptors bound to hERG tetramers had two peaks, at ∼90 and ∼180° in a ratio of ∼2:1, indicating that the sigma-1 receptor interacts with hERG with 4-fold symmetry. Homogeneous time-resolved fluorescence (HTRF®) allowed the detection of the interaction between the sigma-1 receptor and hERG within the plane of the plasma membrane. This interaction was resistant to sigma ligands, but was decreased in response to cholesterol depletion of the membrane. We suggest that the sigma-1 receptor may bind to hERG in the endoplasmic reticulum, aiding its assembly and trafficking to the plasma membrane.
Collapse
Affiliation(s)
- Dilshan Balasuriya
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, United Kingdom
| | - Lauren D'Sa
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, United Kingdom
| | - Ronel Talker
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, United Kingdom
| | - Elodie Dupuis
- CisBio Bioassays, Parc Marcel Boiteux BP 84175, 30200 Codolet, France, and
| | - Fabrice Maurin
- CisBio Bioassays, Parc Marcel Boiteux BP 84175, 30200 Codolet, France, and
| | - Patrick Martin
- Institut de Biologie de Valrose (iBV), CNRS UMR 7277, INSERM U1091 UNS, Faculté des Sciences, Université de Nice Sophia Antipolis, 06108 Nice Cedex 2, France
| | - Franck Borgese
- Institut de Biologie de Valrose (iBV), CNRS UMR 7277, INSERM U1091 UNS, Faculté des Sciences, Université de Nice Sophia Antipolis, 06108 Nice Cedex 2, France
| | - Olivier Soriani
- Institut de Biologie de Valrose (iBV), CNRS UMR 7277, INSERM U1091 UNS, Faculté des Sciences, Université de Nice Sophia Antipolis, 06108 Nice Cedex 2, France.
| | - J Michael Edwardson
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, United Kingdom,.
| |
Collapse
|
44
|
Pabba M, Wong AYC, Ahlskog N, Hristova E, Biscaro D, Nassrallah W, Ngsee JK, Snyder M, Beique JC, Bergeron R. NMDA receptors are upregulated and trafficked to the plasma membrane after sigma-1 receptor activation in the rat hippocampus. J Neurosci 2014; 34:11325-38. [PMID: 25143613 PMCID: PMC6615506 DOI: 10.1523/jneurosci.0458-14.2014] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 07/08/2014] [Accepted: 07/14/2014] [Indexed: 11/21/2022] Open
Abstract
Sigma-1 receptors (σ-1Rs) are endoplasmic reticulum resident chaperone proteins implicated in many physiological and pathological processes in the CNS. A striking feature of σ-1Rs is their ability to interact and modulate a large number of voltage- and ligand-gated ion channels at the plasma membrane. We have reported previously that agonists for σ-1Rs potentiate NMDA receptor (NMDAR) currents, although the mechanism by which this occurs is still unclear. In this study, we show that in vivo administration of the selective σ-1R agonists (+)-SKF 10,047 [2S-(2α,6α,11R*]-1,2,3,4,5,6-hexahydro-6,11-dimethyl-3-(2-propenyl)-2,6-methano-3-benzazocin-8-ol hydrochloride (N-allylnormetazocine) hydrochloride], PRE-084 (2-morpholin-4-ylethyl 1-phenylcyclohexane-1-carboxylate hydrochloride), and (+)-pentazocine increases the expression of GluN2A and GluN2B subunits, as well as postsynaptic density protein 95 in the rat hippocampus. We also demonstrate that σ-1R activation leads to an increased interaction between GluN2 subunits and σ-1Rs and mediates trafficking of NMDARs to the cell surface. These results suggest that σ-1R may play an important role in NMDAR-mediated functions, such as learning and memory. It also opens new avenues for additional studies into a multitude of pathological conditions in which NMDARs are involved, including schizophrenia, dementia, and stroke.
Collapse
Affiliation(s)
- Mohan Pabba
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, K1H 8M5, and
| | - Adrian Y C Wong
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, K1H 8M5, and
| | - Nina Ahlskog
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, K1H 8M5, and
| | - Elitza Hristova
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, K1H 8M5, and
| | - Dante Biscaro
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, K1H 8M5, and
| | - Wissam Nassrallah
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, K1H 8M5, and
| | - Johnny K Ngsee
- Department of Cellular and Molecular Medicine, University of Ottawa, Ontario, Canada, K1H 8M5
| | - Melissa Snyder
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, K1H 8M5, and
| | - Jean-Claude Beique
- Department of Cellular and Molecular Medicine, University of Ottawa, Ontario, Canada, K1H 8M5
| | - Richard Bergeron
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, K1H 8M5, and
| |
Collapse
|
45
|
Matsumoto RR, Nguyen L, Kaushal N, Robson MJ. Sigma (σ) receptors as potential therapeutic targets to mitigate psychostimulant effects. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:323-86. [PMID: 24484982 DOI: 10.1016/b978-0-12-420118-7.00009-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Many psychostimulants, including cocaine and methamphetamine, interact with sigma (σ) receptors at physiologically relevant concentrations. The potential therapeutic relevance of this interaction is underscored by the ability to selectively target σ receptors to mitigate many behavioral and physiological effects of psychostimulants in animal and cell-based model systems. This chapter begins with an overview of these enigmatic proteins. Provocative preclinical data showing that σ ligands modulate an array of cocaine and methamphetamine effects are summarized, along with emerging areas of research. Together, the literature suggests targeting of σ receptors as an innovative option for combating undesired actions of psychostimulants through both neuronal and glial mechanisms.
Collapse
Affiliation(s)
- Rae R Matsumoto
- West Virginia University, One Medical Center Drive, Morgantown, West Virginia, USA.
| | - Linda Nguyen
- West Virginia University, One Medical Center Drive, Morgantown, West Virginia, USA
| | - Nidhi Kaushal
- West Virginia University, One Medical Center Drive, Morgantown, West Virginia, USA
| | - Matthew J Robson
- West Virginia University, One Medical Center Drive, Morgantown, West Virginia, USA
| |
Collapse
|
46
|
Crottès D, Guizouarn H, Martin P, Borgese F, Soriani O. The sigma-1 receptor: a regulator of cancer cell electrical plasticity? Front Physiol 2013; 4:175. [PMID: 23882221 PMCID: PMC3712323 DOI: 10.3389/fphys.2013.00175] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 06/21/2013] [Indexed: 12/18/2022] Open
Abstract
Originally mistaken as an opioid receptor, the sigma-1 receptor (Sig1R) is a ubiquitous membrane protein that has been involved in many cellular processes. While the precise function of Sig1R has long remained mysterious, recent studies have shed light on its role and the molecular mechanisms triggered. Sig1R is in fact a stress-activated chaperone mainly associated with the ER-mitochondria interface that can regulate cell survival through the control of calcium homeostasis. Sig1R functionally regulates ion channels belonging to various molecular families and it has thus been involved in neuronal plasticity and central nervous system diseases. Interestingly, Sig1R is frequently expressed in tumors but its function in cancer has not been yet clarified. In this review, we discuss the current understanding of Sig1R. We suggest herein that Sig1R shapes cancer cell electrical signature upon environmental conditions. Thus, Sig1R may be used as a novel therapeutic target to specifically abrogate pro-invasive functions of ion channels in cancer tissue.
Collapse
Affiliation(s)
- David Crottès
- Université de Nice, UMR 7277 Nice, France ; Institut de Biologie de Valrose, CNRS UMR 7277, INSERM U1091, Université de Nice Nice, France
| | | | | | | | | |
Collapse
|
47
|
Kourrich S, Hayashi T, Chuang JY, Tsai SY, Su TP, Bonci A. Dynamic interaction between sigma-1 receptor and Kv1.2 shapes neuronal and behavioral responses to cocaine. Cell 2013; 152:236-47. [PMID: 23332758 PMCID: PMC4159768 DOI: 10.1016/j.cell.2012.12.004] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 10/05/2012] [Accepted: 11/21/2012] [Indexed: 11/21/2022]
Abstract
The sigma-1 receptor (Sig-1R), an endoplasmic reticulum (ER) chaperone protein, is an interorganelle signaling modulator that potentially plays a role in drug-seeking behaviors. However, the brain site of action and underlying cellular mechanisms remain unidentified. We found that cocaine exposure triggers a Sig-1R-dependent upregulation of D-type K(+) current in the nucleus accumbens (NAc) that results in neuronal hypoactivity and thereby enhances behavioral cocaine response. Combining ex vivo and in vitro studies, we demonstrated that this neuroadaptation is caused by a persistent protein-protein association between Sig-1Rs and Kv1.2 channels, a phenomenon that is associated to a redistribution of both proteins from intracellular compartments to the plasma membrane. In conclusion, the dynamic Sig-1R-Kv1.2 complex represents a mechanism that shapes neuronal and behavioral response to cocaine. Functional consequences of Sig-1R binding to K(+) channels may have implications for other chronic diseases where maladaptive intrinsic plasticity and Sig-1Rs are engaged.
Collapse
Affiliation(s)
- Saïd Kourrich
- Cellular Neurobiology Branch, Intramural Research Program, National Institute on Drug Abuse, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Teruo Hayashi
- Cellular Stress Signaling Unit, Intramural Research Program, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD 21224, USA
- Cellular Pathobiology Section, Intramural Research Program, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Jian-Ying Chuang
- Cellular Pathobiology Section, Intramural Research Program, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Shang-Yi Tsai
- Cellular Pathobiology Section, Intramural Research Program, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Intramural Research Program, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Antonello Bonci
- Cellular Neurobiology Branch, Intramural Research Program, National Institute on Drug Abuse, 251 Bayview Boulevard, Baltimore, MD 21224, USA
- Department of Neurology, University of California, San Francisco, CA, USA
- Solomon H Snyder Neuroscience Institute, Johns Hopkins University School of Medicine, Baltimore, CA, USA
| |
Collapse
|
48
|
Kourrich S, Su TP, Fujimoto M, Bonci A. The sigma-1 receptor: roles in neuronal plasticity and disease. Trends Neurosci 2012; 35:762-71. [PMID: 23102998 PMCID: PMC3587126 DOI: 10.1016/j.tins.2012.09.007] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 08/14/2012] [Accepted: 09/20/2012] [Indexed: 12/31/2022]
Abstract
Sigma-1 receptors (Sig-1Rs) have been implicated in many neurological and psychiatric conditions. Sig-1Rs are intracellular chaperones that reside specifically at the endoplasmic reticulum (ER)-mitochondrion interface, referred to as the mitochondrion-associated ER membrane (MAM). Here, Sig-1Rs regulate ER-mitochondrion Ca(2+) signaling. In this review, we discuss the current understanding of Sig-1R functions. Based on this, we suggest that the key cellular mechanisms linking Sig-1Rs to neurological disorders involve the translocation of Sig-1Rs from the MAM to other parts of the cell, whereby Sig-1Rs bind and modulate the activities of various ion channels, receptors, or kinases. Thus, Sig-1Rs and their associated ligands may represent new avenues for treating aspects of neurological and psychiatric diseases.
Collapse
Affiliation(s)
- Saïd Kourrich
- Synaptic Plasticity Section, Intramural Research Program, National Institute on Drug Abuse (NIDA), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Baltimore, MD, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD USA
| | - Michiko Fujimoto
- Cellular Pathobiology Section, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD USA
| | - Antonello Bonci
- Synaptic Plasticity Section, Intramural Research Program, National Institute on Drug Abuse (NIDA), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Baltimore, MD, USA
- Department of Neurology, University of California, San Francisco, CA, USA
- Solomon H. Snyder Neuroscience Institute, Johns Hopkins University School of Medicine, Baltimore, CA, USA
| |
Collapse
|
49
|
Balasuriya D, Stewart AP, Crottès D, Borgese F, Soriani O, Edwardson JM. The sigma-1 receptor binds to the Nav1.5 voltage-gated Na+ channel with 4-fold symmetry. J Biol Chem 2012; 287:37021-9. [PMID: 22952230 PMCID: PMC3481303 DOI: 10.1074/jbc.m112.382077] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/20/2012] [Indexed: 12/19/2022] Open
Abstract
The sigma-1 receptor (Sig1R) is up-regulated in many human tumors and plays a role in the control of cancer cell proliferation and invasiveness. At the molecular level, the Sig1R modulates the activity of various ion channels, apparently through a direct interaction. We have previously shown using atomic force microscopy imaging that the Sig1R binds to the trimeric acid-sensing ion channel 1A with 3-fold symmetry. Here, we investigated the interaction between the Sig1R and the Nav1.5 voltage-gated Na(+) channel, which has also been implicated in promoting the invasiveness of cancer cells. We show that the Sig1R and Nav1.5 can be co-isolated from co-transfected cells, consistent with an intimate association between the two proteins. Atomic force microscopy imaging of the co-isolated proteins revealed complexes in which Nav1.5 was decorated by Sig1Rs. Frequency distributions of angles between pairs of bound Sig1Rs had two peaks, at ∼90° and ∼180°, and the 90° peak was about twice the size of the 180° peak. These results demonstrate that the Sig1R binds to Nav1.5 with 4-fold symmetry. Hence, each set of six transmembrane regions in Nav1.5 likely constitutes a Sig1R binding site, suggesting that the Sig1R interacts with the transmembrane regions of its partners. Interestingly, two known Sig1R ligands, haloperidol and (+)-pentazocine, disrupted the Nav1.5/Sig1R interaction both in vitro and in living cells. Finally, we show that endogenously expressed Sig1R and Nav1.5 also functionally interact.
Collapse
Affiliation(s)
- Dilshan Balasuriya
- From the Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom and
| | - Andrew P. Stewart
- From the Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom and
| | - David Crottès
- Institut de Biologie de Valrose, CNRS UMR 7277, INSERM U1091 UNS, Faculté des Sciences, Université de Nice Sophia Antipolis, 06108 Nice Cedex 2, France
| | - Franck Borgese
- Institut de Biologie de Valrose, CNRS UMR 7277, INSERM U1091 UNS, Faculté des Sciences, Université de Nice Sophia Antipolis, 06108 Nice Cedex 2, France
| | - Olivier Soriani
- Institut de Biologie de Valrose, CNRS UMR 7277, INSERM U1091 UNS, Faculté des Sciences, Université de Nice Sophia Antipolis, 06108 Nice Cedex 2, France
| | - J. Michael Edwardson
- From the Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom and
| |
Collapse
|