1
|
Ventura-Martínez R, Ángeles-López GE, González-Ugalde D, Domínguez-Páez T, Navarrete-Vázquez G, Jaimez R, Déciga-Campos M. Antinociceptive effect of LMH-2, a new sigma-1 receptor antagonist analog of haloperidol, on the neuropathic pain of diabetic mice. Biomed Pharmacother 2024; 174:116524. [PMID: 38574622 DOI: 10.1016/j.biopha.2024.116524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024] Open
Abstract
This study evaluates the antiallodynic and antihyperalgesic effects of LMH-2, a new haloperidol (HAL) analog that acts as sigma-1 receptor (σ1 R) antagonist, in diabetic mice using a model of neuropathic pain induced by chronic hyperglycemia. Additionally, we compared its effects with those of HAL. Hyperglycemia was induced in mice by nicotinamide-streptozotocin administration (NA-STZ, 50-130 mg/kg). Four weeks later, mechanical allodynia was assessed using the up-down method, and hyperalgesia was evoked with formalin 0.5%. We evaluated antiallodynic and antihyperalgesic effects of LMH-2 (5.6-56.2 mg/kg), HAL (0.018-0.18 mg/kg) and gabapentin (GBP, 5.6-56.2 mg/kg). The results showed that LMH-2 had a more significant antiallodynic effect compared to HAL and GBP (90.4±8.7 vs 75.1±3.1 and 41.9±2.3%, respectively; P<0.05), as well as an antihyperalgesic effect (96.3±1.2 vs 86.9±7.41 and 86.9±4.8%, respectively; P<0.05). Moreover, the antiallodynic and antihyperalgesic effect of both LMH-2 and HAL were completely abolished by PRE-084 (σ1 R agonist); and partially by pramipexole (a D2-like receptor agonist). Finally, the effect of all treatments on the rotarod test, barra, open field and exploratory behaviors showed that LMH-2 did not alter the animals' balance or the exploratory behavior, unlike as HAL or GBP. The molecular docking included indicate that LMH-2 has lower affinity to the D2R than HAL. These results provide evidence that LMH-2 exerts its antinociceptive effects as a σ1 R antagonist without the adverse effects induced by HAL or GBP. Consequently, LMH-2 can be considered a good and safe strategy for treating neuropathic pain caused by hyperglycemia in patients with diabetes.
Collapse
Affiliation(s)
- Rosa Ventura-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Av Universidad No. 3000, Col. Ciudad Universitaria, Alcaldía Coyoacán, Ciudad de México 04510, Mexico.
| | - Guadalupe Esther Ángeles-López
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Av Universidad No. 3000, Col. Ciudad Universitaria, Alcaldía Coyoacán, Ciudad de México 04510, Mexico
| | - Diana González-Ugalde
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico
| | - Tania Domínguez-Páez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico
| | - Gabriel Navarrete-Vázquez
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca, Morelos, Mexico
| | - Ruth Jaimez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Av Universidad No. 3000, Col. Ciudad Universitaria, Alcaldía Coyoacán, Ciudad de México 04510, Mexico
| | - Myrna Déciga-Campos
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico.
| |
Collapse
|
2
|
Couly S, Yasui Y, Foncham S, Grammatikakis I, Lal A, Shi L, Su TP. Benzomorphan and non-benzomorphan agonists differentially alter sigma-1 receptor quaternary structure, as does types of cellular stress. Cell Mol Life Sci 2024; 81:14. [PMID: 38191696 PMCID: PMC10774196 DOI: 10.1007/s00018-023-05023-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 01/10/2024]
Abstract
Sigma-1 receptor (S1R) is a calcium-sensitive, ligand-operated receptor chaperone present on the endoplasmic reticulum (ER) membrane. S1R plays an important role in ER-mitochondrial inter-organelle calcium signaling and cell survival. S1R and its agonists confer resilience against various neurodegenerative diseases; however, the molecular mechanism of S1R is not yet fully understood. At resting state, S1R is either in a monomeric or oligomeric state but the ratio of these concentrations seems to change upon activation of S1R. S1R is activated by either cellular stress, such as ER-calcium depletion, or ligands. While the effect of ligands on S1R quaternary structure remains unclear, the effect of cellular stress has not been studied. In this study we utilize cellular and an in-vivo model to study changes in quaternary structure of S1R upon activation. We incubated cells with cellular stressors (H2O2 and thapsigargin) or exogenous ligands, then quantified monomeric and oligomeric forms. We observed that benzomorphan-based S1R agonists induce monomerization of S1R and decrease oligomerization, which was confirmed in the liver tissue of mice injected with (+)-Pentazocine. Antagonists block this effect but do not induce any changes when used alone. Oxidative stress (H2O2) increases the monomeric/oligomeric S1R ratio whereas ER calcium depletion (thapsigargin) has no effect. We also analyzed the oligomerization ability of various truncated S1R fragments and identified the fragments favorizing oligomerization. In this publication we demonstrate that quaternary structural changes differ according to the mechanism of S1R activation. Therefore, we offer a novel perspective on S1R activation as a nuanced phenomenon dependent on the type of stimulus.
Collapse
Affiliation(s)
- Simon Couly
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute On Drug Abuse, NIH/DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute On Drug Abuse, NIH/DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Semnyonga Foncham
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute On Drug Abuse, NIH/DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Ioannis Grammatikakis
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, 20892, USA
| | - Ashish Lal
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, 20892, USA
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute On Drug Abuse, NIH/DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute On Drug Abuse, NIH/DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| |
Collapse
|
3
|
Kim M, Bezprozvanny I. Structure-Based Modeling of Sigma 1 Receptor Interactions with Ligands and Cholesterol and Implications for Its Biological Function. Int J Mol Sci 2023; 24:12980. [PMID: 37629160 PMCID: PMC10455549 DOI: 10.3390/ijms241612980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The sigma 1 receptor (S1R) is a 223-amino-acid-long transmembrane endoplasmic reticulum (ER) protein. The S1R plays an important role in neuronal health and it is an established therapeutic target for neurodegenerative and neuropsychiatric disorders. Despite its importance in physiology and disease, the biological function of S1R is poorly understood. To gain insight into the biological and signaling functions of S1R, we took advantage of recently reported crystal structures of human and Xenopus S1Rs and performed structural modeling of S1R interactions with ligands and cholesterol in the presence of the membrane. By combining bioinformatics analysis of S1R sequence and structural modelling approaches, we proposed a model that suggests that S1R may exist in two distinct conformations-"dynamic monomer" (DM) and "anchored monomer" (AM). We further propose that equilibrium between AM and DM conformations of S1R is essential for its biological function in cells, with AM conformation facilitating the oligomerization of S1R and DM conformation facilitating deoligomerization. Consistent with experimental evidence, our hypothesis predicts that increased levels of membrane cholesterol and S1R antagonists should promote the oligomeric state of S1R, but S1R agonists and pathogenic mutations should promote its deoligomerization. Obtained results provide mechanistic insights into signaling functions of S1R in cells, and the proposed model may help to explain neuroprotective effects of S1R modulators.
Collapse
Affiliation(s)
- Meewhi Kim
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
- Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg State Polytechnical University, 195251 St. Petersburg, Russia
| |
Collapse
|
4
|
Couly S, Yasui Y, Su TP. SIGMAR1 Confers Innate Resilience against Neurodegeneration. Int J Mol Sci 2023; 24:ijms24097767. [PMID: 37175473 PMCID: PMC10178636 DOI: 10.3390/ijms24097767] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
The sigma-1 receptor (SIGMAR1) is one of a kind: a receptor chaperone protein. This 223 amino acid-long protein is enriched at the mitochondria-associated endoplasmic reticulum membrane (MAM), a specialized microdomain of the endoplasmic reticulum that is structurally and functionally connected to the mitochondria. As a receptor, SIGMAR1 binds a wide spectrum of ligands. Numerous molecules targeting SIGMAR1 are currently in pre-clinical or clinical development. Interestingly, the range of pathologies covered by these studies is broad, especially with regard to neurodegenerative disorders. Upon activation, SIGMAR1 can translocate and interact with other proteins, mostly at the MAM but also in other organelles, which allows SIGMAR1 to affect many cellular functions. During these interactions, SIGMAR1 exhibits chaperone protein behavior by participating in the folding and stabilization of its partner. In this short communication, we will shed light on how SIGMAR1 confers protection against neurodegeneration to the cells of the nervous system and why this ability makes SIGMAR1 a multifunctional therapeutic prospect.
Collapse
Affiliation(s)
- Simon Couly
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, 333 Cassell Drive, Baltimore, MD 21224, USA
| |
Collapse
|
5
|
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
|
6
|
VanderZwaag J, Halvorson T, Dolhan K, Šimončičová E, Ben-Azu B, Tremblay MÈ. The Missing Piece? A Case for Microglia's Prominent Role in the Therapeutic Action of Anesthetics, Ketamine, and Psychedelics. Neurochem Res 2023; 48:1129-1166. [PMID: 36327017 DOI: 10.1007/s11064-022-03772-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
There is much excitement surrounding recent research of promising, mechanistically novel psychotherapeutics - psychedelic, anesthetic, and dissociative agents - as they have demonstrated surprising efficacy in treating central nervous system (CNS) disorders, such as mood disorders and addiction. However, the mechanisms by which these drugs provide such profound psychological benefits are still to be fully elucidated. Microglia, the CNS's resident innate immune cells, are emerging as a cellular target for psychiatric disorders because of their critical role in regulating neuroplasticity and the inflammatory environment of the brain. The following paper is a review of recent literature surrounding these neuropharmacological therapies and their demonstrated or hypothesized interactions with microglia. Through investigating the mechanism of action of psychedelics, such as psilocybin and lysergic acid diethylamide, ketamine, and propofol, we demonstrate a largely under-investigated role for microglia in much of the emerging research surrounding these pharmacological agents. Among others, we detail sigma-1 receptors, serotonergic and γ-aminobutyric acid signalling, and tryptophan metabolism as pathways through which these agents modulate microglial phagocytic activity and inflammatory mediator release, inducing their therapeutic effects. The current review includes a discussion on future directions in the field of microglial pharmacology and covers bidirectional implications of microglia and these novel pharmacological agents in aging and age-related disease, glial cell heterogeneity, and state-of-the-art methodologies in microglial research.
Collapse
Affiliation(s)
- Jared VanderZwaag
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Torin Halvorson
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Vancouver, BC, Canada
- Department of Biology, University of Victoria, Vancouver, BC, Canada
| | - Eva Šimončičová
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de médecine moléculaire, Université Laval, Québec City, QC, Canada.
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
7
|
Zhang Y, Ye G, Chen Y, Sheng C, Wang J, Kong L, Yuan L, Lin C. Veratramine ameliorates pain symptoms in rats with diabetic peripheral neuropathy by inhibiting activation of the SIGMAR1-NMDAR pathway. PHARMACEUTICAL BIOLOGY 2022; 60:2145-2154. [PMID: 36373991 PMCID: PMC9665081 DOI: 10.1080/13880209.2022.2136207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/19/2022] [Accepted: 10/08/2022] [Indexed: 06/16/2023]
Abstract
CONTEXT Veratramine may have a potential therapeutic effect for diabetic peripheral neuropathy (DPN). OBJECTIVE To evaluate whether veratramine ameliorates neuropathic pain in a rat diabetic model. MATERIALS AND METHODS Sprague-Dawley rats were used for a diabetic model induced by a streptozotocin + high-fat diet. Two months after the induction of the diabetic model, the rats with DPN were screened according to the mechanical pain threshold. The rats with DPN were divided into a model group (n = 12) and a treated group (n = 12). Rats with diabetes, but without peripheral neuropathy, were used in the vehicle group (n = 9). The treatment group received 50 μg/kg veratramine via the tail vein once a day for 4 weeks. During modelling and treatment, rats in all three groups were fed a high-fat diet. RESULTS The mechanical withdrawal threshold increased from 7.5 ± 1.9 N to 17.9 ± 2.6 N in DPN rats treated with veratramine. The tolerance time of the treated group to hot and cold ectopic pain increased from 11.8 ± 4.2 s and 3.4 ± 0.8 s to 20.4 ± 4.1 s and 5.9 ± 1.7 s, respectively. Veratramine effectively alleviated L4-L5 spinal cord and sciatic nerve pathological injury. Veratramine inhibited the expression of SIGMAR1 and the phosphorylation of the N-methyl-d-aspartate receptor (NMDAR) Ser896 site in spinal cord tissue, as well as inhibited the formation of SIGMAR1-NMDAR and NMDAR-CaMKII complexes. DISCUSSION AND CONCLUSIONS Veratramine may alleviate the occurrence of pain symptoms in rats with DPN by inhibiting activation of the SIGMAR1-NMDAR pathway.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo, P. R. China
| | - Guangyao Ye
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo, P. R. China
| | - Yuebo Chen
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo, P. R. China
| | - Chaoxu Sheng
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo, P. R. China
| | - Jianlin Wang
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo, P. R. China
| | - Lingsi Kong
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo, P. R. China
| | - Liyong Yuan
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo, P. R. China
| | - Chunyan Lin
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo, P. R. China
| |
Collapse
|
8
|
Antidepressive-like Behavior-Related Metabolomic Signatures of Sigma-1 Receptor Knockout Mice. Biomedicines 2022; 10:biomedicines10071572. [PMID: 35884876 PMCID: PMC9313356 DOI: 10.3390/biomedicines10071572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023] Open
Abstract
Sigma-1 receptor (Sig1R) has been proposed as a therapeutic target for neurological, neurodegenerative, and psychiatric disorders, including depression and anxiety. Identifying metabolites that are affected by Sig1R absence and cross-referencing them with specific mood-related behaviors would be helpful for the development of new therapies for Sig1R-associated disorders. Here, we examined metabolic profiles in the blood and brains of male CD-1 background Sig1R knockout (KO) mice in adulthood and old age and correlated them with the assessment of depression- and anxiety-related behaviors. The most pronounced changes in the metabolic profile were observed in the plasma of adult Sig1R KO mice. In adult mice, the absence of Sig1R significantly influenced the amino acid, sphingolipid (sphingomyelin and ceramide (18:1)), and serotonin metabolic pathways. There were higher serotonin levels in plasma and brain tissue and higher histamine levels in the plasma of Sig1R KO mice than in their age-matched wild-type counterparts. This increase correlated with the reduced behavioral despair in the tail suspension test and lack of anhedonia in the sucrose preference test. Overall, these results suggest that Sig1R regulates behavior by altering serotonergic and histaminergic systems and the sphingolipid metabolic pathway.
Collapse
|
9
|
Couly S, Goguadze N, Yasui Y, Kimura Y, Wang SM, Sharikadze N, Wu HE, Su TP. Knocking Out Sigma-1 Receptors Reveals Diverse Health Problems. Cell Mol Neurobiol 2022; 42:597-620. [PMID: 33095392 PMCID: PMC8062587 DOI: 10.1007/s10571-020-00983-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
Sigma-1 receptor (Sig-1R) is a protein present in several organs such as brain, lung, and heart. In a cell, Sig-1R is mainly located across the membranes of the endoplasmic reticulum and more specifically at the mitochondria-associated membranes. Despite numerous studies showing that Sig-1R could be targeted to rescue several cellular mechanisms in different pathological conditions, less is known about its fundamental relevance. In this review, we report results from various studies and focus on the importance of Sig-1R in physiological conditions by comparing Sig-1R KO mice to wild-type mice in order to investigate the fundamental functions of Sig-1R. We note that the Sig-1R deletion induces cognitive, psychiatric, and motor dysfunctions, but also alters metabolism of heart. Finally, taken together, observations from different experiments demonstrate that those dysfunctions are correlated to poor regulation of ER and mitochondria metabolism altered by stress, which could occur with aging.
Collapse
Affiliation(s)
- Simon Couly
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA.
| | - Nino Goguadze
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Yuriko Kimura
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Shao-Ming Wang
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Nino Sharikadze
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Hsiang-En Wu
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| |
Collapse
|
10
|
Martin P, Maurice T, Gammaitoni A, Farfel G, Boyd B, Galer B. Fenfluramine modulates the anti-amnesic effects induced by sigma-1 receptor agonists and neuro(active)steroids in vivo. Epilepsy Behav 2022; 127:108526. [PMID: 35007961 DOI: 10.1016/j.yebeh.2021.108526] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022]
Abstract
Fenfluramine (N-ethyl-α-methl-3-(trifluoromethyl)phenethylamine) is an anti-seizure medication (ASM) particularly effective in patients with Dravet syndrome, a severe treatment-resistant epileptic encephalopathy. Fenfluramine acts not only as neuronal serotonin (5-HT) releaser but also as a positive modulator of the sigma-1 receptor (S1R). We here examined the modulatory activity of Fenfluramine on the S1R-mediated anti-amnesic response in mice using combination analyses. Fenfluramine and Norfenfluramine, racemate and isomers, were combined with either the S1R agonist (PRE-084) or the S1R-acting neuro(active)steroids, pregnenolone sulfate (PREGS), Dehydroepiandrosterone sulfate (DHEAS), or progesterone. We report that Fenfluramine racemate or (+)-Fenfluramine, in the 0.1-1 mg/kg dose range, attenuated the dizocilpine-induced learning deficits in spontaneous alternation and passive avoidance, and showed low-dose synergies in combination with PRE-084. These effects were blocked by the S1R antagonist NE-100. Dehydroepiandrosterone sulfate or PREGS attenuated dizocilpine-induced learning deficits in the 5-20 mg/kg dose range. Co-treatments at low dose between steroids and Fenfluramine or (+)-Fenfluramine were synergistic. Progesterone blocked Fenfluramine effect. Finally, Fenfluramine and (+)-Fenfluramine effects were prevented by the 5-HT1A receptor antagonist WAY-100635 or 5-HT2A antagonist RS-127445, but not by the 5-HT1B/1D antagonist GR 127935 or the 5-HT2C antagonist SB 242084, confirming a 5-HT1A and 5-HT2A receptor involvement in the drug effect on memory. We therefore confirmed the positive modulation of Fenfluramine racemate or dextroisomer on S1R and showed that, in physiological conditions, the drug potentiated the low dose effects of neuro(active)steroids, endogenous S1R modulators. The latter are potent modulators of the excitatory/inhibitory balance in the brain, and their levels must be considered in the antiepileptic action of Fenfluramine.
Collapse
Affiliation(s)
| | - Tangui Maurice
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France.
| | | | | | | | | |
Collapse
|
11
|
Dai LA, Chen XY, Li WJ, Yang JH, Lin MJ, Li XS, Zeng YF, Chen SW, Xie ZL, Zhu ZL, Li XJ, Huang HS. Sigma-1 Receptor and Binding Immunoglobulin Protein Interact with Ulinastatin Contributing to a Protective Effect in Rat Cerebral Ischemia/Reperfusion. World Neurosurg 2022; 158:e488-e494. [PMID: 34767993 DOI: 10.1016/j.wneu.2021.11.008] [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: 06/10/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To investigate impact of ulinastatin (UTI) on sigma-1 receptor (σ1R) and binding immunoglobulin protein (BiP) after cerebral ischemia/reperfusion injury. METHODS The middle cerebral artery occlusion (MCAO) model was used to induce cerebral ischemia/reperfusion injury. Eighty male Sprague Dawley rats were randomly divided into 6 groups: control, MCAO, MCAO+50,000 U/kg UTI, MCAO+100,000 U/kg UTI, MCAO+200,000 U/kg UTI, MCAO+300,000 U/kg UTI. At 24 and 48 hours after MCAO, infarct volume, neurological dysfunction, and grip strength test were measured, and level of σ1R and BiP proteins was further detected using Western blot. Molecular docking assays were carried out to verify interaction between σ1R, BiP, and UTI. The serum concentration of BiP and the binding assay between σ1R, BiP, and UTI were determined using enzyme-linked immunosorbent assay. RESULTS UTI increased the modified neurological severity score and upregulated σ1R and BiP expression in the cerebral cortex after MCAO. The grip strength of forelimbs increased significantly in the MCAO+200,000 U/kg UTI and MCAO+300,000 U/kg UTI groups compared with the MCAO group, while BiP serum levels remained unchanged. The molecular docking assay indicated putative binding between σ1R, BiP, and UTI. The binding assay also revealed that both σ1R and BiP could be combined with UTI. CONCLUSIONS UTI displays a neuroprotective effect via upregulation of σ1R and BiP during ischemia/reperfusion injury, suggesting that UTI modulates σ1R and BiP and their interaction may provide a novel insight into potential therapeutic mechanisms for stroke.
Collapse
Affiliation(s)
- Ling-Ao Dai
- Department of Anesthesiology, Second Clinical College of Guangzhou Medical University, Guangzhou, China
| | - Xiao-Yin Chen
- Department of Anesthesiology, Second Clinical College of Guangzhou Medical University, Guangzhou, China
| | - Wen-Jing Li
- Department of Anesthesiology, Second Clinical College of Guangzhou Medical University, Guangzhou, China
| | - Jia-Hao Yang
- Department of Anesthesiology, Second Clinical College of Guangzhou Medical University, Guangzhou, China
| | - Ming-Jie Lin
- Department of Anesthesiology, Second Clinical College of Guangzhou Medical University, Guangzhou, China
| | - Xiao-Shan Li
- Department of Anesthesiology, Second Clinical College of Guangzhou Medical University, Guangzhou, China
| | - Yu-Fu Zeng
- Department of Anesthesiology, Second Clinical College of Guangzhou Medical University, Guangzhou, China
| | - Shu-Wen Chen
- Department of Anesthesiology, Second Clinical College of Guangzhou Medical University, Guangzhou, China
| | - Zhu-Liang Xie
- Department of Anesthesiology, Second Clinical College of Guangzhou Medical University, Guangzhou, China
| | - Zhuo-Li Zhu
- Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiong-Juan Li
- Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huan-Sen Huang
- Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Rodríguez-Muñoz M, Cortés-Montero E, Onetti Y, Sánchez-Blázquez P, Garzón-Niño J. The σ1 Receptor and the HINT1 Protein Control α2δ1 Binding to Glutamate NMDA Receptors: Implications in Neuropathic Pain. Biomolecules 2021; 11:1681. [PMID: 34827679 PMCID: PMC8615847 DOI: 10.3390/biom11111681] [Citation(s) in RCA: 3] [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: 09/14/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 02/01/2023] Open
Abstract
Nerve injury produces neuropathic pain through the binding of α2δ1 proteins to glutamate N-methyl-D-aspartate receptors (NMDARs). Notably, mice with a targeted deletion of the sigma 1 receptor (σ1R) gene do not develop neuropathy, whereas mice lacking the histidine triad nucleotide-binding protein 1 (Hint1) gene exhibit exacerbated allodynia. σ1R antagonists more effectively diminish neuropathic pain of spinal origin when administered by intracerebroventricular injection than systemically. Thus, in mice subjected to unilateral sciatic nerve chronic constriction injury (CCI), we studied the participation of σ1Rs and HINT1 proteins in the formation of α2δ1-NMDAR complexes within the supraspinal periaqueductal gray (PAG). We found that δ1 peptides required σ1Rs in order to interact with the NMDAR NR1 variant that contains the cytosolic C1 segment. σ1R antagonists or low calcium levels provoke the dissociation of σ1R-NR1 C1 dimers, while they barely affect the integrity of δ1-σ1R-NR1 C1 trimers. However, HINT1 does remove δ1 peptides from the trimer, thereby facilitating the subsequent dissociation of σ1Rs from NMDARs. In σ1R-/- mice, CCI does not promote the formation of NMDAR-α2δ1 complexes and allodynia does not develop. The levels of α2δ1-σ1R-NMDAR complexes increase in HINT1-/- mice and after inducing CCI, degradation of α2δ1 proteins is observed. Notably, σ1R antagonists but not gabapentinoids alleviate neuropathic pain in these mice. During severe neuropathy, the metabolism of α2δ1 proteins may account for the failure of many patients to respond to gabapentinoids. Therefore, σ1Rs promote and HINT1 proteins hinder the formation α2δ1-NMDAR complexes in the PAG, and hence, the appearance of mechanical allodynia depends on the interplay between these proteins.
Collapse
Affiliation(s)
- María Rodríguez-Muñoz
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, 28002 Madrid, Spain; (M.R.-M.); (E.C.-M.); (Y.O.); (P.S.-B.)
| | - Elsa Cortés-Montero
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, 28002 Madrid, Spain; (M.R.-M.); (E.C.-M.); (Y.O.); (P.S.-B.)
| | - Yara Onetti
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, 28002 Madrid, Spain; (M.R.-M.); (E.C.-M.); (Y.O.); (P.S.-B.)
| | - Pilar Sánchez-Blázquez
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, 28002 Madrid, Spain; (M.R.-M.); (E.C.-M.); (Y.O.); (P.S.-B.)
| | - Javier Garzón-Niño
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Doctor Arce 37, 28002 Madrid, Spain
| |
Collapse
|
13
|
Current development of sigma-2 receptor radioligands as potential tumor imaging agents. Bioorg Chem 2021; 115:105163. [PMID: 34289426 DOI: 10.1016/j.bioorg.2021.105163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/24/2022]
Abstract
Sigma receptors are transmembrane proteins with two different subtypes: σ1 and σ2. Because of its overexpression in tumors, the σ2 receptor (σ2R) is a well-known biomarker for cancer cells. A large number of small-molecule ligands for the σ2Rs have been identified and tested for imaging the proliferative status of tumors using single photon emission computed tomography (SPECT) and positron emission tomography (PET). These small molecules include derivatives of bicyclic amines, indoles, cyclohexylpiperazines and tetrahydroisoquinolines. This review discusses various aspects of small molecule ligands, such as chemical composition, labeling strategy, affinity for σ2Rs, and in vitro/in vivo investigations. The recent studies described here could be useful for the development of σ2R radioligands as potential tumor imaging agents.
Collapse
|
14
|
Zhemkov V, Geva M, Hayden MR, Bezprozvanny I. Sigma-1 Receptor (S1R) Interaction with Cholesterol: Mechanisms of S1R Activation and Its Role in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:4082. [PMID: 33920913 PMCID: PMC8071319 DOI: 10.3390/ijms22084082] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
The sigma-1 receptor (S1R) is a 223 amino acid-long transmembrane endoplasmic reticulum (ER) protein. The S1R modulates the activity of multiple effector proteins, but its signaling functions are poorly understood. S1R is associated with cholesterol, and in our recent studies we demonstrated that S1R association with cholesterol induces the formation of S1R clusters. We propose that these S1R-cholesterol interactions enable the formation of cholesterol-enriched microdomains in the ER membrane. We hypothesize that a number of secreted and signaling proteins are recruited and retained in these microdomains. This hypothesis is consistent with the results of an unbiased screen for S1R-interacting partners, which we performed using the engineered ascorbate peroxidase 2 (APEX2) technology. We further propose that S1R agonists enable the disassembly of these cholesterol-enriched microdomains and the release of accumulated proteins such as ion channels, signaling receptors, and trophic factors from the ER. This hypothesis may explain the pleotropic signaling functions of the S1R, consistent with previously observed effects of S1R agonists in various experimental systems.
Collapse
Affiliation(s)
- Vladimir Zhemkov
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Michal Geva
- Prilenia Therapeutics Development LTD, Herzliya 4673304, Israel; (M.G.); (M.R.H.)
| | - Michael R. Hayden
- Prilenia Therapeutics Development LTD, Herzliya 4673304, Israel; (M.G.); (M.R.H.)
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V6H 3V5, Canada
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
- Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg State Polytechnic University, 195251 St. Petersburg, Russia
| |
Collapse
|
15
|
van der Aart J, Yaqub M, Kooijman EJM, Bakker J, Langermans JAM, Schuit RC, Hofman MBM, Christiaans JAM, Lammertsma AA, Windhorst AD, van Berckel BNM. Evaluation of the Novel PET Tracer [ 11C]HACH242 for Imaging the GluN2B NMDA Receptor in Non-Human Primates. Mol Imaging Biol 2020; 21:676-685. [PMID: 30306318 DOI: 10.1007/s11307-018-1284-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE There are currently no positron emission tomography (PET) radiotracers for the GluN2B (NR2B) binding sites of brain N-methyl-D-aspartate (NMDA) receptors. In rats, the GluN2B antagonist Ro25-6981 reduced the binding of N-((5-(4-fluoro-2-[11C]methoxyphenyl)pyridin-3-yl)methyl)cyclopentanamin ([11C]HACH242). This paper reports the evaluation of [11C]HACH242 PET in non-human primates at baseline and following administration of the GluN2B negative allosteric modulator radiprodil. PROCEDURES Eight 90-min dynamic [11C]HACH242 PET scans were acquired in three male anaesthetised rhesus monkeys, including a retest session of subject 1, at baseline and 10 min after intravenous 10 mg/kg radiprodil. Standardised uptake values (SUV) were calculated for 9 brain regions. Arterial blood samples were taken at six timepoints to characterise pharmacokinetics in blood and plasma. Reliable input functions for kinetic modelling could not be generated due to variability in the whole-blood radioactivity measurements. RESULTS [11C]HACH242 entered the brain and displayed fairly uniform uptake. The mean (± standard deviation, SD) Tmax was 17 ± 7 min in baseline scans and 24 ± 15 min in radiprodil scans. The rate of radioligand metabolism in plasma (primarily to polar metabolites) was high, with mean parent fractions of 26 ± 10 % at 20 min and 8 ± 5 % at 85 min. Radiprodil increased [11C]HACH242 whole-brain SUV in the last PET frame by 25 %, 1 %, 3 and 17 % for subjects 1, 2, 3 and retest of subject 1, respectively. The mean brain to plasma ratio was 5.4 ± 2.6, and increased by 39 to 110 % in the radiprodil condition, partly due to lower parent plasma radioactivity of -11 to -56 %. CONCLUSIONS The present results show that [11C]HACH242 has a suitable kinetic profile in the brain and low accumulation of lipophilic radiometabolites. Radiprodil did not consistently change [11C]HACH242 brain uptake. These findings may be explained by variations in cerebral blood flow, a low fraction of specifically bound tracer, or interactions with endogenous NMDA receptor ligands at the binding site. Further experiments of ligand interactions are necessary to facilitate the development of radiotracers for in vivo imaging of the ionotropic NMDA receptor.
Collapse
Affiliation(s)
- Jasper van der Aart
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands. .,Centre for Human Drug Research, Leiden, The Netherlands.
| | - Maqsood Yaqub
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Esther J M Kooijman
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Jaco Bakker
- Animal Science Department, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Jan A M Langermans
- Animal Science Department, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Robert C Schuit
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Mark B M Hofman
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Johannes A M Christiaans
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Cortés-Montero E, Sánchez-Blázquez P, Onetti Y, Merlos M, Garzón J. Ligands Exert Biased Activity to Regulate Sigma 1 Receptor Interactions With Cationic TRPA1, TRPV1, and TRPM8 Channels. Front Pharmacol 2019; 10:634. [PMID: 31249525 PMCID: PMC6582314 DOI: 10.3389/fphar.2019.00634] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 05/17/2019] [Indexed: 11/13/2022] Open
Abstract
The sigma 1 receptor (σ1R) and the mu-opioid receptor (MOR) regulate the transient receptor potential (TRP) V1 calcium channel. A series of proteins are involved in the cross-regulation between MORs and calcium channels like the glutamate N-methyl-D-aspartate receptor (NMDAR), including the histidine triad nucleotide-binding protein 1 (HINT1), calmodulin (CaM), and the σ1R. Thus, we assessed whether similar mechanisms also apply to the neural TRP ankyrin member 1 (TRPA1), TRP vanilloid member 1 (TRPV1), and TRP melastatin member 8 (TRPM8). Our results indicate that σ1R and CaM bound directly to cytosolic regions of these TRPs, and this binding increased in the presence of calcium. By contrast, the association of HINT1 with these TRPs was moderately dependent on calcium. The σ1R always competed with CaM for binding to the TRPs, except for its binding to the TRPA1 C-terminal where σ1R binding cooperated with that of CaM. However, σ1R dampened HINT1 binding to the TRPA1 N-terminal. When the effect of σ1R ligands was addressed, the σ1R agonists PRE084 and pregnenolone sulfate enhanced the association of the σ1R with the TRPM8 N-terminal and TRPV1 C-terminal in the presence of physiological calcium, as seen for the σ1R-NMDAR interactions. However, these agonists dampened σ1R binding to the TRPA1 and TRPV1 N-terminal domains, and also to the TRPA1 C-terminal, as seen for σ1R-binding immunoglobulin protein (BiP) interactions in the endoplasmic reticulum (ER). By contrast, the σ1R antagonists progesterone and S1RA reduced the association of σ1R with TRPA1 and TRPV1 C-terminal regions, as seen for the σ1R-NMDAR interactions. Conversely, they enhanced the σ1R interaction with the TRPA1 N-terminal, as seen for σ1R-BiP interactions, whereas they barely affected the association of σ1R with the TRPV1 N-terminal. Thus, depending on the calcium channel and the cytosolic region examined, the σ1R agonists pregnenolone sulfate and PRE084 opposed or collaborated with the σ1R antagonists progesterone and S1RA to disrupt or promote such interactions. Through the use of cloned cytosolic regions of selected TRP calcium channels, we were able to demonstrate that σ1R ligands exhibit biased activity to regulate particular σ1R interactions with other proteins. Since σ1Rs are implicated in essential physiological processes, exploiting such ligand biases may represent a means to develop more selective and efficacious pharmacological interventions.
Collapse
Affiliation(s)
- Elsa Cortés-Montero
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, Madrid, Spain
| | - Pilar Sánchez-Blázquez
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, Madrid, Spain
| | - Yara Onetti
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, Madrid, Spain
| | - Manuel Merlos
- Drug Discovery & Preclinical Development, Esteve, Barcelona, Spain
| | - Javier Garzón
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, Madrid, Spain
| |
Collapse
|
17
|
Carcolé M, Zamanillo D, Merlos M, Fernández-Pastor B, Cabañero D, Maldonado R. Blockade of the Sigma-1 Receptor Relieves Cognitive and Emotional Impairments Associated to Chronic Osteoarthritis Pain. Front Pharmacol 2019; 10:468. [PMID: 31130863 PMCID: PMC6510262 DOI: 10.3389/fphar.2019.00468] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/12/2019] [Indexed: 12/30/2022] Open
Abstract
Osteoarthritis is the most common musculoskeletal disease worldwide, often characterized by degradation of the articular cartilage, chronic joint pain and disability. Cognitive dysfunction, anxiety and depression are common comorbidities that impact the quality of life of these patients. In this study, we evaluated the involvement of sigma-1 receptor (σ1R) on the nociceptive, cognitive and emotional alterations associated with chronic osteoarthritis pain. Monosodium iodoacetate (MIA) was injected into the knee of Swiss-albino CD1 mice to induce osteoarthritis pain, which then received a repeated treatment with the σ1R antagonist E-52862 or its vehicle. Nociceptive responses and motor performance were assessed with the von Frey and the Catwalk gait tests. Cognitive alterations were evaluated using the novel object recognition task, anxiety-like behavior with the elevated plus maze and the zero-maze tests, whereas depressive-like responses were determined using the forced swimming test. We also studied the local effect of the σ1R antagonist on cartilage degradation, and its central effects on microglial reactivity in the medial prefrontal cortex. MIA induced mechanical allodynia and gait abnormalities that were prevented by the chronic treatment with the σ1R antagonist. E-52862 also reduced the memory impairment and the depressive-like behavior associated to osteoarthritis pain. Interestingly, the effect of E-52862 on depressive-like behavior was not accompanied by a modification of anxiety-like behavior. The pain-relieving effects of the σ1R antagonist were not due to a local effect on the articular cartilage, since E-52862 treatment did not modify the histological alterations of the knee joints. However, E-52862 induced central effects revealed by a reduction of the cortical microgliosis observed in mice with osteoarthritis pain. These findings show that σ1R antagonism inhibits mechanical hypersensitivity, cognitive deficits and depressive-like states associated with osteoarthritis pain in mice. These effects are associated with central modulation of glial activity but are unrelated to changes in cartilage degradation. Therefore, targeting the σ1R with E-52862 represents a promising pharmacological approach with effects on multiple aspects of chronic osteoarthritis pain that may go beyond the strict inhibition of nociception.
Collapse
Affiliation(s)
- Mireia Carcolé
- Neuropharmacology Laboratory, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Daniel Zamanillo
- Drug Discovery and Preclinical Development, Laboratories Esteve, Barcelona Science Park, Barcelona, Spain
| | - Manuel Merlos
- Drug Discovery and Preclinical Development, Laboratories Esteve, Barcelona Science Park, Barcelona, Spain
| | - Begoña Fernández-Pastor
- Drug Discovery and Preclinical Development, Laboratories Esteve, Barcelona Science Park, Barcelona, Spain
| | - David Cabañero
- Neuropharmacology Laboratory, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Rafael Maldonado
- Neuropharmacology Laboratory, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| |
Collapse
|
18
|
Jia J, Cheng J, Wang C, Zhen X. Sigma-1 Receptor-Modulated Neuroinflammation in Neurological Diseases. Front Cell Neurosci 2018; 12:314. [PMID: 30294261 PMCID: PMC6158303 DOI: 10.3389/fncel.2018.00314] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/29/2018] [Indexed: 02/02/2023] Open
Abstract
A large body of evidence indicates that sigma-1 receptors (Sig-1R) are important drug targets for a number of neuropsychiatric disorders. Sig-1Rs are enriched in central nervous system (CNS). In addition to neurons, both cerebral microglia and astrocytes express Sig-1Rs. Activation of Sig-1Rs is known to elicit potent neuroprotective effects and promote neuronal survival via multiple mechanisms, including promoting mitochondrial functions, decreasing oxidative stress and regulating neuroimmnological functions. In this review article, we focus on the emerging role of Sig-1Rs in regulating neuroinflammation and discuss the recent advances on the Sig-1R-modulating neuroinflammation in the pathophysiology and therapy of neurodegenerative disorders.
Collapse
Affiliation(s)
- Jia Jia
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jian Cheng
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Cheng Wang
- Department of Pharmacy, Suzhou Science and Technology Town Hospital, Suzhou, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
19
|
Imaging sigma receptors in the brain: New opportunities for diagnosis of Alzheimer's disease and therapeutic development. Neurosci Lett 2018; 691:3-10. [PMID: 30040970 DOI: 10.1016/j.neulet.2018.07.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/09/2018] [Accepted: 07/20/2018] [Indexed: 10/28/2022]
Abstract
The sigma-1 (σ1) receptor is a chaperone protein located on the mitochondria-associated membrane of the endoplasmic reticulum, while the sigma-2 receptor (σ2) is an endoplasmic reticulum-resident membrane protein. Recent evidence indicates that both of these receptors figure prominently in the pathophysiology of Alzheimer's disease (AD) and thus are targets for the development of novel, disease-modifying therapeutic strategies. Radioligand-based molecular imaging technique such as positron emission tomography (PET) imaging is a powerful tool for the investigation of protein target expression and function in living subjects. In this review, we survey the development of PET radioligands for the σ1 or σ2 receptors and assess their potential for human imaging applications. The availability of PET imaging with σ1 or σ2 receptor-specific radioligands in humans will allow the investigation of these receptors in vivo and lead to further understanding of their respective roles in AD pathogenesis and progression. Moreover, PET imaging can be used in target occupancy studies to assess target engagement and correlate receptor occupancy and therapeutic response of σ1 receptor agonists and σ2 receptor antagonists currently in clinical trials. It is expected that neuroimaging of σ1 and σ2 receptors in the brain will shed new light on AD pathophysiology and may provide us with new biomarkers for diagnosis of AD and efficacy monitoring of emerging AD therapeutic strategies.
Collapse
|
20
|
Montilla-García Á, Perazzoli G, Tejada MÁ, González-Cano R, Sánchez-Fernández C, Cobos EJ, Baeyens JM. Modality-specific peripheral antinociceptive effects of μ-opioid agonists on heat and mechanical stimuli: Contribution of sigma-1 receptors. Neuropharmacology 2018; 135:328-342. [PMID: 29580951 DOI: 10.1016/j.neuropharm.2018.03.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 03/05/2018] [Accepted: 03/20/2018] [Indexed: 11/16/2022]
Abstract
Morphine induces peripherally μ-opioid-mediated antinociception to heat but not to mechanical stimulation. Peripheral sigma-1 receptors tonically inhibit μ-opioid antinociception to mechanical stimuli, but it is unknown whether they modulate μ-opioid heat antinociception. We hypothesized that sigma-1 receptors might play a role in the modality-specific peripheral antinociceptive effects of morphine and other clinically relevant μ-opioid agonists. Mechanical nociception was assessed in mice with the paw pressure test (450 g), and heat nociception with the unilateral hot plate (55 °C) test. Local peripheral (intraplantar) administration of morphine, buprenorphine or oxycodone did not induce antinociception to mechanical stimulation but had dose-dependent antinociceptive effects on heat stimuli. Local sigma-1 antagonism unmasked peripheral antinociception by μ-opioid agonists to mechanical stimuli, but did not modify their effects on heat stimulation. TRPV1+ and IB4+ cells are segregated populations of small neurons in the dorsal root ganglia (DRG) and the density of sigma-1 receptors was higher in IB4+ cells than in the rest of small nociceptive neurons. The in vivo ablation of TRPV1-expressing neurons with resiniferatoxin did not alter IB4+ neurons in the DRG, mechanical nociception, or the effects of sigma-1 antagonism on local morphine antinociception in this type of stimulus. However, it impaired the responses to heat stimuli and the effect of local morphine on heat nociception. In conclusion, peripheral opioid antinociception to mechanical stimuli is limited by sigma-1 tonic inhibitory actions, whereas peripheral opioid antinociception to heat stimuli (produced in TRPV1-expressing neurons) is not. Therefore, sigma-1 receptors contribute to the modality-specific peripheral effects of opioid analgesics.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/pathology
- Hot Temperature
- Hyperalgesia/drug therapy
- Hyperalgesia/metabolism
- Hyperalgesia/pathology
- Mice, Knockout
- Nociceptors/drug effects
- Nociceptors/metabolism
- Nociceptors/pathology
- Random Allocation
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Receptors, sigma/agonists
- Receptors, sigma/antagonists & inhibitors
- Receptors, sigma/genetics
- Receptors, sigma/metabolism
- TRPV Cation Channels/metabolism
- Touch
- Sigma-1 Receptor
Collapse
Affiliation(s)
- Ángeles Montilla-García
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Gloria Perazzoli
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Miguel Á Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Rafael González-Cano
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Cristina Sánchez-Fernández
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain; Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain; Teófilo Hernando Institute for Drug Discovery, 28029 Madrid, Spain.
| | - José M Baeyens
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain; Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain.
| |
Collapse
|
21
|
Castany S, Gris G, Vela JM, Verdú E, Boadas-Vaello P. Critical role of sigma-1 receptors in central neuropathic pain-related behaviours after mild spinal cord injury in mice. Sci Rep 2018; 8:3873. [PMID: 29497125 PMCID: PMC5832850 DOI: 10.1038/s41598-018-22217-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/19/2018] [Indexed: 02/06/2023] Open
Abstract
Sigma-1 receptor (σ1R) knockout (KO) CD1 mice, generated by homologous recombination, and separate pharmacological studies in wild type (WT) mice were done to investigate the role of this receptor in the development of pain-related behaviours (thermal hyperalgesia and mechanical allodynia) in mice after spinal cord contusion injury (SCI) - a model of central neuropathic pain. The modulatory effect of σ1R KO on extracellular mediators and signalling pathways in the spinal cord was also investigated. In particular, changes in the expression of inflammatory cytokines (tumour necrosis factor TNF-α, interleukin IL-1β) and both the expression and activation (phosphorylation) of the N-methyl-D-aspartate receptor subunit 2B (NR2B-NMDA) and extracellular signal-regulated kinases (ERK1/2) were analysed. Compared with WT mice, both mechanical and thermal hypersensitivity were attenuated in σ1R KO mice following SCI. Accordingly, treatment of WT mice with the σ1R antagonist MR309 (previously developed as E-52862; S1RA) after SCI exerted antinociceptive effects (i.e. reduced mechanical allodynia and thermal hyperalgesia). Attenuated nociceptive responses in σ1R KO were accompanied by reduced expression of TNF- α and IL-1β as well as decreased activation/phosphorylation of NR2B-NMDA receptors and ERK1/2. These findings suggest that σ1R may modulate central neuropathic pain and point to regulation of sensitization-related phenomena as a possible mechanism.
Collapse
Affiliation(s)
- Sílvia Castany
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, Universitat de Girona (UdG), Girona, Spain
- ESTEVE, Drug Discovery and Preclinical Development, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
| | - Georgia Gris
- ESTEVE, Drug Discovery and Preclinical Development, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
| | - José Miguel Vela
- ESTEVE, Drug Discovery and Preclinical Development, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
| | - Enrique Verdú
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, Universitat de Girona (UdG), Girona, Spain.
| | - Pere Boadas-Vaello
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, Universitat de Girona (UdG), Girona, Spain.
| |
Collapse
|
22
|
Endocannabinoid control of glutamate NMDA receptors: the therapeutic potential and consequences of dysfunction. Oncotarget 2018; 7:55840-55862. [PMID: 27323834 PMCID: PMC5342457 DOI: 10.18632/oncotarget.10095] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 06/06/2016] [Indexed: 01/04/2023] Open
Abstract
Glutamate is probably the most important excitatory neurotransmitter in the brain. The glutamate N-methyl-D-aspartate receptor (NMDAR) is a calcium-gated channel that coordinates with G protein-coupled receptors (GPCRs) to establish the efficiency of the synaptic transmission. Cross-regulation between these receptors requires the concerted activity of the histidine triad nucleotide-binding protein 1 (HINT1) and of the sigma receptor type 1 (σ1R). Essential brain functions like learning, memory formation and consolidation, mood and behavioral responses to exogenous stimuli depend on the activity of NMDARs. In this biological context, endocannabinoids are released to retain NMDAR activity within physiological limits. The efficacy of such control depends on HINT1/σ1R assisting in the physical coupling between cannabinoid type 1 receptors (CB1Rs) and NMDARs to dampen their activity. Subsequently, the calcium-regulated HINT1/σ1R protein tandem uncouples CB1Rs to prevent NMDAR hypofunction. Thus, early recruitment or a disproportionate cannabinoid induced response can bring about excess dampening of NMDAR activity, impeding its adequate integration with GPCR signaling. Alternatively, this control circuit can apparently be overridden in situations where bursts of NMDAR overactivity provoke convulsive syndromes. In this review we will discuss the possible relevance of the HINT1/σ1R tandem and its use by endocannabinoids to diminish NMDAR activity and their implications in psychosis/schizophrenia, as well as in NMDAR-mediated convulsive episodes.
Collapse
|
23
|
Loiseau C, Cayetanot F, Joubert F, Perrin-Terrin AS, Cardot P, Fiamma MN, Frugiere A, Straus C, Bodineau L. Current Perspectives for the use of Gonane Progesteronergic Drugs in the Treatment of Central Hypoventilation Syndromes. Curr Neuropharmacol 2018; 16:1433-1454. [PMID: 28721821 PMCID: PMC6295933 DOI: 10.2174/1570159x15666170719104605] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/30/2017] [Accepted: 07/12/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Central alveolar hypoventilation syndromes (CHS) encompass neurorespiratory diseases resulting from congenital or acquired neurological disorders. Hypercapnia, acidosis, and hypoxemia resulting from CHS negatively affect physiological functions and can be lifethreatening. To date, the absence of pharmacological treatment implies that the patients must receive assisted ventilation throughout their lives. OBJECTIVE To highlight the relevance of determining conditions in which using gonane synthetic progestins could be of potential clinical interest for the treatment of CHS. METHODS The mechanisms by which gonanes modulate the respiratory drive were put into the context of those established for natural progesterone and other synthetic progestins. RESULTS The clinical benefits of synthetic progestins to treat respiratory diseases are mixed with either positive outcomes or no improvement. A benefit for CHS patients has only recently been proposed. We incidentally observed restoration of CO2 chemosensitivity, the functional deficit of this disease, in two adult CHS women by desogestrel, a gonane progestin, used for contraception. This effect was not observed by another group, studying a single patient. These contradictory findings are probably due to the complex nature of the action of desogestrel on breathing and led us to carry out mechanistic studies in rodents. Our results show that desogestrel influences the respiratory command by modulating the GABAA and NMDA signaling in the respiratory network, medullary serotoninergic systems, and supramedullary areas. CONCLUSION Gonanes show promise for improving ventilation of CHS patients, although the conditions of their use need to be better understood.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Laurence Bodineau
- Address correspondence to this author at the Sorbonne Universités, UPMC Univ. Paris 06, INSERM, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, F-75013, Paris, France; Tel: 33 1 40 77 97 15; Fax: 33 1 40 77 97 89; E-mail:
| |
Collapse
|
24
|
Di T, Zhang S, Hong J, Zhang T, Chen L. Hyperactivity of Hypothalamic-Pituitary-Adrenal Axis Due to Dysfunction of the Hypothalamic Glucocorticoid Receptor in Sigma-1 Receptor Knockout Mice. Front Mol Neurosci 2017; 10:287. [PMID: 28932185 PMCID: PMC5592243 DOI: 10.3389/fnmol.2017.00287] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/24/2017] [Indexed: 01/07/2023] Open
Abstract
Sigma-1 receptor knockout (σ1R-KO) mice exhibit a depressive-like phenotype. Because σ1R is highly expressed in the neuronal cells of hypothalamic paraventricular nuclei (PVN), this study investigated the influence of σ1R deficiency on the regulation of the hypothalamic-pituitary-adrenocortical (HPA) axis. Here, we show that the levels of basal serum corticosterone (CORT), adrenocorticotropic hormone (ACTH) and corticotrophin releasing factor (CRF) as well as the level of CRF mRNA in PVN did not significantly differ between adult male σ1R-KO mice and wild-type (WT) mice. Acute mild restraint stress (AMRS) induced a higher and more sustainable increase in activity of HPA axis and CRF expression in σ1R-KO mice. Percentage of dexamethasone (Dex)-induced reduction in level of CORT was markedly attenuated in σ1R-/- mice. The levels of glucocorticoid receptor (GR) and protein kinase C (PKC) phosphorylation were reduced in the PVN of σ1R-KO mice and σ1R antagonist NE100-treated WT mice. The exposure to AMRS in σ1R-KO mice induced a stronger phosphorylation of cAMP-response element binding protein (CREB) in PVN than that in WT mice. Intracerebroventricular (i.c.v.) injection of PKC activator PMA for 3 days in σ1R-KO mice not only recovered the GR phosphorylation and the percentage of Dex-reduced CORT but also corrected the AMRS-induced hyperactivity of HPA axis and enhancement of CRF mRNA and CREB phosphorylation. Furthermore, the injection (i.c.v.) of PMA in σ1R-KO mice corrected the prolongation of immobility time in forced swim test (FST) and tail suspension test (TST). These results indicate that σ1R deficiency causes down-regulation of GR by reducing PKC phosphorylation, which attenuates GR-mediated feedback inhibition of HPA axis and facilitates the stress response of HPA axis leading to the production of depressive-like behaviors.
Collapse
Affiliation(s)
- Tingting Di
- State Key Laboratory of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Suyun Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical UniversityNanjing, China
| | - Juan Hong
- State Key Laboratory of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Tingting Zhang
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Ling Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| |
Collapse
|
25
|
Georgiadis MO, Karoutzou O, Foscolos AS, Papanastasiou I. Sigma Receptor (σR) Ligands with Antiproliferative and Anticancer Activity. Molecules 2017; 22:E1408. [PMID: 28841173 PMCID: PMC6151391 DOI: 10.3390/molecules22091408] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 01/01/2023] Open
Abstract
Sigma receptor (σR) ligands have proven to be useful as cancer diagnostics and anticancer therapeutics and their ligands have been developed as molecular probes in oncology. Moreover, various σR ligands generate cancer cell death in vitro and in vivo. These σR ligands have exhibited promising results against numerous human and rodent cancers and are investigated under preclinical and clinical study trials, indicating a new category of drugs in cancer therapy.
Collapse
Affiliation(s)
- Markos-Orestis Georgiadis
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 15784 Athens, Greece.
| | - Olga Karoutzou
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 15784 Athens, Greece.
| | - Angeliki-Sofia Foscolos
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 15784 Athens, Greece.
| | - Ioannis Papanastasiou
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 15784 Athens, Greece.
| |
Collapse
|
26
|
Albayrak Y, Hashimoto K. Sigma-1 Receptor Agonists and Their Clinical Implications in Neuropsychiatric Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:153-161. [PMID: 28315270 DOI: 10.1007/978-3-319-50174-1_11] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Accumulating evidence suggests that sigma-1 receptors play a role in the pathophysiology of neuropsychiatric diseases, as well as in the mechanisms of some selective serotonin reuptake inhibitors (SSRIs). Among the SSRIs, the order of affinity for sigma-1 receptors is as follows: fluvoxamine > sertraline > fluoxetine > escitalopram > citalopram >> paroxetine. Some SSRIs (e.g., fluvoxamine, fluoxetine and escitalopram) and other drugs (donepezil , ifenprodil , dehydroepiandeterone (DHEA)) potentiate nerve-growth factor (NGF)-induced neurite outgrowth in PC12 cells, and these effects could be antagonized by the selective sigma-1 receptor antagonist NE-100. Furthermore, fluvoxamine, donepezil, and DHEA, but not paroxetine or sertraline, improved phencyclidine-induced cognitive deficits in mice, and these effects could be antagonized by NE-100. Several clinical studies showed that sigma-1 receptor agonists such as fluvoxamine and ifenprodil could have beneficial effects in patients with neuropsychiatric disorders. In this chapter, the authors will discuss the role of sigma-1 receptors in the mechanistic action of some SSRIs, donepezil, neurosteroids, and ifenprodil, and the clinical implications for sigma-1 receptor agonists .
Collapse
|
27
|
Moradpour F, Fathollahi Y, Naghdi N, Hosseinmardi N, Javan M. Prepubertal castration-associated developmental changes in sigma-1 receptor gene expression levels regulate hippocampus area CA1 activity during adolescence. Hippocampus 2016; 26:933-46. [PMID: 26860755 DOI: 10.1002/hipo.22576] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2016] [Indexed: 11/08/2022]
Abstract
The functional relevance of sigma-1 (σ1 ) receptor expression in the rat hippocampal CA1 during adolescence (i.e., 35-60 days old) was explored. A selective antagonist for the σ1 receptor subtype, BD-1047, was applied to study hippocampal long-term potentiation (LTP) and spatial learning performance. Changes in the expression of the σ1 receptor subtype and its function were compared between castrated and sham-castrated rats. Castration reduced the magnitude of both field excitatory postsynaptic potential (fEPSP)-LTP and population spike (PS)-LTP at 35 days (d). BD-1047 decreased PS-LTP in sham-castrated rats, whereas BD-1047 reversed the effect of castration on fEPSP-LTP at 35 d. In addition, BD1047 impaired spatial learning and augmented σ1 receptor mRNA levels in castrated rats at 35 d. Surprisingly, neither castration nor BD1047 had an effect on fEPSP-LTP and PS-LTP, spatial learning ability or gene expression levels at 45 d. Castration had no effect on fEPSP-LTP but reduced PS-LTP at 60 d. BD1047 increased the magnitude of fEPSP-LTP, but had no effect on PS-LTP in castrated rats at 60 d. However, BD1047 reduced spatial learning ability, and σ1 receptor mRNA levels were decreased in castrated rats at 60 d. This study shows that σ1 receptors play a role in the regulation of both CA1 synaptic efficacy and spatial learning performance. The regulatory role of σ1 receptors in activity-dependent CA1-LTP is locality- and age-dependent, whereas its role in spatial learning ability is only age-dependent. Prepubertal castration-associated changes in the expression and function of the σ1 receptor during adolescence may play a developmental role in the regulation of hippocampal area CA1 activity and plasticity. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Farshad Moradpour
- Department of Physiology Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Physiology School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yaghoub Fathollahi
- Department of Physiology Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nasser Naghdi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Nargess Hosseinmardi
- Department of Physiology School of Medicine, Shahid Behsheti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
28
|
Kwon SG, Roh DH, Yoon SY, Choi SR, Choi HS, Moon JY, Kang SY, Kim HW, Han HJ, Beitz AJ, Oh SB, Lee JH. Role of peripheral sigma-1 receptors in ischaemic pain: Potential interactions with ASIC and P2X receptors. Eur J Pain 2016; 20:594-606. [PMID: 26358747 DOI: 10.1002/ejp.774] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND The role of peripheral sigma-1 receptors (Sig-1Rs) in normal nociception and in pathologically induced pain conditions has not been thoroughly investigated. Since there is mounting evidence that Sig-1Rs modulate ischaemia-induced pathological conditions, we investigated the role of Sig-1Rs in ischaemia-induced mechanical allodynia (MA) and addressed their possible interaction with acid-sensing ion channels (ASICs) and P2X receptors at the ischaemic site. METHODS We used a rodent model of hindlimb thrombus-induced ischaemic pain (TIIP) to investigate their role. Western blot was performed to observe changes in Sig-1R expression in peripheral nervous tissues. MA was measured after intraplantar (i.pl.) injections of antagonists for the Sig-1, ASIC and P2X receptors in TIIP rats or agonists of each receptor in naïve rats. RESULTS Sig-1R expression significantly increased in skin, sciatic nerve and dorsal root ganglia at 3 days post-TIIP surgery. I.pl. injections of the Sig-1R antagonist, BD-1047 on post-operative days 0-3 significantly attenuated the development of MA during the induction phase, but had no effect on MA when given during the maintenance phase (days 3-6 post-surgery). BD-1047 synergistically increased amiloride (an ASICs blocker)- and TNP-ATP (a P2X antagonist)-induced analgesic effects in TIIP rats. In naïve rats, i.pl. injection of Sig-1R agonist PRE-084 alone did not produce MA; but it did induce MA when co-administered with either an acidic pH solution or a sub-effective dose of αβmeATP. CONCLUSION Peripheral Sig-1Rs contribute to the induction of ischaemia-induced MA via facilitation of ASICs and P2X receptors. Thus, peripheral Sig-1Rs represent a novel therapeutic target for the treatment of ischaemic pain.
Collapse
Affiliation(s)
- S G Kwon
- Department of Veterinary Physiology, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Korea
| | - D H Roh
- Department of Maxillofacial Tissue Regeneration, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - S Y Yoon
- Pain Cognitive Function Research Center, Department of Brain and Cognitive Sciences College of Natural Sciences, Seoul National University, Korea
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Korea
| | - S R Choi
- Department of Veterinary Physiology, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Korea
| | - H S Choi
- Department of Veterinary Physiology, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Korea
| | - J Y Moon
- KM Fundamental Research Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - S Y Kang
- KM Fundamental Research Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - H W Kim
- Department of Physiology, Institute of Brain Research, Chungnam National University Medical School, Daejeon, Korea
| | - H J Han
- Department of Veterinary Physiology, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Korea
| | - A J Beitz
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, USA
| | - S B Oh
- Pain Cognitive Function Research Center, Department of Brain and Cognitive Sciences College of Natural Sciences, Seoul National University, Korea
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Korea
| | - J H Lee
- Department of Veterinary Physiology, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Korea
| |
Collapse
|
29
|
Romero L, Merlos M, Vela JM. Antinociception by Sigma-1 Receptor Antagonists: Central and Peripheral Effects. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 75:179-215. [PMID: 26920013 DOI: 10.1016/bs.apha.2015.11.003] [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] [Indexed: 12/15/2022]
Abstract
There is plenty of evidence supporting the modulatory role of sigma-1 receptors (σ1Rs) in nociception, mainly based on the pain-attenuated phenotype of σ1R knockout mice and on the antinociceptive effect exerted by σ1R antagonists, particularly in nonacute sensitizing conditions involving sustained afferent drive, activity-dependent plasticity/sensitization, and ultimately pain hypersensitivity, as it is the case in chronic pains of different etiology. Antinociceptive effects of σ1R antagonists both when acting alone and in combination with opioids (to enhance opioid analgesia) have been reported at both central and peripheral sites. At the central level, findings at the behavioral (animal pain models), electrophysiological (spinal wind-up recordings), neurochemical (spinal release of neurotransmitters) and molecular (NMDAR function) level supports a role for σ1R antagonists in inhibiting augmented excitability secondary to sustained afferent input. Attenuation of activity-induced plastic changes (central sensitization) following tissue injury/inflammation or nerve damage could thus underlie the central inhibitory effect of σ1R antagonists. Moreover, recent pieces of information confirm the involvement of σ1R in mechanisms regulating pain at the periphery, where σ1Rs are highly expressed, particularly in dorsal root ganglia. Indeed, local peripheral administration of σ1R antagonists reduces inflammatory hyperalgesia. Potentiation of opioid analgesia is also supported, particularly at supraspinal sites and at the periphery, where locally administered σ1R antagonists unmask opioid analgesia. Altogether, whereas σ1R activation is coupled to pain facilitation and inhibition of opioid antinociception, σ1R antagonism inhibits pain hypersensitivity and "releases the brake" enabling opioids to exert enhanced antinociceptive effects, both at the central nervous system and at the periphery.
Collapse
Affiliation(s)
- Luz Romero
- Drug Discovery and Preclinical Development, ESTEVE, Parc Científic de Barcelona, Baldiri Reixac 4-8, Barcelona, Spain
| | - Manuel Merlos
- Drug Discovery and Preclinical Development, ESTEVE, Parc Científic de Barcelona, Baldiri Reixac 4-8, Barcelona, Spain
| | - José Miguel Vela
- Drug Discovery and Preclinical Development, ESTEVE, Parc Científic de Barcelona, Baldiri Reixac 4-8, Barcelona, Spain.
| |
Collapse
|
30
|
Abstract
This review compares the biological and physiological function of Sigma receptors [σRs] and their potential therapeutic roles. Sigma receptors are widespread in the central nervous system and across multiple peripheral tissues. σRs consist of sigma receptor one (σ1R) and sigma receptor two (σ2R) and are expressed in numerous regions of the brain. The sigma receptor was originally proposed as a subtype of opioid receptors and was suggested to contribute to the delusions and psychoses induced by benzomorphans such as SKF-10047 and pentazocine. Later studies confirmed that σRs are non-opioid receptors (not an µ opioid receptor) and play a more diverse role in intracellular signaling, apoptosis and metabolic regulation. σ1Rs are intracellular receptors acting as chaperone proteins that modulate Ca2+ signaling through the IP3 receptor. They dynamically translocate inside cells, hence are transmembrane proteins. The σ1R receptor, at the mitochondrial-associated endoplasmic reticulum membrane, is responsible for mitochondrial metabolic regulation and promotes mitochondrial energy depletion and apoptosis. Studies have demonstrated that they play a role as a modulator of ion channels (K+ channels; N-methyl-d-aspartate receptors [NMDAR]; inositol 1,3,5 triphosphate receptors) and regulate lipid transport and metabolism, neuritogenesis, cellular differentiation and myelination in the brain. σ1R modulation of Ca2+ release, modulation of cardiac myocyte contractility and may have links to G-proteins. It has been proposed that σ1Rs are intracellular signal transduction amplifiers. This review of the literature examines the mechanism of action of the σRs, their interaction with neurotransmitters, pharmacology, location and adverse effects mediated through them.
Collapse
Affiliation(s)
- Colin G Rousseaux
- a Department of Pathology and Laboratory Medicine , University of Ottawa , Ottawa , ON , Canada and
| | | |
Collapse
|
31
|
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
|
32
|
Gris G, Cobos EJ, Zamanillo D, Portillo-Salido E. Sigma-1 receptor and inflammatory pain. Inflamm Res 2015; 64:377-81. [PMID: 25902777 DOI: 10.1007/s00011-015-0819-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/23/2015] [Accepted: 04/08/2015] [Indexed: 01/28/2023] Open
Abstract
INTRODUCTION The sigma-1 receptor (Sig-1R) is a unique ligand-regulated molecular chaperone that interacts with several protein targets such as G protein-coupled receptors and ion channels to modulate their activity. Sig-1R is located in areas of the central and peripheral nervous system that are key to pain control. Previous preclinical studies have suggested a potential therapeutic use of Sig-1R antagonists for the management of neuropathic pain. DISCUSSION Recent studies using pharmacological and genetic tools have explored the role of Sig-1R in inflammatory pain conditions. Mice lacking the Sig-1R have shown different patterns of phenotypic responses to inflammatory injury. Systemic or peripheral administration of several Sig-1R antagonists, including the selective Sig-1R antagonist S1RA, inhibited both mechanical and thermal hypersensitivity in several preclinical models of inflammatory pain. These recent studies are summarized in the present commentary. CONCLUSION Central and peripheral pharmacological blockade of Sig-1R could be an effective option to treat inflammatory pain.
Collapse
Affiliation(s)
- Georgia Gris
- Drug Discovery and Preclinical Development, ESTEVE, Baldiri Reixach, 4-8, 08028, Barcelona, Spain
| | | | | | | |
Collapse
|
33
|
Malik M, Rangel-Barajas C, Sumien N, Su C, Singh M, Chen Z, Huang RQ, Meunier J, Maurice T, Mach RH, Luedtke RR. The effects of sigma (σ1) receptor-selective ligands on muscarinic receptor antagonist-induced cognitive deficits in mice. Br J Pharmacol 2015; 172:2519-31. [PMID: 25573298 PMCID: PMC4409904 DOI: 10.1111/bph.13076] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 12/12/2014] [Accepted: 12/29/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Cognitive deficits in patients with Alzheimer's disease, Parkinson's disease, traumatic brain injury and stroke often involve alterations in cholinergic signalling. Currently available therapeutic drugs provide only symptomatic relief. Therefore, novel therapeutic strategies are needed to retard and/or arrest the progressive loss of memory. EXPERIMENTAL APPROACH Scopolamine-induced memory impairment provides a rapid and reversible phenotypic screening paradigm for cognition enhancement drug discovery. Male C57BL/6J mice given scopolamine (1 mg·kg(-1) ) were used to evaluate the ability of LS-1-137, a novel sigma (σ1) receptor-selective agonist, to improve the cognitive deficits associated with muscarinic antagonist administration. KEY RESULTS LS-1-137 is a high-affinity (Ki = 3.2 nM) σ1 receptor agonist that is 80-fold selective for σ1, compared with σ2 receptors. LS-1-137 binds with low affinity at D2-like (D2, D3 and D4) dopamine and muscarinic receptors. LS-1-137 was found to partially reverse the learning deficits associated with scopolamine administration using a water maze test and an active avoidance task. LS-1-137 treatment was also found to trigger the release of brain-derived neurotrophic factor from rat astrocytes. CONCLUSIONS AND IMPLICATIONS The σ1 receptor-selective compound LS-1-137 may represent a novel candidate cognitive enhancer for the treatment of muscarinic receptor-dependent cognitive deficits.
Collapse
Affiliation(s)
- Maninder Malik
- Department of Pharmacology and Neuroscience, University of North Texas Health Science CenterFort Worth, TX, USA
| | - Claudia Rangel-Barajas
- Department of Pharmacology and Neuroscience, University of North Texas Health Science CenterFort Worth, TX, USA
| | - Nathalie Sumien
- Department of Pharmacology and Neuroscience, University of North Texas Health Science CenterFort Worth, TX, USA
| | - Chang Su
- Department of Pharmacology and Neuroscience, University of North Texas Health Science CenterFort Worth, TX, USA
| | - Meharvan Singh
- Department of Pharmacology and Neuroscience, University of North Texas Health Science CenterFort Worth, TX, USA
| | - Zhenglan Chen
- Department of Pharmacology and Neuroscience, University of North Texas Health Science CenterFort Worth, TX, USA
| | - Ren-Qi Huang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science CenterFort Worth, TX, USA
| | - Johann Meunier
- AMYLGENMontferrier-surLez, France
- 3 INSERM U. 710Montpellier, France
| | - Tangui Maurice
- AMYLGENMontferrier-surLez, France
- 3 INSERM U. 710Montpellier, France
- University of MontpellierMontpellier, France
| | - Robert H Mach
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA, USA
| | - Robert R Luedtke
- Department of Pharmacology and Neuroscience, University of North Texas Health Science CenterFort Worth, TX, USA
| |
Collapse
|
34
|
Rodríguez-Muñoz M, Sánchez-Blázquez P, Herrero-Labrador R, Martínez-Murillo R, Merlos M, Vela JM, Garzón J. The σ1 receptor engages the redox-regulated HINT1 protein to bring opioid analgesia under NMDA receptor negative control. Antioxid Redox Signal 2015; 22:799-818. [PMID: 25557043 PMCID: PMC4367239 DOI: 10.1089/ars.2014.5993] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 12/16/2014] [Accepted: 01/01/2015] [Indexed: 12/12/2022]
Abstract
AIMS The in vivo pharmacology of the sigma 1 receptor (σ1R) is certainly complex; however, σ1R antagonists are of therapeutic interest, because they enhance mu-opioid receptor (MOR)-mediated antinociception and reduce neuropathic pain. Thus, we investigated whether the σ1R is involved in the negative control that glutamate N-methyl-d-aspartate acid receptors (NMDARs) exert on opioid antinociception. RESULTS The MOR C terminus carries the histidine triad nucleotide-binding protein 1 (HINT1) coupled to the regulator of G-protein signaling RGSZ2-neural nitric oxide synthase assembly. Activated MORs stimulate the production of nitric oxide (NO), and the redox zinc switch RGSZ2 converts this signal into free zinc ions that are required to recruit the redox sensor PKCγ to HINT1 proteins. Then, PKCγ impairs HINT1-RGSZ2 association and enables σ1R-NR1 interaction with MOR-HINT1 complexes to restrain opioid signaling. The inhibition of NOS or the absence of σ1Rs prevents HINT1-PKCγ interaction, and MOR-NMDAR cross-regulation fails. The σ1R antagonists transitorily remove the binding of σ1Rs to NR1 subunits, facilitate the entrance of negative regulators of NMDARs, likely Ca(2+)-CaM, and prevent NR1 interaction with HINT1, thereby impairing the negative feedback of glutamate on opioid analgesia. INNOVATION A redox-regulated process situates MOR signaling under NMDAR control, and in this context, the σ1R binds to the cytosolic C terminal region of the NMDAR NR1 subunit. CONCLUSION The σ1R antagonists enhance opioid analgesia in naïve mice by releasing MORs from the negative influence of NMDARs, and they also reset antinociception in morphine tolerant animals. Moreover, σ1R antagonists alleviate neuropathic pain, probably by driving the inhibition of up-regulated NMDARs.
Collapse
Affiliation(s)
- María Rodríguez-Muñoz
- Neurofarmacología, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Pilar Sánchez-Blázquez
- Neurofarmacología, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Raquel Herrero-Labrador
- Neurofarmacología, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Ricardo Martínez-Murillo
- Neurofarmacología, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Manuel Merlos
- Drug Discovery & Preclinical Development, Esteve, Barcelona, Spain
| | - José Miguel Vela
- Drug Discovery & Preclinical Development, Esteve, Barcelona, Spain
| | - Javier Garzón
- Neurofarmacología, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
35
|
Jaramillo-Loranca BE, Garcés-Ramírez L, Munguía Rosales AA, Luna Ramírez C, Vargas Hernández G, Morales-Dionisio O, González-Elizalde K, Flores G, Zamudio S, De La Cruz-López F. The sigma agonist 1,3-Di-o-tolyl-guanidine reduces the morphological and behavioral changes induced by neonatal ventral hippocampus lesion in rats. Synapse 2015; 69:213-25. [DOI: 10.1002/syn.21811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/11/2014] [Accepted: 02/06/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Blanca Estela Jaramillo-Loranca
- Departamento De Fisiología, Escuela Nacional De Ciencias Biológicas; Instituto Politécnico Nacional; México D. F. México
- Programa Educativo De La Licenciatura En Terapia Física, Universidad Politécnica De Pachuca; Zempoala Hidalgo México
| | - Linda Garcés-Ramírez
- Departamento De Fisiología, Escuela Nacional De Ciencias Biológicas; Instituto Politécnico Nacional; México D. F. México
| | | | - Carolina Luna Ramírez
- Programa Educativo De La Ingeniería En Biotecnología, Universidad Politécnica De Pachuca; Zempoala Hidalgo México
| | - Genaro Vargas Hernández
- Programa Educativo De La Ingeniería En Biotecnología, Universidad Politécnica De Pachuca; Zempoala Hidalgo México
| | - Oscar Morales-Dionisio
- Departamento De Fisiología, Escuela Nacional De Ciencias Biológicas; Instituto Politécnico Nacional; México D. F. México
| | - Kateri González-Elizalde
- Departamento De Fisiología, Escuela Nacional De Ciencias Biológicas; Instituto Politécnico Nacional; México D. F. México
| | - Gonzalo Flores
- Laboratorio De Neuropsiquiatría, Instituto De Fisiología, Universidad Autónoma De Puebla; Puebla México
| | - Sergio Zamudio
- Departamento De Fisiología, Escuela Nacional De Ciencias Biológicas; Instituto Politécnico Nacional; México D. F. México
| | - Fidel De La Cruz-López
- Departamento De Fisiología, Escuela Nacional De Ciencias Biológicas; Instituto Politécnico Nacional; México D. F. México
| |
Collapse
|
36
|
Tejada MA, Montilla-García A, Sánchez-Fernández C, Entrena JM, Perazzoli G, Baeyens JM, Cobos EJ. Sigma-1 receptor inhibition reverses acute inflammatory hyperalgesia in mice: role of peripheral sigma-1 receptors. Psychopharmacology (Berl) 2014; 231:3855-69. [PMID: 24639046 DOI: 10.1007/s00213-014-3524-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 02/26/2014] [Indexed: 11/29/2022]
Abstract
RATIONALE Sigma-1 (σ1) receptor inhibition ameliorates neuropathic pain by inhibiting central sensitization. However, it is unknown whether σ1 receptor inhibition also decreases inflammatory hyperalgesia, or whether peripheral σ1 receptors are involved in this process. OBJECTIVE The purpose of this study was to determine the role of σ1 receptors in carrageenan-induced inflammatory hyperalgesia, particularly at the inflammation site. RESULTS The subcutaneous (s.c.) administration of the selective σ1 antagonists BD-1063 and S1RA to wild-type mice dose-dependently and fully reversed inflammatory mechanical (paw pressure) and thermal (radiant heat) hyperalgesia. These antihyperalgesic effects were abolished by the s.c. administration of the σ1 agonist PRE-084 and also by the intraplantar (i.pl.) administration of this compound in the inflamed paw, suggesting that blockade of peripheral σ1 receptors in the inflamed site is involved in the antihyperalgesic effects induced by σ1 antagonists. In fact, the i.pl. administration of σ1 antagonists in the inflamed paw (but not in the contralateral paw) was sufficient to completely reverse inflammatory hyperalgesia. σ1 knockout (σ1-KO) mice did not develop mechanical hyperalgesia but developed thermal hypersensitivity; however, the s.c. administration of BD-1063 or S1RA had no effect on thermal hyperalgesia in σ1-KO mice, supporting on-target mechanisms for the effects of both drugs. The antiedematous effects of σ1 inhibition do not account for the decreased hyperalgesia, since carrageenan-induced edema was unaffected by σ1 knockout or systemic σ1 pharmacological antagonism. CONCLUSIONS σ1 receptors play a major role in inflammatory hyperalgesia. Targeting σ1 receptors in the inflamed tissue may be useful for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- M A Tejada
- Department of Pharmacology and Institute of Neuroscience, Faculty of Medicine, University of Granada, Avenida de Madrid 11, 18012, Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
37
|
Parenti C, Marrazzo A, Aricò G, Parenti R, Pasquinucci L, Ronsisvalle S, Ronsisvalle G, Scoto GM. The antagonistic effect of the sigma 1 receptor ligand (+)-MR200 on persistent pain induced by inflammation. Inflamm Res 2014; 63:231-7. [PMID: 24316864 DOI: 10.1007/s00011-013-0692-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/17/2013] [Accepted: 11/24/2013] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE AND DESIGN The sigma 1 (σ1) receptor, which is widely distributed in the CNS in areas that are known to be important for pain control, may play a role in persistent pain characterized by the hypersensitivity of nociceptive transmission. Here, we investigated the effect of σ1 blockade in an inflammatory pain model. TREATMENT AND METHODS An intraplantar injection of carrageenan (2 %) was used to induce paw inflammation. The effects of the σ1 antagonist (+)-MR200, given subcutaneously at a dose of 0.1, 0.25, 0.5,1, 1.5, and 2 mg/kg prior to injection of carrageenan, on inflammatory pain and inflammation were assessed. Mechanical allodynia with von Frey filaments, thermal hyperalgesia with the plantar test and edema evaluation with a plethysmometer were measured. Intergroup comparisons were assessed by one- or two-way analysis of variance when appropriate, followed by post-hoc tests (Dunnett's test for one-way or Bonferroni for two-way ANOVA). RESULTS (+)-MR200 dose-dependently prevented allodynia and hyperalgesia induced by carrageenan. Furthermore, it reduced paw edema with a significant inhibition percentage of 37.82 % at 3 h after carrageenan treatment. CONCLUSIONS The blockade of the σ1 receptor with the selective antagonist (+)-MR200 may contribute to the suppression of the typical symptoms of inflammatory pain.
Collapse
Affiliation(s)
- Carmela Parenti
- Pharmacology and Toxicology Section, Department of Drug Sciences, University of Catania, 95125, Catania, Italy
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Nieto FR, Cendán CM, Cañizares FJ, Cubero MA, Vela JM, Fernández-Segura E, Baeyens JM. Genetic inactivation and pharmacological blockade of sigma-1 receptors prevent paclitaxel-induced sensory-nerve mitochondrial abnormalities and neuropathic pain in mice. Mol Pain 2014; 10:11. [PMID: 24517272 PMCID: PMC3924235 DOI: 10.1186/1744-8069-10-11] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 02/07/2014] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Paclitaxel, a widely-used antineoplastic drug, produces a painful peripheral neuropathy that in rodents is associated with peripheral-nerve mitochondrial alterations. The sigma-1 receptor (σ1R) is a ligand-regulated molecular chaperone involved in mitochondrial calcium homeostasis and pain hypersensitivity. This receptor plays a key role in paclitaxel-induced neuropathic pain, but it is not known whether it also modulates mitochondrial abnormalities.In this study, we used a mouse model of paclitaxel-induced neuropathic pain to test the involvement of the σ1R in the mitochondrial abnormalities associated with paclitaxel, by using genetic (σ1R knockout mice) and pharmacological (σ1R antagonist) approaches. RESULTS Paclitaxel administration to wild-type (WT) mice produced cold- and mechanical-allodynia, and an increase in the frequency of swollen and vacuolated mitochondria in myelinated A-fibers, but not in C-fibers, of the saphenous nerve. Behavioral and mitochondrial alterations were marked at 10 days after paclitaxel-administration and had resolved at day 28. In contrast, paclitaxel treatment did not induce allodynia or mitochondrial abnormalities in σ1R knockout mice. Moreover, the prophylactic treatment of WT mice with BD-1063 also prevented the neuropathic pain and mitochondrial abnormalities induced by paclitaxel. CONCLUSIONS These results suggest that activation of the σ1R is necessary for development of the sensory nerve mitochondrial damage and neuropathic pain produced by paclitaxel. Therefore, σ1R antagonists might have therapeutic value for the prevention of paclitaxel-induced neuropathy.
Collapse
Affiliation(s)
- Francisco R Nieto
- Department of Pharmacology, Biomedical Research Centre and Institute of Neuroscience, University of Granada, 18012 Granada, Spain
- Current address: Wolfson Centre for Age-Related Diseases, King’s College London, Wolfson Wing, Hodgkin Building, SE1 1UL London, UK
| | - Cruz M Cendán
- Department of Pharmacology, Biomedical Research Centre and Institute of Neuroscience, University of Granada, 18012 Granada, Spain
| | - Francisco J Cañizares
- Department of Histology, Biomedical Research Centre and Institute of Neuroscience, University of Granada, 18012 Granada, Spain
| | - María A Cubero
- Department of Histology, Biomedical Research Centre and Institute of Neuroscience, University of Granada, 18012 Granada, Spain
| | - José M Vela
- Esteve, Drug Discovery and Preclinical Development, Parc Científic de Barcelona, Carrer Baldiri Reixac, 4-8, 08028 Barcelona, Spain
| | - Eduardo Fernández-Segura
- Department of Histology, Biomedical Research Centre and Institute of Neuroscience, University of Granada, 18012 Granada, Spain
| | - José M Baeyens
- Department of Pharmacology, Biomedical Research Centre and Institute of Neuroscience, University of Granada, 18012 Granada, Spain
| |
Collapse
|
39
|
Maisonial-Besset A, Funke U, Wenzel B, Fischer S, Holl K, Wünsch B, Steinbach J, Brust P. Automation of the radiosynthesis and purification procedures for [18F]Fluspidine preparation, a new radiotracer for clinical investigations in PET imaging of σ1 receptors in brain. Appl Radiat Isot 2014; 84:1-7. [DOI: 10.1016/j.apradiso.2013.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 10/05/2013] [Accepted: 10/26/2013] [Indexed: 10/26/2022]
|
40
|
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: 3.9] [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
|
41
|
Jin H, Fan J, Zhang X, Li J, Flores HP, Perlmutter JS, Parsons SM, Tu Z. Radiosynthesis and in vivo evaluation of a novel σ 1 selective PET ligand. MEDCHEMCOMM 2014; 5:1669-1677. [PMID: 25584182 DOI: 10.1039/c4md00240g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The σ1 receptor is an important target for CNS disorders. We previously identified a σ1 ligand TZ3108 having highly potent (Ki-σ1 = 0.48 nM) and selective affinity for σ1 versus σ2 receptors. TZ3108 was 18F-labeled with F-18 for in vivo evaluation. Biodistribution and blocking studies of [18F]TZ3108 in male Sprague-Dawley rats demonstrated high brain uptake, which was σ1-specific with no in vivo defluorination. MicroPET studies in cynomolgus macaques showed high brain penetration of [18F]TZ3108; the regional brain distribution was consistent with that of the σ1 receptor. Pseudo-equilibrium in the brain was reached ~ 45 min post-injection. Metabolite analysis of [18F]TZ3108 in NHP blood and rodent blood and brain revealed that ~ 70% parent remained in the plasma of NHPs 60 min post-injection and the major radiometabolite did not cross the blood-brain barrier in rats. In summary, the potent, selective and metabolically stable σ1 specific radioligand [18F]TZ3108 represents a potentially useful PET radioligand for quantifying the σ1 receptor in the brain.
Collapse
Affiliation(s)
- Hongjun Jin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jinda Fan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xiang Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Junfeng Li
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hubert P Flores
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA. ; Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Stanley M Parsons
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, 93106, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
42
|
Zamanillo D, Romero L, Merlos M, Vela JM. Sigma 1 receptor: a new therapeutic target for pain. Eur J Pharmacol 2013; 716:78-93. [PMID: 23500210 DOI: 10.1016/j.ejphar.2013.01.068] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 12/15/2012] [Accepted: 01/09/2013] [Indexed: 01/05/2023]
Abstract
Sigma 1 receptor (σ₁ receptor) is a unique ligand-regulated molecular chaperone located mainly in the endoplasmic reticulum and the plasma membrane. σ₁ receptor is activated under stress or pathological conditions and interacts with several neurotransmitter receptors and ion channels to modulate their function. The effects reported preclinically with σ₁ receptor ligands are consistent with a role for σ₁ receptor in central sensitization and pain hypersensitivity and suggest a potential therapeutic use of σ₁ receptor antagonists for the management of neuropathic pain as monotherapy. Moreover, data support their use in opioid adjuvant therapy: combination of σ₁ receptor antagonists and opioids results in potentiation of opioid analgesia, without significant increases in opioid-related unwanted effects. Results from clinical trials using selective σ₁ receptor antagonists in several pain conditions are eagerly awaited to ascertain the potential of σ₁ receptor modulation in pain therapy.
Collapse
Affiliation(s)
- Daniel Zamanillo
- Esteve, Drug Discovery and Preclinical Development. Parc Científic de Barcelona. Carrer Baldiri Reixac, 4-8. 08028 Barcelona, Spain
| | | | | | | |
Collapse
|
43
|
Sánchez-Fernández C, Nieto FR, González-Cano R, Artacho-Cordón A, Romero L, Montilla-García Á, Zamanillo D, Baeyens JM, Entrena JM, Cobos EJ. Potentiation of morphine-induced mechanical antinociception by σ₁ receptor inhibition: role of peripheral σ₁ receptors. Neuropharmacology 2013; 70:348-58. [PMID: 23524304 DOI: 10.1016/j.neuropharm.2013.03.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 02/25/2013] [Accepted: 03/04/2013] [Indexed: 11/21/2022]
Abstract
We studied the modulation of morphine-induced mechanical antinociception and side effects by σ₁ receptor inhibition. Both wild-type (WT) and σ₁ receptor knockout (σ₁-KO) mice showed similar responses to paw pressure (100-600 g). The systemic (subcutaneous) or local (intraplantar) administration of σ₁ antagonists (BD-1063, BD-1047, NE-100 and S1RA) was devoid of antinociceptive effects in WT mice. However, σ₁-KO mice exhibited an enhanced mechanical antinociception in response to systemic morphine (1-16 mg/kg). Similarly, systemic treatment of WT mice with σ₁ antagonists markedly potentiated morphine-induced antinociception, and its effects were reversed by the selective σ₁ agonist PRE-084. Although the local administration of morphine (50-200 μg) was devoid of antinociceptive effects in WT mice, it induced dose-dependent antinociception in σ₁-KO mice. This effect was limited to the injected paw. Enhancement of peripheral morphine antinociception was replicated in WT mice locally co-administered with σ₁ antagonists and the opioid. None of the σ₁ antagonists tested enhanced morphine-antinociception in σ₁-KO mice, confirming a σ₁-mediated action. Morphine-induced side-effects (hyperlocomotion and inhibition of gastrointestinal transit) were unaltered in σ₁-KO mice. These results cannot be explained by a direct interaction of σ₁ ligands with μ-opioid receptors or adaptive changes of μ-receptors in σ₁-KO mice, given that [(3)H]DAMGO binding in forebrain, spinal cord, and hind-paw skin membranes was unaltered in mutant mice, and none of the σ₁ drugs tested bound to μ-opioid receptors. These results show that σ₁ receptor inhibition potentiates morphine-induced mechanical analgesia but not its acute side effects, and that this enhanced analgesia can be induced at peripheral level.
Collapse
Affiliation(s)
- Cristina Sánchez-Fernández
- Department of Pharmacology, School of Medicine, University of Granada, Avenida de Madrid 11, 18012 Granada, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
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: 181] [Impact Index Per Article: 15.1] [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
|
45
|
Nieto FR, Cendán CM, Sánchez-Fernández C, Cobos EJ, Entrena JM, Tejada MA, Zamanillo D, Vela JM, Baeyens JM. Role of sigma-1 receptors in paclitaxel-induced neuropathic pain in mice. THE JOURNAL OF PAIN 2012; 13:1107-21. [PMID: 23063344 DOI: 10.1016/j.jpain.2012.08.006] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 08/04/2012] [Accepted: 08/20/2012] [Indexed: 01/22/2023]
Abstract
UNLABELLED Sigma-1 (σ(1)) receptors play a role in different types of pain and in central sensitization mechanisms; however, it is unknown whether they are involved in chemotherapy-induced neuropathic pain. We compared the ability of paclitaxel to induce cold (acetone test) and mechanical (electronic Von Frey test) allodynia in wild-type (WT) and σ(1) receptor knockout (σ(1)-KO) mice. We also tested the effect on paclitaxel-induced painful neuropathy of BD-1063 (16-64 mg/kg, subcutaneously) and S1RA (32-128 mg/kg, subcutaneously), 2 selective σ(1) receptor antagonists that bind to the σ(1) receptor with high affinity and competitively. The responses to cold and mechanical stimuli were similar in WT and σ(1)-KO mice not treated with paclitaxel; however, treatment with paclitaxel (2 mg/kg, intraperitoneally, once per day during 5 consecutive days) produced cold and mechanical allodynia and an increase in spinal cord diphosphorylated extracellular signal-regulated kinase (pERK) in WT but not in σ(1)-KO mice. The administration of BD-1063 or S1RA 30 minutes before each paclitaxel dose prevented the development of cold and mechanical allodynia in WT mice. Moreover, the acute administration of both σ(1) receptor antagonists dose dependently reversed both types of paclitaxel-induced allodynia after they had fully developed. These results suggest that σ(1) receptors play a key role in paclitaxel-induced painful neuropathy. PERSPECTIVE Antagonists of the σ(1) receptor may have therapeutic value for the treatment and/or prevention of paclitaxel-induced neuropathic pain. This possibility is especially interesting in the context of chemotherapy-induced neuropathy, where the onset of nerve damage is predictable and preventive treatment could be administered.
Collapse
Affiliation(s)
- Francisco Rafael Nieto
- Department of Pharmacology and Institute of Neuroscience, Faculty of Medicine, University of Granada, Granada, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Martin-Fardon R, Strong EM, Weiss F. Effect of σ1 receptor antagonism on ethanol and natural reward seeking. Neuroreport 2012; 23:809-13. [DOI: 10.1097/wnr.0b013e32835717c8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
47
|
Microwave assisted synthesis of spirocyclic pyrrolidines – σ1 receptor ligands with modified benzene-N-distance. Eur J Med Chem 2012; 53:327-36. [DOI: 10.1016/j.ejmech.2012.04.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 04/12/2012] [Accepted: 04/13/2012] [Indexed: 11/23/2022]
|
48
|
Maisonial A, Große Maestrup E, Wiese C, Hiller A, Schepmann D, Fischer S, Deuther-Conrad W, Steinbach J, Brust P, Wünsch B. Synthesis, radiofluorination and pharmacological evaluation of a fluoromethyl spirocyclic PET tracer for central σ1 receptors and comparison with fluoroalkyl homologs. Bioorg Med Chem 2012; 20:257-69. [DOI: 10.1016/j.bmc.2011.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 10/31/2011] [Accepted: 11/02/2011] [Indexed: 11/17/2022]
|
49
|
Schläger T, Schepmann D, Lehmkuhl K, Holenz J, Vela JM, Buschmann H, Wünsch B. Combination of two pharmacophoric systems: synthesis and pharmacological evaluation of spirocyclic pyranopyrazoles with high σ₁ receptor affinity. J Med Chem 2011; 54:6704-13. [PMID: 21859078 DOI: 10.1021/jm200585k] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The novel class of spirocyclic σ(1) ligands 3 (6',7'-dihydro-1'H-spiro[piperidine-4,4'-pyrano[4,3-c]pyrazoles]) was designed by the combination of the potent σ(1) ligands 1 and 2 in one molecule. Thorough structure affinity relationships were derived by the variation of the substituents in position 1', 1, and 6'. Whereas the small electron rich methylpyrazole heterocycle was less tolerated by the σ(1) receptor protein, the introduction of a phenyl substituent instead of the methyl group led to ligands with a high σ(1) affinity. It is postulated that the additional phenyl substituent occupies a previously unrecognized hydrophobic region of the σ(1) receptor resulting in additional lipophilic interactions. The spirocyclic pyranopyrazoles are very selective against the σ(2) subtype, the PCP binding site of the NMDA receptor, and further targets. Despite high σ(1) affinity, the cyclohexylmethyl derivative 17i (K(i) (σ(1)) = 0.55 nM) and the isopentenyl derivative 17p (K(i) (σ(1)) = 1.6 nM) showed only low antiallodynic activity in the capsaicin assay.
Collapse
Affiliation(s)
- Torsten Schläger
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Hittorfstrasse 58-62, D-48149 Münster, Germany
| | | | | | | | | | | | | |
Collapse
|
50
|
Wang W, Cui J, Lu X, Padakanti PK, Xu J, Parsons SM, Luedtke RR, Rath NP, Tu Z. Synthesis and in vitro biological evaluation of carbonyl group-containing analogues for σ1 receptors. J Med Chem 2011; 54:5362-72. [PMID: 21732626 DOI: 10.1021/jm200203f] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
To identify the ligands for σ(1) receptors that are potent and selective, analogues of prezamicol and trozamicol scaffolds of carbonyl-containing vesicular acetylcholine transporter (VAChT) inhibitors were explored. Of the 23 analogues synthesized and tested, 5 displayed very high affinity for σ(1) (K(i) = 0.48-4.05 nM) and high selectivity for σ(1) relative to σ(2) receptors (σ(1)/σ(2) selectivity of >749-fold). Four of the five compounds (14a, 14b, 14c, and 14e) showed very low affinity for VAChT (K(i) > 290 nM), and the fifth compound (14g) showed moderate affinity for VAChT (K(i) = 44.2 nM). The compound [1'-(4-fluorobenzyl)-3'-hydroxy[1,4']bipiperidinyl-4-yl]-(4-fluorophenyl)methanone (14a) displayed very high affinity and selectivity for σ(1) receptor (K(i) = 0.48 nM, σ(1)/σ(2) > 3600). All four of these most promising compounds (14a, 14b, 14c, and 14e) can be radiosynthesized with fluorine-18 or carbon-11, which will allow further evaluation of their properties as PET probes for imaging σ(1) receptor in vivo.
Collapse
Affiliation(s)
- Wei Wang
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | | | | | | | | | | | | | | | | |
Collapse
|