1
|
Peng Y, Zhang AH, Wei L, Welsh WJ. Preclinical Evaluation of Sigma 1 Receptor Antagonists as a Novel Treatment for Painful Diabetic Neuropathy. ACS Pharmacol Transl Sci 2024; 7:2358-2368. [PMID: 39144554 PMCID: PMC11320727 DOI: 10.1021/acsptsci.4c00186] [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: 04/01/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024]
Abstract
The global prevalence of diabetes is steadily rising, with an estimated 537 million adults affected by diabetes in 2021, projected to reach 783 million by 2045. A severe consequence of diabetes is the development of painful diabetic neuropathy (PDN), afflicting approximately one in every three diabetic patients and significantly compromising their quality of life. Current pharmacotherapies for PDN provide inadequate pain relief for many patients, underscoring the need for novel treatments that are both safe and effective. The Sigma 1 Receptor (S1R) is a ligand-operated chaperone protein that resides at the mitochondria-associated membrane of the endoplasmic reticulum. The S1R has been shown to play crucial roles in regulating cellular processes implicated in pain modulation. This study explores the potential of PW507, a novel S1R antagonist, as a therapeutic candidate for PDN. PW507 exhibited promising in vitro and in vivo properties in terms of ADME, toxicity, pharmacokinetics, and safety. In preclinical rat models of Streptozotocin-induced diabetic neuropathy, PW507 demonstrated significant efficacy in alleviating mechanical allodynia and thermal hyperalgesia following both acute and chronic (2-week) administration, without inducing tolerance and visual evidence of toxicity. To the best of our knowledge, this is the first report to evaluate an S1R antagonist in STZ-induced diabetic rats following both acute and 2-week chronic administration, offering compelling preclinical evidence for the potential use of PW507 as a promising therapeutic option for PDN.
Collapse
Affiliation(s)
- Youyi Peng
- Biomedical
Informatics Shared Resource, Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08903, United States
| | - Allen H. Zhang
- Department
of Biology, Emory College of Arts and Sciences, Atlanta, Georgia 30322, United States
| | - Liping Wei
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - William J. Welsh
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
2
|
Shen B, Yoon D, Castillo J, Biswal S. A Practical Guide to Sigma-1 Receptor Positron Emission Tomography/Magnetic Resonance Imaging: A New Clinical Molecular Imaging Method to Identify Peripheral Pain Generators in Patients with Chronic Pain. Semin Musculoskelet Radiol 2023; 27:601-617. [PMID: 37935207 PMCID: PMC10629991 DOI: 10.1055/s-0043-1775744] [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] [Indexed: 11/09/2023]
Abstract
Accurately identifying the peripheral pain generator in patients with chronic pain remains a major challenge for modern medicine. Millions of patients around the world suffer endlessly from difficult-to-manage debilitating pain because of very limited diagnostic tests and a paucity of pain therapies. To help these patients, we have developed a novel clinical molecular imaging approach, and, in its early stages, it has been shown to accurately identify the exact site of pain generation using an imaging biomarker for the sigma-1 receptor and positron emission tomography/magnetic resonance imaging. We hope the description of the work in this article can help others begin their own pain imaging programs at their respective institutions.
Collapse
Affiliation(s)
- Bin Shen
- Cyclotron Radiochemistry Facility, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Daehyun Yoon
- Department of Radiology, University of California San Francisco School of Medicine, San Francisco, California
| | - Jessa Castillo
- Radiochemistry Facility, University of California San Francisco School of Medicine, San Francisco, California
| | - Sandip Biswal
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
3
|
de la Puente B, Zamanillo D, Romero L, Carceller A, Vela JM, Merlos M, Portillo-Salido E. Resilience to Pain-Related Depression in σ 1 Receptor Knockout Mice Is Associated with the Reversal of Pain-Induced Brain Changes in Affect-Related Genes. ACS Chem Neurosci 2023; 14:3714-3725. [PMID: 37738096 DOI: 10.1021/acschemneuro.3c00502] [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] [Indexed: 09/24/2023] Open
Abstract
Mice lacking the σ1 receptor chaperone (σ1R-/-) are resilient to depressive-like behaviors secondary to neuropathic pain. Examining the resilience's brain mechanisms could help develop conceptually novel therapeutic strategies. We explored the diminished motivation for a natural reinforcer (white chocolate) in the partial sciatic nerve ligation (PSNL) model in wild-type (WT) and σ1R-/- mice. In the same mice, we performed a comprehensive reverse transcription quantitative PCR (qPCR) analysis across ten brain regions of seven genes implicated in pain regulation and associated affective disorders, such as anxiety and depression. PSNL induced anhedonic-like behavior in WT but not in σ1R-/- mice. In WT mice, PSNL up-regulated dopamine transporter (DAT) and its rate-limiting enzyme, tyrosine hydroxylase (Th), in the ventral tegmental area (VTA) and periaqueductal gray (PAG) as well as the serotonin transporters (SERT) and its rate-limiting enzyme tryptophan hydroxylase 2 (Tph2) in VTA. In addition, μ-opioid receptor (MOR) and σ1R were up-regulated in PAG, and MOR was also elevated in the somatosensory cortex (SS) but down-regulated in the striatum (STR). Finally, increased BDNF was found in the medial prefrontal cortex (mPFC) and hypothalamus (HPT). Sham surgery also produced PSNL-like expression changes in VTA, HPT, and STR. Genetic deletion of the σ1R in mice submitted to PSNL or sham surgery prevented changes in the expression of most of these genes. σ1R is critically involved in the supraspinal gene expression changes produced by PSNL and sham surgery. The changes in gene expression observed in WT mice may be related to pain-related depression, and the absence of these changes observed in σ1R-/- mice may be related to resilience.
Collapse
Affiliation(s)
| | - Daniel Zamanillo
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Luz Romero
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Alicia Carceller
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - José Miguel Vela
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Manuel Merlos
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | | |
Collapse
|
4
|
Malik JA, Agrewala JN. Future perspectives of emerging novel drug targets and immunotherapies to control drug addiction. Int Immunopharmacol 2023; 119:110210. [PMID: 37099943 DOI: 10.1016/j.intimp.2023.110210] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 04/28/2023]
Abstract
Substance Use Disorder (SUD) is one of the major mental illnesses that is terrifically intensifying worldwide. It is becoming overwhelming due to limited options for treatment. The complexity of addiction disorders is the main impediment to understanding the pathophysiology of the illness. Hence, unveiling the complexity of the brain through basic research, identification of novel signaling pathways, the discovery of new drug targets, and advancement in cutting-edge technologies will help control this disorder. Additionally, there is a great hope of controlling the SUDs through immunotherapeutic measures like therapeutic antibodies and vaccines. Vaccines have played a cardinal role in eliminating many diseases like polio, measles, and smallpox. Further, vaccines have controlled many diseases like cholera, dengue, diphtheria, Haemophilus influenza type b (Hib), human papillomavirus, influenza, Japanese encephalitis, etc. Recently, COVID-19 was controlled in many countries by vaccination. Currently, continuous effort is done to develop vaccines against nicotine, cocaine, morphine, methamphetamine, and heroin. Antibody therapy against SUDs is another important area where serious attention is required. Antibodies have contributed substantially against many serious diseases like diphtheria, rabies, Crohn's disease, asthma, rheumatoid arthritis, and bladder cancer. Antibody therapy is gaining immense momentum due to its success rate in cancer treatment. Furthermore, enormous advancement has been made in antibody therapy due to the generation of high-efficiency humanized antibodies with a long half-life. The advantage of antibody therapy is its instant outcome. This article's main highlight is discussing the drug targets of SUDs and their associated mechanisms. Importantly, we have also discussed the scope of prophylactic measures to eliminate drug dependence.
Collapse
Affiliation(s)
- Jonaid Ahmad Malik
- Immunology laboratory, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Javed N Agrewala
- Immunology laboratory, Indian Institute of Technology Ropar, Rupnagar, Punjab, India.
| |
Collapse
|
5
|
Vavers E, Zvejniece L, Dambrova M. Sigma-1 receptor and seizures. Pharmacol Res 2023; 191:106771. [PMID: 37068533 PMCID: PMC10176040 DOI: 10.1016/j.phrs.2023.106771] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 04/19/2023]
Abstract
Over the last decade, sigma-1 receptor (Sig1R) has been recognized as a valid target for the treatment of seizure disorders and seizure-related comorbidities. Clinical trials with Sig1R ligands are underway testing therapies for the treatment of drug-resistant seizures, developmental and epileptic encephalopathies, and photosensitive epilepsy. However, the direct molecular mechanism by which Sig1R modulates seizures and the balance between excitatory and inhibitory pathways has not been fully elucidated. This review article aims to summarize existing knowledge of Sig1R and its involvement in seizures by focusing on the evidence obtained from Sig1R knockout animals and the anti-seizure effects of Sig1R ligands. In addition, this review article includes a discussion of the advantages and disadvantages of the use of existing compounds and describes the challenges and future perspectives on the use of Sig1R as a target for the treatment of seizure disorders.
Collapse
Affiliation(s)
- Edijs Vavers
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles 21, LV-1006, Riga, Latvia; University of Tartu, Faculty of Science and Technology, Institute of Chemistry, Ravila 14a, 50411, Tartu, Estonia.
| | - Liga Zvejniece
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles 21, LV-1006, Riga, Latvia
| | - Maija Dambrova
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles 21, LV-1006, Riga, Latvia; Riga Stradiņš University, Faculty of Pharmacy, Konsula 21, LV-1007, Riga, Latvia
| |
Collapse
|
6
|
Ruiz-Cantero MC, Cortés-Montero E, Jain A, Montilla-García Á, Bravo-Caparrós I, Shim J, Sánchez-Blázquez P, Woolf CJ, Baeyens JM, Cobos EJ. The sigma-1 receptor curtails endogenous opioid analgesia during sensitization of TRPV1 nociceptors. Br J Pharmacol 2023; 180:1148-1167. [PMID: 36478100 DOI: 10.1111/bph.16003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Peripheral sensitization contributes to pathological pain. While prostaglandin E2 (PGE2) and nerve growth factor (NGF) sensitize peptidergic C-nociceptors (TRPV1+), glial cell line-derived neurotrophic factor (GDNF) sensitizes non-peptidergic C-neurons (IB4+). The sigma-1 receptor (sigma-1R) is a Ca2+ -sensing chaperone known to modulate opoid analgesia. This receptor binds both to TRPV1 and the μ opioid receptor, although the functional repercussions of these physical interactions in peripheral sensitization are unknown. EXPERIMENTAL APPROACH We tested the effects of sigma-1 antagonism on PGE2-, NGF-, and GDNF-induced mechanical and heat hyperalgesia in mice. We used immunohistochemistry to determine the presence of endomorphin-2, an endogenous μ receptor agonist, on dorsal root ganglion (DRG) neurons. Recombinant proteins were used to study the interactions between sigma-1R, μ- receptor, and TRPV1. We used calcium imaging to study the effects of sigma-1 antagonism on PGE2-induced sensitization of TRPV1+ nociceptors. KEY RESULTS Sigma1 antagonists reversed PGE2- and NGF-induced hyperalgesia but not GDNF-induced hyperalgesia. Endomorphin-2 was detected on TRPV1+ but not on IB4+ neurons. Peripheral opioid receptor antagonism by naloxone methiodide or administration of an anti-endomorphin-2 antibody to a sensitized paw reversed the antihyperalgesia induced by sigma-1 antagonists. Sigma-1 antagonism transfers sigma-1R from TRPV1 to μ receptors, suggesting that sigma-1R participate in TRPV1-μ receptor crosstalk. Moreover, sigma-1 antagonism reversed, in a naloxone-sensitive manner, PGE2-induced sensitization of DRG neurons to the calcium flux elicited by capsaicin, the prototypic TRPV1 agonist. CONCLUSION AND IMPLICATIONS Sigma-1 antagonism harnesses endogenous opioids produced by TRPV1+ neurons to reduce hyperalgesia by increasing μ receptor activity.
Collapse
Affiliation(s)
- M Carmen Ruiz-Cantero
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain.,Biosanitary Research Institute ibs. GRANADA, Granada, Spain
| | - Elsa Cortés-Montero
- Department of Translational Neurosciences, Neuropharmacology, Cajal Institute, CSIC, Madrid, Spain
| | - Aakanksha Jain
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Ángeles Montilla-García
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
| | - Inmaculada Bravo-Caparrós
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
| | - Jaehoon Shim
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Pilar Sánchez-Blázquez
- Department of Translational Neurosciences, Neuropharmacology, Cajal Institute, CSIC, Madrid, Spain
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - José M Baeyens
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain.,Biosanitary Research Institute ibs. GRANADA, Granada, Spain
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain.,Biosanitary Research Institute ibs. GRANADA, Granada, Spain.,Teófilo Hernando Institute for Drug Discovery, Madrid, Spain
| |
Collapse
|
7
|
Munguia-Galaviz FJ, Miranda-Diaz AG, Cardenas-Sosa MA, Echavarria R. Sigma-1 Receptor Signaling: In Search of New Therapeutic Alternatives for Cardiovascular and Renal Diseases. Int J Mol Sci 2023; 24:ijms24031997. [PMID: 36768323 PMCID: PMC9916216 DOI: 10.3390/ijms24031997] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Cardiovascular and renal diseases are among the leading causes of death worldwide, and regardless of current efforts, there is a demanding need for therapeutic alternatives to reduce their progression to advanced stages. The stress caused by diseases leads to the activation of protective mechanisms in the cell, including chaperone proteins. The Sigma-1 receptor (Sig-1R) is a ligand-operated chaperone protein that modulates signal transduction during cellular stress processes. Sig-1R interacts with various ligands and proteins to elicit distinct cellular responses, thus, making it a potential target for pharmacological modulation. Furthermore, Sig-1R ligands activate signaling pathways that promote cardioprotection, ameliorate ischemic injury, and drive myofibroblast activation and fibrosis. The role of Sig-1R in diseases has also made it a point of interest in developing clinical trials for pain, neurodegeneration, ischemic stroke, depression in patients with heart failure, and COVID-19. Sig-1R ligands in preclinical models have significantly beneficial effects associated with improved cardiac function, ventricular remodeling, hypertrophy reduction, and, in the kidney, reduced ischemic damage. These basic discoveries could inform clinical trials for heart failure (HF), myocardial hypertrophy, acute kidney injury (AKI), and chronic kidney disease (CKD). Here, we review Sig-1R signaling pathways and the evidence of Sig-1R modulation in preclinical cardiac and renal injury models to support the potential therapeutic use of Sig-1R agonists and antagonists in these diseases.
Collapse
Affiliation(s)
- Francisco Javier Munguia-Galaviz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Division de Ciencias de la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzman 49000, Jalisco, Mexico
| | - Alejandra Guillermina Miranda-Diaz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Miguel Alejandro Cardenas-Sosa
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Raquel Echavarria
- CONACYT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
- Correspondence:
| |
Collapse
|
8
|
Vidal-Torres A, Fernández-Pastor B, García M, Ayet E, Cabot A, Burgueño J, Monroy X, Aubel B, Codony X, Romero L, Pascual R, Serafini MT, Encina G, Almansa C, Zamanillo D, Merlos M, Vela JM. Bispecific sigma-1 receptor antagonism and mu-opioid receptor partial agonism: WLB-73502, an analgesic with improved efficacy and safety profile compared to strong opioids. Acta Pharm Sin B 2023; 13:82-99. [PMID: 36815042 PMCID: PMC9939367 DOI: 10.1016/j.apsb.2022.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/17/2022] [Accepted: 07/18/2022] [Indexed: 11/01/2022] Open
Abstract
Opioids are the most effective painkillers, but their benefit-risk balance often hinder their therapeutic use. WLB-73502 is a dual, bispecific compound that binds sigma-1 (S1R) and mu-opioid (MOR) receptors. WLB-73502 is an antagonist at the S1R. It behaved as a partial MOR agonist at the G-protein pathway and produced no/unsignificant β-arrestin-2 recruitment, thus demonstrating low intrinsic efficacy on MOR at both signalling pathways. Despite its partial MOR agonism, WLB-73502 exerted full antinociceptive efficacy, with potency superior to morphine and similar to oxycodone against nociceptive, inflammatory and osteoarthritis pain, and superior to both morphine and oxycodone against neuropathic pain. WLB-73502 crosses the blood-brain barrier and binds brain S1R and MOR to an extent consistent with its antinociceptive effect. Contrary to morphine and oxycodone, tolerance to its antinociceptive effect did not develop after repeated 4-week administration. Also, contrary to opioid comparators, WLB-73502 did not inhibit gastrointestinal transit or respiratory function in rats at doses inducing full efficacy, and it was devoid of proemetic effect (retching and vomiting) in ferrets at potentially effective doses. WLB-73502 benefits from its bivalent S1R antagonist and partial MOR agonist nature to provide an improved antinociceptive and safety profile respect to strong opioid therapy.
Collapse
|
9
|
Garzón-Niño J, Cortés-Montero E, Rodríguez-Muñoz M, Sánchez-Blázquez P. αN-Acetyl β-Endorphin Is an Endogenous Ligand of σ1Rs That Regulates Mu-Opioid Receptor Signaling by Exchanging G Proteins for σ2Rs in σ1R Oligomers. Int J Mol Sci 2022; 24:ijms24010582. [PMID: 36614024 PMCID: PMC9820303 DOI: 10.3390/ijms24010582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/11/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
The opioid peptide β-endorphin coexists in the pituitary and brain in its αN-acetylated form, which does not bind to opioid receptors. We now report that these neuropeptides exhibited opposite effects in in vivo paradigms, in which ligands of the sigma type 1 receptor (σ1R) displayed positive effects. Thus, αN-acetyl β-Endorphin reduced vascular infarct caused by permanent unilateral middle cerebral artery occlusion and diminished the incidence of N-methyl-D-aspartate acid-promoted convulsive syndrome and mechanical allodynia caused by unilateral chronic constriction of the sciatic nerve. Moreover, αN-acetyl β-Endorphin reduced the analgesia of morphine, β-Endorphin and clonidine but enhanced that of DAMGO. All these effects were counteracted by β-Endorphin and absent in σ1R-/- mice. We observed that σ1Rs negatively regulate mu-opioid receptor (MOR)-mediated morphine analgesia by binding and sequestering G proteins. In this scenario, β-Endorphin promoted the exchange of σ2Rs by G proteins at σ1R oligomers and increased the regulation of G proteins by MORs. The opposite was observed for the αN-acetyl derivative, as σ1R oligomerization decreased and σ2R binding was favored, which displaced G proteins; thus, MOR-regulated transduction was reduced. Our findings suggest that the pharmacological β-Endorphin-specific epsilon receptor is a σ1R-regulated MOR and that β-Endorphin and αN-acetyl β-Endorphin are endogenous ligands of σ1R.
Collapse
|
10
|
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
|
11
|
Hashemi M, Karami M, Zarrindast MR. The regulatory role of nitric oxide in morphine-induced analgesia in the descending path of pain from the dorsal hippocampus to the dorsolateral periaqueductal gray. Eur J Pain 2022; 26:888-901. [PMID: 35090066 DOI: 10.1002/ejp.1916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 12/30/2021] [Accepted: 01/23/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Nitric oxide (NO) levels in brain nuclei, such as the hippocampus and brainstem, are involved in morphine analgesia, but the relationship between the dorsal hippocampus (dH) and the dorsolateral periaqueductal gray matter (dlPAG) needs to be clarified, which is our goal. METHODS Wistar rats were simultaneously equipped with a stereotaxic device with unilateral guide cannula at dH and dlPAG. After recovery, they were divided into control and experimental groups. Formalin (50 μL of 2.5%) was inoculated into the left hind paw of rat. Morphine (6 mg/kg) was administered intraperitoneally (i.p.) 10 min before formalin injection. L-Arginine (0.25, 0.5, 1 and 2 μg/rat), and L-NAME (0.25, 0.5, 1 and 2 μg/rat), unrelatedly or with the respect in the order of injection were used in the nuclei before morphine injection (i.p.). Activation of the neuronal NO synthase (nNOS) in the brains of all animals was measured using NADPH-diaphorase, a selective biochemical marker of nNOS. RESULTS Morphine reduced inflammatory pain in the early and late stages of the rat formalin test. The morphine response was attenuated by before injection of single L-arginine but not L-NAME in the two target areas. However, the acute phase result was stopped due to L-NAME pretreatment. When L-NAME was injected into dlPAG before injecting L-arginine at dH, the morphine response did not decrease at all, indicating a modulatory role of NO in dlPAG, which was confirmed by NADPH-d staining. CONCLUSIONS High levels of NO in dlPAG may regulate pain process in downward synaptic interactions.
Collapse
Affiliation(s)
- Mahboobeh Hashemi
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | - Manizheh Karami
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | | |
Collapse
|
12
|
Forth N, Nguyen M, Grech A. A Case Report of Subanesthetic Ketamine Bolus and Infusion for Opioid Refractory Cancer Pain. J Palliat Med 2022; 25:1161-1165. [PMID: 35085456 DOI: 10.1089/jpm.2021.0239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Opioids and traditional adjuvant medications are frequently prescribed for the management of moderate to severe cancer pain with good effect. However, there are many cases, in which patients experience severe opioid refractory cancer pain. Ketamine is being used more frequently in the hospice and palliative setting to manage opioid refractory pain, although high-quality evidence regarding its effectiveness is lacking. It seems certain patients respond favorably to ketamine, while others experience no effect. Studies have not yet identified factors associated with a favorable response to ketamine. We present a case describing the successful treatment of high-dose opioid refractory cancer pain with a subanesthetic ketamine infusion and propose the novel use of a preinfusion test bolus of ketamine to identify patients who are likely to respond favorably to an infusion.
Collapse
Affiliation(s)
- Nicole Forth
- Department of Hospice and Palliative Medicine, Henry Ford Health System, Detroit, Michigan, USA
| | - Michelle Nguyen
- Department of Hospice and Palliative Medicine, Henry Ford Health System, Detroit, Michigan, USA
| | - Anthony Grech
- Department of Hospice and Palliative Medicine, Henry Ford Health System, Detroit, Michigan, USA
| |
Collapse
|
13
|
Xiong J, Zhuang T, Ma Y, Xu J, Ye J, Ma R, Zhang S, Liu X, Liu BF, Hao C, Zhang G, Chen Y. Optimization of bifunctional piperidinamide derivatives as σ 1R Antagonists/MOR agonists for treating neuropathic pain. Eur J Med Chem 2021; 226:113879. [PMID: 34628236 DOI: 10.1016/j.ejmech.2021.113879] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/11/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022]
Abstract
Here, we describe the optimization, synthesis, and associated pharmacological analgesic activities of a new series of bifunctional piperidinamide derivatives as sigma-1 receptor (σ1R) antagonists and mu opioid receptor (MOR) agonists. The new compounds were evaluated in vitro in σ1R and MOR binding assays. The most promising compound 114 (also called HKC-126), showed superior affinities for σ1R and MOR and good selectivity to additional receptors related to pain. Compound 114 showed powerful dose-dependent analgesic effects in the acetic acid writhing test, formalin test, hot plate test, and chronic constriction injury (CCI) neuropathic pain model. In contrast to an equianalgesic dose of fentanyl, compound 114 produced fewer opioid-like side effects, such as reward liability, respiratory depression, physical dependence, and sedation. Lastly, the pharmacokinetic properties of this drug were also acceptable, and these results suggest that compound 114, as a mixed σ1R/MOR ligand, has potential for treating neuropathic pain.
Collapse
MESH Headings
- Acetic Acid
- Amides/chemical synthesis
- Amides/chemistry
- Amides/pharmacology
- Animals
- Behavior, Animal/drug effects
- Dose-Response Relationship, Drug
- Formaldehyde
- Guinea Pigs
- Mice
- Mice, Inbred ICR
- Molecular Dynamics Simulation
- Molecular Structure
- Neuralgia/chemically induced
- Neuralgia/drug therapy
- Neuralgia/metabolism
- Pain Measurement
- Piperidines/chemical synthesis
- Piperidines/chemistry
- Piperidines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Receptors, sigma/antagonists & inhibitors
- Receptors, sigma/metabolism
- Structure-Activity Relationship
- Sigma-1 Receptor
Collapse
Affiliation(s)
- Jiaying Xiong
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tao Zhuang
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yurong Ma
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Junyi Xu
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jiaqi Ye
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Ru Ma
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Shuang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xin Liu
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chao Hao
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Guisen Zhang
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Yin Chen
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
14
|
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
|
15
|
Zhuang T, Xiong J, Hao S, Du W, Liu Z, Liu B, Zhang G, Chen Y. Bifunctional μ opioid and σ 1 receptor ligands as novel analgesics with reduced side effects. Eur J Med Chem 2021; 223:113658. [PMID: 34175542 DOI: 10.1016/j.ejmech.2021.113658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022]
Abstract
Opioid analgesics are highly effective painkillers for the treatment of moderate or severe pain, but they are associated with a number of undesirable adverse effects, including the development of tolerance, addiction, constipation and life-threatening respiratory depression. The development of new and safer analgesics with innovative mechanisms of action, which can enhance the efficacy in comparison to available treatments and reduce their side effects, is urgently needed. The sigma-1 receptor (σ1R), a unique Ca2+-sensing chaperone protein, is expressed throughout pain-modulating tissues and affects neurotransmission by interacting with different protein partners, including molecular targets that participate in nociceptive signalling, such as the μ-opioid receptor (MOR), N-methyl-d-aspartate receptor (NMDAR) and cannabinoid 1 receptor (CB1R). Overwhelming pharmacological and genetic evidence indicates that σ1R antagonists induce anti-hypersensitive effects in sensitising pain conditions (e.g. chemically induced, inflammatory and neuropathic pain) and enhance opioid analgesia but not opioid-mediated detrimental effects. It has been suggested that balanced modulation of MORs and σ1Rs may improve both the therapeutic efficacy and safety of opioids. This review summarises the functional profiles of ligands with mixed MOR agonist and σ1R antagonist activities and highlights their therapeutic potentials for pain management. Dual MOR agonism/σ1R antagonism represents a promising avenue for the development of potent and safer analgesics.
Collapse
MESH Headings
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/chemistry
- Analgesics, Opioid/metabolism
- Analgesics, Opioid/therapeutic use
- Benzopyrans/chemistry
- Benzopyrans/metabolism
- Humans
- Ligands
- Pain/drug therapy
- Piperazines/chemistry
- Piperazines/metabolism
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Receptors, sigma/antagonists & inhibitors
- Receptors, sigma/metabolism
- Sigma-1 Receptor
Collapse
Affiliation(s)
- Tao Zhuang
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jiaying Xiong
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Shuaishuai Hao
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wei Du
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Bifeng Liu
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Guisen Zhang
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Yin Chen
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
16
|
Romeo G, Bonanno F, Wilson LL, Arena E, Modica MN, Pittalà V, Salerno L, Prezzavento O, McLaughlin JP, Intagliata S. Development of New Benzylpiperazine Derivatives as σ 1 Receptor Ligands with in Vivo Antinociceptive and Anti-Allodynic Effects. ACS Chem Neurosci 2021; 12:2003-2012. [PMID: 34019387 PMCID: PMC8291485 DOI: 10.1021/acschemneuro.1c00106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
σ-1 receptors (σ1R) modulate nociceptive signaling, driving the search for selective antagonists to take advantage of this promising target to treat pain. In this study, a new series of benzylpiperazinyl derivatives has been designed, synthesized, and characterized for their affinities toward σ1R and selectivity over the σ-2 receptor (σ2R). Notably, 3-cyclohexyl-1-{4-[(4-methoxyphenyl)methyl]piperazin-1-yl}propan-1-one (15) showed the highest σ1R receptor affinity (Ki σ1 = 1.6 nM) among the series with a significant improvement of the σ1R selectivity (Ki σ2/Ki σ1= 886) compared to the lead compound 8 (Ki σ2/Ki σ1= 432). Compound 15 was further tested in a mouse formalin assay of inflammatory pain and chronic nerve constriction injury (CCI) of neuropathic pain, where it produced dose-dependent (3-60 mg/kg, i.p.) antinociception and anti-allodynic effects. Moreover, compound 15 demonstrated no significant effects in a rotarod assay, suggesting that this σ1R antagonist did not produce sedation or impair locomotor responses. Overall, these results encourage the further development of our benzylpiperazine-based σ1R antagonists as potential therapeutics for chronic pain.
Collapse
Affiliation(s)
- Giuseppe Romeo
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Federica Bonanno
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Lisa L. Wilson
- Department
of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Emanuela Arena
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Maria N. Modica
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Valeria Pittalà
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Loredana Salerno
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Jay P. McLaughlin
- Department
of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Sebastiano Intagliata
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
17
|
Voronin MV, Vakhitova YV, Tsypysheva IP, Tsypyshev DO, Rybina IV, Kurbanov RD, Abramova EV, Seredenin SB. Involvement of Chaperone Sigma1R in the Anxiolytic Effect of Fabomotizole. Int J Mol Sci 2021; 22:5455. [PMID: 34064275 PMCID: PMC8196847 DOI: 10.3390/ijms22115455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/19/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
Sigma-1 receptor (chaperone Sigma1R) is an intracellular protein with chaperone functions, which is expressed in various organs, including the brain. Sigma1R participates in the regulation of physiological mechanisms of anxiety (Su, T. P. et al., 2016) and reactions to emotional stress (Hayashi, T., 2015). In 2006, fabomotizole (ethoxy-2-[2-(morpholino)-ethylthio]benzimidazole dihydrochloride) was registered in Russia as an anxiolytic (Seredenin S. and Voronin M., 2009). The molecular targets of fabomotizole are Sigma1R, NRH: quinone reductase 2 (NQO2), and monoamine oxidase A (MAO-A) (Seredenin S. and Voronin M., 2009). The current study aimed to clarify the dependence of fabomotizole anxiolytic action on its interaction with Sigma1R and perform a docking analysis of fabomotizole interaction with Sigma1R. An elevated plus maze (EPM) test revealed that the anxiolytic-like effect of fabomotizole (2.5 mg/kg i.p.) administered to male BALB/c mice 30 min prior EPM exposition was blocked by Sigma1R antagonists BD-1047 (1.0 mg/kg i.p.) and NE-100 (1.0 mg/kg i.p.) pretreatment. Results of initial in silico study showed that fabomotizole locates in the active center of Sigma1R, reproducing the interactions with the site's amino acids common for established Sigma1R ligands, with the ΔGbind value closer to that of agonist (+)-pentazocine in the 6DK1 binding site.
Collapse
Affiliation(s)
- Mikhail V. Voronin
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (I.P.T.); (D.O.T.); (I.V.R.); (R.D.K.); (E.V.A.)
| | - Yulia V. Vakhitova
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (I.P.T.); (D.O.T.); (I.V.R.); (R.D.K.); (E.V.A.)
| | | | | | | | | | | | - Sergei B. Seredenin
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (I.P.T.); (D.O.T.); (I.V.R.); (R.D.K.); (E.V.A.)
| |
Collapse
|
18
|
Cortés-Montero E, Rodríguez-Muñoz M, Sánchez-Blázquez P, Garzón-Niño J. Human HINT1 Mutant Proteins that Cause Axonal Motor Neuropathy Exhibit Anomalous Interactions with Partner Proteins. Mol Neurobiol 2021; 58:1834-1845. [PMID: 33404983 DOI: 10.1007/s12035-020-02265-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/16/2020] [Indexed: 11/25/2022]
Abstract
The 14 kDa histidine triad nucleotide-binding protein 1 (HINT1) is critical to maintain the normal function of motor neurons. Thus, a series of human HINT1 mutants cause autosomal recessive axonal neuropathy with neuromyotonia. HINT1 establishes a series of regulatory interactions with signaling proteins, some of which are enriched in motor neurons, such as the type 1 sigma receptor or intracellular domain (ICD) of transmembrane teneurin 1, both of which are also implicated in motor disturbances. In a previous study, we reported the capacity of HINT1 to remove the small ubiquitin-like modifier (SUMO) from a series of substrates and the influence of HINT1 mutants on this activity. We now report how human HINT1 mutations affect the interaction of HINT1 with the regulator of its SUMOylase activity, calcium-activated calmodulin, and its substrate SUMO. Moreover, HINT1 mutants exhibited anomalous interactions with G protein coupled receptors, such as the mu-opioid, and with glutamate N-methyl-D-aspartate receptors as well. Additionally, these HINT1 mutants showed impaired associations with transcriptional regulators such as the regulator of G protein signaling Z2 protein and the cleaved N-terminal ICD of teneurin 1. Thus, the altered enzymatic activity of human HINT1 mutants and their anomalous interactions with partner proteins may disrupt signaling pathways essential to the normal function of human motor neurons.
Collapse
Affiliation(s)
- Elsa Cortés-Montero
- Neuropharmacology, Cajal Institute, Department of Translational Neuroscience, CSIC, Madrid, Spain
| | - María Rodríguez-Muñoz
- Neuropharmacology, Cajal Institute, Department of Translational Neuroscience, CSIC, Madrid, Spain
| | - Pilar Sánchez-Blázquez
- Neuropharmacology, Cajal Institute, Department of Translational Neuroscience, CSIC, Madrid, Spain
| | - Javier Garzón-Niño
- Neuropharmacology, Cajal Institute, Department of Translational Neuroscience, CSIC, Madrid, Spain.
| |
Collapse
|
19
|
Liu P, Chu Z, Lei G, Deng L, Yang L, Dang Y. The role of HINT1 protein in morphine addiction: An animal model-based study. Addict Biol 2021; 26:e12897. [PMID: 32171181 DOI: 10.1111/adb.12897] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/25/2020] [Accepted: 03/06/2020] [Indexed: 01/17/2023]
Abstract
Drug addiction is a recurrent, chronic brain disease. The existing treatment methods have limitations, such as poor adherence and inability to completely avoid relapse. Histidine triad nucleotide-binding protein 1 (HINT1) is involved in many neuropsychiatric diseases, such as schizophrenia, pain, and drug dependence. Studies have confirmed that there is a genetic link between HINT1 and addictions such as nicotine and cocaine. However, there is no research on the role of HINT1 protein in morphine addiction at home and abroad. Thus, we designed this project by constructing different types of morphine addiction animal models, including conditioned place preference and behavioral sensitization. We comprehensively examined the participation of HINT1 protein in key brain regions associated with addiction, including prefrontal cortex, nucleus accumbens, corpus striatum, and hippocampus, in different stages of different models. In addition, we used HINT1 knockout mice to establish the above models and physical dependence model to investigate the effect of HINT1 protein deletion on morphine addiction-related behaviors. We found that HINT1 has varying degrees of involvement in different stages of multiple addictive animal models. The absence of HINT1 can attenuate morphine-mediated addictive behavior to a certain extent and can alleviate the withdrawal symptoms of morphine.
Collapse
Affiliation(s)
- Peng Liu
- College of Medicine and Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry Xi'an Jiaotong University Health Science Center Xi'an 710061 China
- Department of Pharmacology and Toxicology Institute of Basic Medicine Science, Xi'an Medical University Xi'an 710021 China
| | - Zheng Chu
- College of Medicine and Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry Xi'an Jiaotong University Health Science Center Xi'an 710061 China
| | - Gang Lei
- College of Medicine and Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry Xi'an Jiaotong University Health Science Center Xi'an 710061 China
| | - Li‐sha Deng
- College of Medicine and Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry Xi'an Jiaotong University Health Science Center Xi'an 710061 China
| | - Liu Yang
- College of Medicine and Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry Xi'an Jiaotong University Health Science Center Xi'an 710061 China
| | - Yong‐hui Dang
- College of Medicine and Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry Xi'an Jiaotong University Health Science Center Xi'an 710061 China
| |
Collapse
|
20
|
Mena-Valdés LC, Blanco-Hernández Y, Espinosa-Juárez JV, López-Muñoz FJ. Haloperidol potentiates antinociceptive effects of morphine and disrupt opioid tolerance. Eur J Pharmacol 2021; 893:173825. [PMID: 33347818 DOI: 10.1016/j.ejphar.2020.173825] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 01/06/2023]
Abstract
Haloperidol is an antipsychotic agent recently described as an antinociceptive drug able to mediate the antagonism of sigma-1 receptors while morphine is an opioid used in the treatment of neuropathic pain. The objectives of this work were to determine the type of interaction generated by the combination of morphine and haloperidol in neuropathic pain induced by chronic constriction injury and to evaluate morphine tolerance and side effects. The antiallodynic and anti-hyperalgesic effects of morphine (0.01-3.16 mg/kg, s.c.) and haloperidol (0.0178-0.1778 mg/kg, s.c.) were determined after single-doses, in monotherapy and combined, using the acetone and von Frey tests, respectively. Evaluations were performed until 10-days postsurgery. Data were processed using "Surface of Synergic Interaction analysis". The rotarod test was used to evaluate motor coordination, and the constipation test was performed using 5% charcoal. The effects of haloperidol and BD-1063, sigma-1 receptor antagonists, naloxone and PRE-084 (sigma-1 agonist) were determined using the morphine-tolerance model. Morphine (0.0316 mg/kg)+haloperidol (0.0178 mg/kg) was determined to be the optimal combination. Morphine-tolerance was observed on day 5 after 11 administrations, although in animals that received the combination, tolerance was delayed until day 8. PRE-084 and naloxone administered on day 5 in animals treated with the combination resulted in a blockade of its antiallodynic effects. Adverse effects of constipation or motor incoordination were not shown in animals treated with morphine + haloperidol. In conclusion, haloperidol enhances the antinociceptive effects of morphine without significant adverse effects, as it is able to disrupt or delay the morphine-tolerance in neuropathic pain.
Collapse
Affiliation(s)
- Licet Caridad Mena-Valdés
- Laboratorio No.7 "Dolor y Analgesia" Del Departamento de Farmacobiología, Cinvestav-Sede Sur, Calz. de Los Tenorios No. 235, Col. Granjas Coapa, C.P. 14330, Ciudad de México, Mexico.
| | - Yisel Blanco-Hernández
- Laboratorio No.7 "Dolor y Analgesia" Del Departamento de Farmacobiología, Cinvestav-Sede Sur, Calz. de Los Tenorios No. 235, Col. Granjas Coapa, C.P. 14330, Ciudad de México, Mexico.
| | - Josué Vidal Espinosa-Juárez
- Escuela de Ciencias Químicas Sede Ocozocoautla, Universidad Autónoma de Chiapas, Ocozocoautla de Espinosa, Chiapas, Mexico.
| | - Francisco Javier López-Muñoz
- Laboratorio No.7 "Dolor y Analgesia" Del Departamento de Farmacobiología, Cinvestav-Sede Sur, Calz. de Los Tenorios No. 235, Col. Granjas Coapa, C.P. 14330, Ciudad de México, Mexico.
| |
Collapse
|
21
|
Peng Y, Zhang Q, Welsh WJ. Novel Sigma 1 Receptor Antagonists as Potential Therapeutics for Pain Management. J Med Chem 2021; 64:890-904. [PMID: 33372782 DOI: 10.1021/acs.jmedchem.0c01964] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The sigma 1 receptor (S1R) is a molecular chaperone protein located in the endoplasmic reticulum and plasma membranes and has been shown to play important roles in various pathological disorders including pain and, as recently discovered, COVID-19. Employing structure- and QSAR-based drug design strategies, we rationally designed, synthesized, and biologically evaluated a series of novel triazole-based S1R antagonists. Compound 10 exhibited potent binding affinity for S1R, high selectivity over S2R and 87 other human targets, acceptable in vitro metabolic stability, slow clearance in liver microsomes, and excellent blood-brain barrier permeability in rats. Further in vivo studies in rats showed that 10 exhibited negligible acute toxicity in the rotarod test and statistically significant analgesic effects in the formalin test for acute inflammatory pain and paclitaxel-induced neuropathic pain models during cancer chemotherapy. These encouraging results promote further development of our triazole-based S1R antagonists as novel treatments for pain of different etiologies.
Collapse
Affiliation(s)
- Youyi Peng
- Biomedical Informatics Shared Resource, Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, 195 Little Albany Street, New Brunswick, New Jersey 08903, United States
| | - Qiang Zhang
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, 661 Hoes Lane West, Piscataway, New Jersey 08854, United States
| | - William J Welsh
- Biomedical Informatics Shared Resource, Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, 195 Little Albany Street, New Brunswick, New Jersey 08903, United States
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, 661 Hoes Lane West, Piscataway, New Jersey 08854, United States
| |
Collapse
|
22
|
Dash R, Ali MC, Jahan I, Munni YA, Mitra S, Hannan MA, Timalsina B, Oktaviani DF, Choi HJ, Moon IS. Emerging potential of cannabidiol in reversing proteinopathies. Ageing Res Rev 2021; 65:101209. [PMID: 33181336 DOI: 10.1016/j.arr.2020.101209] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/22/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022]
Abstract
The aberrant accumulation of disease-specific protein aggregates accompanying cognitive decline is a pathological hallmark of age-associated neurological disorders, also termed as proteinopathies, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and multiple sclerosis. Along with oxidative stress and neuroinflammation, disruption in protein homeostasis (proteostasis), a network that constitutes protein surveillance system, plays a pivotal role in the pathobiology of these dementia disorders. Cannabidiol (CBD), a non-psychotropic phytocannabinoid of Cannabis sativa, is known for its pleiotropic neuropharmacological effects on the central nervous system, including the ability to abate oxidative stress, neuroinflammation, and protein misfolding. Over the past years, compelling evidence has documented disease-modifying role of CBD in various preclinical and clinical models of neurological disorders, suggesting the potential therapeutic implications of CBD in these disorders. Because of its putative role in the proteostasis network in particular, CBD could be a potent modulator for reversing not only age-associated neurodegeneration but also other protein misfolding disorders. However, the current understanding is insufficient to underpin this proposition. In this review, we discuss the potentiality of CBD as a pharmacological modulator of the proteostasis network, highlighting its neuroprotective and aggregates clearing roles in the neurodegenerative disorders. We anticipate that the current effort will advance our knowledge on the implication of CBD in proteostasis network, opening up a new therapeutic window for aging proteinopathies.
Collapse
|
23
|
Identifying Musculoskeletal Pain Generators Using Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00076-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
24
|
Calmodulin Supports TRPA1 Channel Association with Opioid Receptors and Glutamate NMDA Receptors in the Nervous Tissue. Int J Mol Sci 2020; 22:ijms22010229. [PMID: 33379368 PMCID: PMC7795679 DOI: 10.3390/ijms22010229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/24/2022] Open
Abstract
Transient receptor potential ankyrin member 1 (TRPA1) belongs to the family of thermo TRP cation channels that detect harmful temperatures, acids and numerous chemical pollutants. TRPA1 is expressed in nervous tissue, where it participates in the genesis of nociceptive signals in response to noxious stimuli and mediates mechanical hyperalgesia and allodynia associated with different neuropathies. The glutamate N-methyl-d-aspartate receptor (NMDAR), which plays a relevant role in allodynia to mechanical stimuli, is connected via histidine triad nucleotide-binding protein 1 (HINT1) and type 1 sigma receptor (σ1R) to mu-opioid receptors (MORs), which mediate the most potent pain relief. Notably, neuropathic pain causes a reduction in MOR antinociceptive efficacy, which can be reversed by blocking spinal NMDARs and TRPA1 channels. Thus, we studied whether TRPA1 channels form complexes with MORs and NMDARs that may be implicated in the aforementioned nociceptive signals. Our data suggest that TRPA1 channels functionally associate with MORs, delta opioid receptors and NMDARs in the dorsal root ganglia, the spinal cord and brain areas. These associations were altered in response to pharmacological interventions and the induction of inflammatory and also neuropathic pain. The MOR-TRPA1 and NMDAR-TRPA1 associations do not require HINT1 or σ1R but appear to be mediated by calcium-activated calmodulin. Thus, TRPA1 channels may associate with NMDARs to promote ascending acute and chronic pain signals and to control MOR antinociception.
Collapse
|
25
|
Sánchez-Blázquez P, Cortés-Montero E, Rodríguez-Muñoz M, Merlos M, Garzón-Niño J. The Sigma 2 receptor promotes and the Sigma 1 receptor inhibits mu-opioid receptor-mediated antinociception. Mol Brain 2020; 13:150. [PMID: 33176836 PMCID: PMC7659117 DOI: 10.1186/s13041-020-00676-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/22/2020] [Indexed: 01/10/2023] Open
Abstract
The Sigma-1 receptor (σ1R) has emerged as an interesting pharmacological target because it inhibits analgesia mediated by mu-opioid receptors (MOR), and also facilitates the development of neuropathic pain. Based on these findings, the recent cloning of the Sigma-2 receptor (σ2R) led us to investigate its potential role as a regulator of opioid analgesia and of pain hypersensitivity in σ2R knockout mice. In contrast to σ1R deficient mice, σ2R knockout mice developed mechanical allodynia following establishment of chronic constriction injury-induced neuropathic pain, which was alleviated by the σ1R antagonist S1RA. The analgesic effects of morphine, [D-Ala, N-MePhe, Gly-ol]-encephalin (DAMGO) and β-endorphin increased in σ1R-/- mice and diminished in σ2R-/- mice. The analgesic effect of morphine was increased in σ2R-/- mice by treatment with S1RA. However, σ2R-/- mice and wild-type mice exhibited comparable antinociceptive responses to the delta receptor agonist [D-Pen2,5]-encephalin (DPDPE), the cannabinoid type 1 receptor agonist WIN55,212-2 and the α2-adrenergic receptor agonist clonidine. Therefore, while σR1 inhibits and σ2R facilitates MOR-mediated analgesia these receptors exchange their roles when regulating neuropathic pain perception. Our study may help identify new pharmacological targets for diminishing pain perception and improving opioid detoxification therapies.
Collapse
MESH Headings
- Analgesics/pharmacology
- Animals
- Constriction, Pathologic
- Hyperalgesia/metabolism
- Hyperalgesia/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Morphine/pharmacology
- Nociception/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Receptors, sigma/metabolism
- Sigma-1 Receptor
Collapse
Affiliation(s)
- Pilar Sánchez-Blázquez
- Neuropharmacology, Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Doctor Arce 37, 28002, Madrid, Spain.
| | - Elsa Cortés-Montero
- Neuropharmacology, Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Doctor Arce 37, 28002, Madrid, Spain
| | - María Rodríguez-Muñoz
- Neuropharmacology, Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Doctor Arce 37, 28002, Madrid, Spain
| | - Manuel Merlos
- Drug Discovery & Preclinical Development, Esteve, Barcelona, Spain
| | - Javier Garzón-Niño
- Neuropharmacology, Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Doctor Arce 37, 28002, Madrid, Spain
| |
Collapse
|
26
|
Rodríguez-Muñoz M, Cortés-Montero E, Garzón-Niño J, Sánchez-Blázquez P. The ALS-related σ1R E102Q Mutant Eludes Ligand Control and Exhibits Anomalous Response to Calcium. Int J Mol Sci 2020; 21:E7339. [PMID: 33020464 PMCID: PMC7582951 DOI: 10.3390/ijms21197339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 01/28/2023] Open
Abstract
Sigma receptor type 1 (σ1R) is a transmembrane protein expressed throughout the central nervous system and in certain peripheral tissues. The human σ1R E102Q mutation causes juvenile amyotrophic lateral sclerosis (ALS), likely by inducing a series of alterations in calcium efflux from the endoplasmic reticulum (ER) to mitochondria that affects calcium homeostasis and cellular survival. Here, we report the influence of calcium on σ1R E102Q associations with glutamate N-methyl-D-aspartate receptors (NMDARs), binding immunoglobulin protein (BiP), and transient receptor potential calcium channels A1, V1, and M8. The mutant protein inhibited the binding of calmodulin to these calcium channels and interacted less with BiP than wild-type σ1R, thereby contributing to calcium homeostasis dysfunction. Mutant σ1R, but not wild-type σ1R, strongly bound to histidine triad nucleotide binding protein 1, which regulates neuromuscular synaptic organization and target selection through teneurin 1. While ligands regulated the association of σ1R wild-type with NMDARs and BiP, they failed to modulate the interaction between these proteins and the σ1R E102Q mutant. Thus, the σ1R E102Q mutant exhibited an anomalous response to cytosolic calcium levels, altered affinity for target proteins, and a loss of response to regulatory ligands. We believe that these modifications may contribute to the onset of juvenile ALS.
Collapse
Affiliation(s)
| | | | | | - Pilar Sánchez-Blázquez
- Neuropharmacology, Cajal Institute, CSIC, Avenida Doctor Arce, 37. 28002 Madrid, Spain; (M.R.-M.); (E.C.-M.); (J.G.-N.)
| |
Collapse
|
27
|
Yoon D, Kogan F, Gold GE, Biswal S. Identifying Musculoskeletal Pain Generators Using Clinical PET. Semin Musculoskelet Radiol 2020; 24:441-450. [DOI: 10.1055/s-0040-1713607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AbstractIdentifying the source of a person's pain is a significant clinical challenge because the physical sensation of pain is believed to be subjective and difficult to quantify. The experience of pain is not only modulated by the individual's threshold to painful stimuli but also a product of the person's affective contributions, such as fear, anxiety, and previous experiences. Perhaps then to quantify pain is to examine the degree of nociception and pro-nociceptive inflammation, that is, the extent of cellular, chemical, and molecular changes that occur in pain-generating processes. Measuring changes in the local density of receptors, ion channels, mediators, and inflammatory/immune cells that are involved in the painful phenotype using targeted, highly sensitive, and specific positron emission tomography (PET) radiotracers is therefore a promising approach toward objectively identifying peripheral pain generators. Although several preclinical radiotracer candidates are being developed, a growing number of ongoing clinical PET imaging approaches can measure the degree of target concentration and thus serve as a readout for sites of pain generation. Further, when PET is combined with the spatial and contrast resolution afforded by magnetic resonance imaging, nuclear medicine physicians and radiologists can potentially identify pain drivers with greater accuracy and confidence. Clinical PET imaging approaches with fluorine-18 fluorodeoxyglucose, fluorine-18 sodium fluoride, and sigma-1 receptor PET radioligand and translocator protein radioligands to isolate the source of pain are described here.
Collapse
Affiliation(s)
- Daehyun Yoon
- Division of Musculoskeletal Radiology, Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Feliks Kogan
- Division of Musculoskeletal Radiology, Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Garry E. Gold
- Division of Musculoskeletal Radiology, Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Sandip Biswal
- Division of Musculoskeletal Radiology, Department of Radiology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
28
|
Herrando-Grabulosa M, Gaja-Capdevila N, Vela JM, Navarro X. Sigma 1 receptor as a therapeutic target for amyotrophic lateral sclerosis. Br J Pharmacol 2020; 178:1336-1352. [PMID: 32761823 DOI: 10.1111/bph.15224] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/13/2020] [Accepted: 07/25/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult disease causing a progressive loss of upper and lower motoneurons, muscle paralysis and early death. ALS has a poor prognosis of 3-5 years after diagnosis with no effective cure. The aetiopathogenic mechanisms involved include glutamate excitotoxicity, oxidative stress, protein misfolding, mitochondrial alterations, disrupted axonal transport and inflammation. Sigma non-opioid intracellular receptor 1 (sigma 1 receptor) is a protein expressed in motoneurons, mainly found in the endoplasmic reticulum (ER) on the mitochondria-associated ER membrane (MAM) or in close contact with cholinergic postsynaptic sites. MAMs are sites that allow the assembly of several complexes implicated in essential survival cell functions. The sigma 1 receptor modulates essential mechanisms for motoneuron survival including excitotoxicity, calcium homeostasis, ER stress and mitochondrial dysfunction. This review updates sigma 1 receptor mechanisms and its alterations in ALS, focusing on MAM modulation, which may constitute a novel target for therapeutic strategies. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.
Collapse
Affiliation(s)
- Mireia Herrando-Grabulosa
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Núria Gaja-Capdevila
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José M Vela
- Esteve Pharmaceuticals S.A., Drug Discovery and Preclinical Development, Barcelona, Spain
| | - Xavier Navarro
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Institut Guttmann de Neurorehabilitació, Badalona, Spain
| |
Collapse
|
29
|
Lei G, Liu F, Liu P, Jiao T, Yang L, Chu Z, Deng LS, Li Y, Dang YH. Does genetic mouse model of constitutive Hint1 deficiency exhibit schizophrenia-like behaviors? Schizophr Res 2020; 222:304-318. [PMID: 32439293 DOI: 10.1016/j.schres.2020.05.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 01/13/2023]
Abstract
The histidine triad nucleotide binding protein 1 (HINT1) is closely related to many neuropsychiatric disorders. Clinical studies supported that mutations in the Hint1 gene correlated potentially with schizophrenia. In addition, Hint1 gene knockout (KO) mice exhibited hyperactivity induced by amphetamine and apomorphine. However, it is still unclear whether this animal model exhibits schizophrenia-like behaviors and, if so, their underlying mechanisms remain to be elucidated. Thus, our study sought to evaluate schizophrenia-like behaviors in Hint1-KO mice, and explore the associated changes in neuronal structural plasticity and schizophrenia-related molecules. A series of behavioral tests were used to compare Hint1-KO and their wild-type (WT) littermates, alongside a number of morphological and molecular biological methods. Relative to WT mice, Hint1-KO mice exhibited reduced social interaction behaviors, aggressive behavior, sensorimotor gating deficits, apathetic and self-neglect behaviors, and increased MK-801-induced hyperactivity. Hint1-KO mice also showed partly increased dendritic complexity in the hippocampus (Hip) relative to WT mice. Total glutamate was decreased in the medial prefrontal cortex, nucleus accumbens (NAc), and Hip of KO mice. Expression of NR1, NR2A, and D4R was decreased whereas that of D1R was increased in the NAc of KO relative to WT mice. The expression level of NR2B was increased whereas that of D1R was decreased in the Hip of KO mice. Hint1-KO mice exhibited schizophrenia-like behaviors. Partly increased dendritic complexity and dysfunction in both the dopaminergic and glutamatergic systems may be involved in the abnormalities in Hint1-KO mice.
Collapse
Affiliation(s)
- Gang Lei
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Fei Liu
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Peng Liu
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Tong Jiao
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Liu Yang
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Zheng Chu
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Li-Sha Deng
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yan Li
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yong-Hui Dang
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Key Laboratory of the Health Ministry for Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Key Laboratory of Shaanxi Province for Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; State Key Laboratory for Manufacturing Systems Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
30
|
Xiong J, Jin J, Gao L, Hao C, Liu X, Liu BF, Chen Y, Zhang G. Piperidine propionamide as a scaffold for potent sigma-1 receptor antagonists and mu opioid receptor agonists for treating neuropathic pain. Eur J Med Chem 2020; 191:112144. [PMID: 32087465 DOI: 10.1016/j.ejmech.2020.112144] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/11/2020] [Accepted: 02/11/2020] [Indexed: 11/24/2022]
Abstract
We designed and synthesized a novel series of piperidine propionamide derivatives as potent sigma-1 (σ1) receptor antagonists and mu (μ) opioid receptor agonists, and measured their affinity for σ1 and μ receptors in vitro through binding assays. The basic scaffold of the new compounds contained a 4-substituted piperidine ring and N-aryl propionamide. Compound 44, N-(2-(4-(4-fluorobenzyl) piperidin-1-yl) ethyl)-N-(4-methoxy-phenyl) propionamide, showed the highest affinity for σ1 receptor (Ki σ1 = 1.86 nM) and μ receptor (Ki μ = 2.1 nM). It exhibited potent analgesic activity in the formalin test (ED50 = 15.1 ± 1.67 mg/kg) and had equivalent analgesic effects to S1RA (σ1 antagonist) in a CCI model. Therefore, Compound 44, which has mixed σ1/μ receptor profiles, may be a potential candidate for treating neuropathic pain.
Collapse
Affiliation(s)
- Jiaying Xiong
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jian Jin
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Lanchang Gao
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chao Hao
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xin Liu
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bi-Feng Liu
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yin Chen
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Guisen Zhang
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
31
|
Sigma receptor-induced heavy drinking in rats: Modulation by the opioid receptor system. Pharmacol Biochem Behav 2020; 192:172914. [PMID: 32205151 DOI: 10.1016/j.pbb.2020.172914] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 02/04/2023]
Abstract
Alcohol use disorder (AUD) is a major cause of morbidity and mortality worldwide, for which new efficacious treatments are necessary. The opioid receptor system is a mediator of the rewarding effects of alcohol; in particular, while activation of μ opioid receptors enhances ethanol intake in rodents, opioid-receptor antagonists, such as naloxone and naltrexone, reduce its pleasurable and reinforcing effects, thereby decreasing alcohol. Sigma receptors (Sig-Rs) have been proposed as modulators of the effects of alcohol and, therefore, as a potential new pharmacological target for AUD. Somewhat analogously to μ opioid ligands, SigR agonists increase, while SigR antagonists decrease alcohol intake in animal models of excessive alcohol drinking. However, a potential cross-talk between these two receptor systems in relation to alcohol consumption has so far not been investigated. Here, we addressed this question pharmacologically, by testing the effects of either activating or inhibiting opioid receptors on the heavy alcohol drinking induced by chronic stimulation of SigR in alcohol-preferring rats. We found that the opioid receptor agonist morphine, which per se increases ethanol intake, at a sub-threshold dose reduces the binge-like drinking induced by the repeated treatment with the SigR agonist 1,3-di-o-tolylguanidine (DTG); conversely, the opioid receptor antagonist naltrexone, which per se reduces ethanol intake, at a sub-threshold dose potentiates the DTG-induced binge-like drinking. Our data show a cross-talk between the opioid and SigR systems relevant to the modulation of alcohol drinking, which provides important insights into the neurobiology of AUD and may lead to the development of novel therapies, either standalone or in combination.
Collapse
|
32
|
Carcolé M, Kummer S, Gonçalves L, Zamanillo D, Merlos M, Dickenson AH, Fernández‐Pastor B, Cabañero D, Maldonado R. Sigma-1 receptor modulates neuroinflammation associated with mechanical hypersensitivity and opioid tolerance in a mouse model of osteoarthritis pain. Br J Pharmacol 2019; 176:3939-3955. [PMID: 31332781 PMCID: PMC6811737 DOI: 10.1111/bph.14794] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/06/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Osteoarthritic pain is a chronic disabling condition lacking effective treatment. Continuous use of opioid drugs during osteoarthritic pain induces tolerance and may result in dose escalation and abuse. Sigma-1 (σ1) receptors, a chaperone expressed in key areas for pain control, modulates μ-opioid receptor activity and represents a promising target to tackle these problems. The present study investigates the efficacy of the σ1 receptor antagonist E-52862 to inhibit pain sensitization, morphine tolerance, and associated electrophysiological and molecular changes in a murine model of osteoarthritic pain. EXPERIMENTAL APPROACH Mice received an intra-knee injection of monoiodoacetate followed by 14-day treatment with E-52862, morphine, or vehicle, and mechanical sensitivity was assessed before and after the daily doses. KEY RESULTS Monoiodoacetate-injected mice developed persistent mechanical hypersensitivity, which was dose-dependently inhibited by E-52862. Mechanical thresholds assessed before the daily E-52862 dose showed gradual recovery, reaching complete restoration by the end of the treatment. When repeated treatment started 15 days after knee injury, E-52862 produced enhanced short-term analgesia, but recovery to baseline threshold was slower. Both a σ1 receptor agonist and a μ receptor antagonist blocked the analgesic effects of E-52862. An acute, sub-effective dose of E-52862 restored morphine analgesia in opioid-tolerant mice. Moreover, E-52862 abolished spinal sensitization in osteoarthritic mice and inhibited pain-related molecular changes. CONCLUSION AND IMPLICATIONS These findings show dual effects of σ1 receptor antagonism alleviating both short- and long-lasting antinociception during chronic osteoarthritis pain. They identify E-52862 as a promising pharmacological agent to treat chronic pain and avoid opioid tolerance.
Collapse
Affiliation(s)
- Mireia Carcolé
- Neuropharmacology Lab, Department of Experimental and Health SciencesUniversity Pompeu FabraBarcelonaSpain
| | - Sami Kummer
- Neuropharmacology Lab, Department of Experimental and Health SciencesUniversity Pompeu FabraBarcelonaSpain
| | - Leonor Gonçalves
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | - Daniel Zamanillo
- Drug Discovery and Preclinical Development, Laboratories EsteveBarcelona Science ParkBarcelonaSpain
| | - Manuel Merlos
- Drug Discovery and Preclinical Development, Laboratories EsteveBarcelona Science ParkBarcelonaSpain
| | - Anthony H. Dickenson
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | - Begoña Fernández‐Pastor
- Drug Discovery and Preclinical Development, Laboratories EsteveBarcelona Science ParkBarcelonaSpain
| | - David Cabañero
- Neuropharmacology Lab, Department of Experimental and Health SciencesUniversity Pompeu FabraBarcelonaSpain
| | - Rafael Maldonado
- Neuropharmacology Lab, Department of Experimental and Health SciencesUniversity Pompeu FabraBarcelonaSpain
| |
Collapse
|
33
|
Schmidt HR, Kruse AC. The Molecular Function of σ Receptors: Past, Present, and Future. Trends Pharmacol Sci 2019; 40:636-654. [PMID: 31387763 PMCID: PMC6748033 DOI: 10.1016/j.tips.2019.07.006] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/03/2019] [Accepted: 07/10/2019] [Indexed: 10/26/2022]
Abstract
The σ1 and σ2 receptors are enigmatic proteins that have attracted attention for decades due to the chemical diversity and therapeutic potential of their ligands. However, despite ongoing clinical trials with σ receptor ligands for multiple conditions, relatively little is known regarding the molecular function of these receptors. In this review, we revisit past research on σ receptors and discuss the interpretation of these data in light of recent developments. We provide a synthesis of emerging structural and genetic data on the σ1 receptor and discuss the recent cloning of the σ2 receptor. Finally, we discuss the major questions that remain in the study of σ receptors.
Collapse
Affiliation(s)
- Hayden R Schmidt
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
34
|
Cortés-Montero E, Rodríguez-Muñoz M, Sánchez-Blázquez P, Garzón J. The Axonal Motor Neuropathy-Related HINT1 Protein Is a Zinc- and Calmodulin-Regulated Cysteine SUMO Protease. Antioxid Redox Signal 2019; 31:503-520. [PMID: 31088288 PMCID: PMC6648240 DOI: 10.1089/ars.2019.7724] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aims: Histidine triad nucleotide-binding protein 1 (HINT1) exhibits proapoptotic and tumor-suppressive activity. HINT1 binds to transcription factors such as teneurin1 and to the regulator of G protein signaling 17 (RGS) (Z2) protein, which incorporates the small ubiquitin-like modifier (SUMO), and is implicated in several types of cancer. HINT1 interacts with proteins such as PKCγ and Raf-1 through zinc ions provided by the cysteine-rich domain of RGSZ2 and the coupled neural nitric oxide synthase (nNOS). Recently, a series of HINT1 mutants have been reported to cause human autosomal recessive axonal neuropathy with neuromyotonia (ARAN-NM). However, the specific alteration in the function of HINT1 induced by these mutants remains to be elucidated. Because sumoylation modifies protein association and transcriptional regulation, we investigated whether HINT1 exhibits zinc- and redox-regulated sumoylase activity, which may be altered in those mutants. Results: HINT1 exhibits cysteine protease activity to remove SUMO from a variety of signaling proteins. HINT1 sumoylase activity is blocked by zinc, and it is released by nitric oxide or calcium-activated calmodulin (CaM). HINT1 contains a SUMO-interacting motif (110-116 HIHLHVL) and the catalytic triad Cys84-Asp87-His114 in the C-terminal region. Thus, zinc probably provided by the RGSZ2-nNOS complex may bind to Cys84 to block HINT1 isopeptidase activity. Innovation: To date, HINT1 is the only sumoylase that is regulated by two alternate pathways, redox- and calcium-activated CaM. Conclusion: The 15 human HINT1 mutants reported to cause ARAN-NM exhibited altered sumoylase activity, which may contribute to the onset of this human motor disease.
Collapse
Affiliation(s)
- Elsa Cortés-Montero
- Neuropharmacology, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - María Rodríguez-Muñoz
- Neuropharmacology, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Pilar Sánchez-Blázquez
- Neuropharmacology, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Javier Garzón
- Neuropharmacology, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
35
|
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
|
36
|
Vidal-Torres A, Fernández-Pastor B, Carceller A, Vela JM, Merlos M, Zamanillo D. Supraspinal and Peripheral, but Not Intrathecal, σ 1R Blockade by S1RA Enhances Morphine Antinociception. Front Pharmacol 2019; 10:422. [PMID: 31068818 PMCID: PMC6491787 DOI: 10.3389/fphar.2019.00422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/03/2019] [Indexed: 12/26/2022] Open
Abstract
Sigma-1 receptor (σ1R) antagonism increases the effects of morphine on acute nociceptive pain. S1RA (E-52862) is a selective σ1R antagonist widely used to study the role of σ1Rs. S1RA alone exerted antinociceptive effect in the formalin test in rats and increased noradrenaline levels in the spinal cord, thus accounting for its antinociceptive effect. Conversely, while systemic S1RA failed to elicit antinociceptive effect by itself in the tail-flick test in mice, it did potentiate the antinociceptive effect of opioids in this acute pain model. The present study aimed to investigate the site of action and the involvement of spinal noradrenaline on the potentiation of opioid antinociception by S1RA on acute thermal nociception using the tail-flick test in rats. Local administration was performed after intrathecal catheterization or intracerebroventricular and rostroventral medullar (RVM) cannulae implantation. Noradrenaline levels in the spinal cord were evaluated using the concentric microdialysis technique in awake, freely-moving rats. Systemic or supraspinal administration of S1RA alone, while having no effect on antinociception, enhanced the effect of morphine in rats. However, spinal S1RA administration did not potentiate the antinociceptive effect of morphine. Additionally, the peripherally restricted opioid agonist loperamide was devoid of antinociceptive effect but produced antinociception when combined with S1RA. Neurochemical studies revealed that noradrenaline levels in the dorsal horn of the spinal cord were not increased at doses exerting potentiation of the antinociceptive effect of the opioid. In conclusion, the site of action of σ1R for opioid modulation on acute thermal nociception is located at the peripheral and supraspinal levels, and the opioid-potentiating effect is independent of the spinal noradrenaline increase produced by S1RA.
Collapse
Affiliation(s)
- Alba Vidal-Torres
- Drug Discovery and Preclinical Development, Esteve Pharmaceuticals, Parc Científic Barcelona, Barcelona, Spain
| | - Begoña Fernández-Pastor
- Drug Discovery and Preclinical Development, Esteve Pharmaceuticals, Parc Científic Barcelona, Barcelona, Spain
| | - Alicia Carceller
- Drug Discovery and Preclinical Development, Esteve Pharmaceuticals, Parc Científic Barcelona, Barcelona, Spain
| | - José Miguel Vela
- Drug Discovery and Preclinical Development, Esteve Pharmaceuticals, Parc Científic Barcelona, Barcelona, Spain
| | - Manuel Merlos
- Drug Discovery and Preclinical Development, Esteve Pharmaceuticals, Parc Científic Barcelona, Barcelona, Spain
| | - Daniel Zamanillo
- Drug Discovery and Preclinical Development, Esteve Pharmaceuticals, Parc Científic Barcelona, Barcelona, Spain
| |
Collapse
|
37
|
Vavers E, Zvejniece L, Maurice T, Dambrova M. Allosteric Modulators of Sigma-1 Receptor: A Review. Front Pharmacol 2019; 10:223. [PMID: 30941035 PMCID: PMC6433746 DOI: 10.3389/fphar.2019.00223] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/22/2019] [Indexed: 11/13/2022] Open
Abstract
Allosteric modulators of sigma-1 receptor (Sig1R) are described as compounds that can increase the activity of some Sig1R ligands that compete with (+)-pentazocine, one of the classic prototypical ligands that binds to the orthosteric Sig1R binding site. Sig1R is an endoplasmic reticulum membrane protein that, in addition to its promiscuous high-affinity ligand binding, has been shown to have chaperone activity. Different experimental approaches have been used to describe and validate the activity of allosteric modulators of Sig1R. Sig1R-modulatory activity was first found for phenytoin, an anticonvulsant drug that primarily acts by blocking the voltage-gated sodium channels. Accumulating evidence suggests that allosteric Sig1R modulators affect processes involved in the pathophysiology of depression, memory and cognition disorders as well as convulsions. This review will focus on the description of selective and non-selective allosteric modulators of Sig1R, including molecular structure properties and pharmacological activity both in vitro and in vivo, with the aim of providing the latest overview from compound discovery approaches to eventual clinical applications. In this review, the possible mechanisms of action will be discussed, and future challenges in the development of novel compounds will be addressed.
Collapse
Affiliation(s)
- Edijs Vavers
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Liga Zvejniece
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Tangui Maurice
- MMDN, University of Montpellier, INSERM, EPHE, UMR-S1198, Montpellier, France
| | - Maija Dambrova
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| |
Collapse
|
38
|
Montilla-García Á, Tejada MÁ, Ruiz-Cantero MC, Bravo-Caparrós I, Yeste S, Zamanillo D, Cobos EJ. Modulation by Sigma-1 Receptor of Morphine Analgesia and Tolerance: Nociceptive Pain, Tactile Allodynia and Grip Strength Deficits During Joint Inflammation. Front Pharmacol 2019; 10:136. [PMID: 30853912 PMCID: PMC6395397 DOI: 10.3389/fphar.2019.00136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/06/2019] [Indexed: 12/31/2022] Open
Abstract
Sigma-1 receptor antagonism increases the effects of morphine on nociceptive pain, even in morphine-tolerant animals. However, it is unknown whether these receptors are able to modulate morphine antinociception and tolerance during inflammatory pain. Here we used a mouse model to test the modulation of morphine effects by the selective sigma-1 antagonist S1RA (MR309), by determining its effect on inflammatory tactile allodynia (von Frey filaments) and on grip strength deficits induced by joint inflammation (a measure of pain-induced functional disability), and compared the results with those for nociceptive heat pain recorded with the unilateral hot plate (55°C) test. The subcutaneous (s.c.) administration of morphine induced antinociceptive effects to heat stimuli, and restored mechanical threshold and grip strength in mice with periarticular inflammation induced by Complete Freund’s Adjuvant. S1RA (80 mg/kg, s.c.) administered alone did not induce any effect on nociceptive heat pain or inflammatory allodynia, but was able to partially reverse grip strength deficits. The association of S1RA with morphine, at doses inducing little or no analgesic-like effects when administered alone, resulted in a marked antinociceptive effect to heat stimuli and complete reversion of inflammatory tactile allodynia. However, S1RA administration did not increase the effect of morphine on grip strength deficits induced by joint inflammation. When S1RA (80 mg/kg, s.c.) was administered to morphine-tolerant animals, it rescued the analgesic-like effects of this opioid in all three pain measures. However, when S1RA was repeatedly given during the induction of morphine tolerance (and not on the day of behavioral evaluation) it failed to affect tolerance to the effects of morphine on nociceptive heat pain or inflammatory allodynia, but completely preserved the effects of this opioid on grip strength deficits. These effects of S1RA on morphine tolerance cannot be explained by pharmacokinetic interactions, given that the administration of S1RA did not modify concentrations of morphine or morphine-3-glucuronide (a major morphine metabolite) in morphine-tolerant animals in plasma or brain tissue. We conclude that sigma-1 receptors play a pivotal role in the control of morphine analgesia and tolerance in nociceptive and inflammatory pain, although in a manner dependent on the type of painful stimulus explored.
Collapse
Affiliation(s)
- Ángeles Montilla-García
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, The Biomedical Research Centre, University of Granada, Granada, Spain
| | - Miguel Á Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, The Biomedical Research Centre, University of Granada, Granada, Spain
| | - M Carmen Ruiz-Cantero
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, The Biomedical Research Centre, University of Granada, Granada, Spain
| | - Inmaculada Bravo-Caparrós
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, The Biomedical Research Centre, University of Granada, Granada, Spain
| | - Sandra Yeste
- Drug Discovery and Preclinical Development, ESTEVE, Barcelona, Spain
| | - Daniel Zamanillo
- Drug Discovery and Preclinical Development, ESTEVE, Barcelona, Spain
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, The Biomedical Research Centre, University of Granada, Granada, Spain.,Institute of Neuroscience, The Biomedical Research Centre, University of Granada, Granada, Spain.,Institute of Neuroscience, The Biomedical Research Centre, University of Granada, Granada, Spain
| |
Collapse
|
39
|
Aguinaga D, Medrano M, Cordomí A, Jiménez-Rosés M, Angelats E, Casanovas M, Vega-Quiroga I, Canela EI, Petrovic M, Gysling K, Pardo L, Franco R, Navarro G. Cocaine Blocks Effects of Hunger Hormone, Ghrelin, Via Interaction with Neuronal Sigma-1 Receptors. Mol Neurobiol 2019; 56:1196-1210. [PMID: 29876881 DOI: 10.1007/s12035-018-1140-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/21/2018] [Indexed: 10/14/2022]
Abstract
Despite ancient knowledge on cocaine appetite-suppressant action, the molecular basis of such fact remains unknown. Addiction/eating disorders (e.g., binge eating, anorexia, bulimia) share a central control involving reward circuits. However, we here show that the sigma-1 receptor (σ1R) mediates cocaine anorectic effects by interacting in neurons with growth/hormone/secretagogue (ghrelin) receptors. Cocaine increases colocalization of σ1R and GHS-R1a at the cell surface. Moreover, in transfected HEK-293T and neuroblastoma SH-SY5Y cells, and in primary neuronal cultures, pretreatment with cocaine or a σ1R agonist inhibited ghrelin-mediated signaling, in a similar manner as the GHS-R1a antagonist YIL-781. Results were similar in G protein-dependent (cAMP accumulation and calcium release) and in partly dependent or independent (ERK1/2 phosphorylation and label-free) assays. We provide solid evidence for direct interaction between receptors and the functional consequences, as well as a reliable structural model of the macromolecular σ1R-GHS-R1a complex, which arises as a key piece in the puzzle of the events linking cocaine consumption and appetitive/consummatory behaviors.
Collapse
Affiliation(s)
- David Aguinaga
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Mireia Medrano
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Arnau Cordomí
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Mireia Jiménez-Rosés
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Edgar Angelats
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Mireia Casanovas
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Ignacio Vega-Quiroga
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enric I Canela
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Milos Petrovic
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, UK
| | - Katia Gysling
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leonardo Pardo
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Rafael Franco
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain.
- School of Biology, Universitat de Barcelona, Diagonal 643, 08028, Barcelona, Spain.
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Department of Biochemistry and Physiology, Faculty of Pharmacy, Universitat de Barcelona, Barcelona, Spain.
- School of Biology, Universitat de Barcelona, Diagonal 643, 08028, Barcelona, Spain.
| |
Collapse
|
40
|
Sánchez-Blázquez P, Cortés-Montero E, Rodríguez-Muñoz M, Garzón J. Sigma 1 Receptor Antagonists Inhibit Manic-Like Behaviors in Two Congenital Strains of Mice. Int J Neuropsychopharmacol 2018; 21:938-948. [PMID: 29860313 PMCID: PMC6165958 DOI: 10.1093/ijnp/pyy049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 05/04/2018] [Accepted: 05/30/2018] [Indexed: 12/28/2022] Open
Abstract
Background Several currently available animal models reproduce select behavioral facets of human mania as well as the abnormal glutamatergic neurotransmission and dysregulation of glycogen synthase kinase 3β that accompanies this disease. Methods In this study, we addressed the therapeutic potential of ligands of sigma receptor type 1 (σ1R) in 2 putative models of mania: the "manic" Black Swiss outbred mice from Taconic farms (BStac) and mice with the 129 genetic background and histidine triad nucleotide-binding protein 1 (HINT1) deletion (HINT1-/- mice) that exhibit bipolar-like behaviors. Results The activity of control mice, which do not exhibit manic-like behaviors in the forced swim test, was significantly enhanced by MK801, an inhibitor of glutamate N-methyl-D-aspartate receptor activity, an effect that was not or barely observed in manic-like mice. Typical mood stabilizers, such as glycogen synthase kinase 3β inhibitors, but not σ1R ligands, reduced the N-methyl-D-aspartate receptor-mediated behaviors in control mice. Notably, σ1R antagonists S1RA, PD144418, BD1047, and BD1063, but not σ1R agonists PRE084 and PPCC, attenuated the manic-like behaviors of BStac and HINT1-/- mice by increasing antiactivity behaviors. The antimanic effects of a single administration of σ1R antagonists persisted for at least 24 hours, and these drugs did not alter the behavior of the "bipolar" HINT1-/- mice during pro-depressive episodes. Conclusions σ1R antagonists exhibit a selective normalizing effect on specific behavioral domains of mania without altering control (normal) or depressive-like behaviors.
Collapse
Affiliation(s)
- Pilar Sánchez-Blázquez
- Neuropharmacology, Department of Translational Neurosciences, Instituto Cajal, CSIC, Madrid, Spain
| | - Elsa Cortés-Montero
- Neuropharmacology, Department of Translational Neurosciences, Instituto Cajal, CSIC, Madrid, Spain
| | - María Rodríguez-Muñoz
- Neuropharmacology, Department of Translational Neurosciences, Instituto Cajal, CSIC, Madrid, Spain
| | - Javier Garzón
- Neuropharmacology, Department of Translational Neurosciences, Instituto Cajal, CSIC, Madrid, Spain
| |
Collapse
|
41
|
Liu DY, Chi TY, Ji XF, Liu P, Qi XX, Zhu L, Wang ZQ, Li L, Chen L, Zou LB. Sigma-1 receptor activation alleviates blood-brain barrier dysfunction in vascular dementia mice. Exp Neurol 2018; 308:90-99. [PMID: 30006137 DOI: 10.1016/j.expneurol.2018.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/02/2018] [Accepted: 07/02/2018] [Indexed: 10/28/2022]
Abstract
Sigma-1 receptor (Sig-1R) activation has been shown to decrease infarct volume and enhance neuronal survival after brain ischemia-reperfusion (IR) in rodent models. The present study aims to investigate first the effect of Sig-1R activation on blood-brain barrier (BBB) disruption during experimental stroke. Male C57BL/6 mice were subjected to bilateral common carotid artery occlusion (BCCAO) for 15 min, and the worst BBB leakage was observed on the 7th day after brain IR. To confirm the BBB protective role of Sig-1R, mice were divided into five groups (sham group, BCCAO group, PRE084 group, BD1047 group, PRE084 and BD1047 group; 29-35 mice for each group), and treated with agonist PRE084 (1 mg/kg) and/or antagonist BD1047 (1 mg/kg) for 7 days intraperitoneally once a day after BCCAO. Interestingly, PRE084 administration significantly improved neurobehavioral performance as well as healing of neuron damage and white matter lesions. PRE084 also reduced the leakage of Evans blue and IgG and attenuated the disassembly of BBB structural proteins, while the neuroprotective and BBB protective functions of PRE084 were blocked by BD1047. Furthermore, in Sig-1R knockout (Sig-1R KO) mice, brain IR produced more serious IgG leakage and degradation of BBB structural proteins than in wild-type model mice. In addition, the protective effect of PRE084 against the BBB was lost in Sig-1R KO mice after brain IR. Finally, treatment with PRE084 significantly increased the expression of Sig-1R in brain microvascular endothelial cells of mice that were subjected to brain IR and increased translocation of Sig-1R to the cell plasmalemma. Thus, we identified a previously unexplored role of Sig-1R in alleviating BBB disruption in stroke processes and have demonstrated that reversing BBB rupture through Sig-1R activation may be another promising method for cerebral protection against IR injury.
Collapse
Affiliation(s)
- Dan-Yang Liu
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tian-Yan Chi
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xue-Fei Ji
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Peng Liu
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiao-Xiao Qi
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lin Zhu
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zi-Qi Wang
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lin Li
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Ling Chen
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China.
| | - Li-Bo Zou
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
42
|
Rodríguez-Muñoz M, Onetti Y, Cortés-Montero E, Garzón J, Sánchez-Blázquez P. Cannabidiol enhances morphine antinociception, diminishes NMDA-mediated seizures and reduces stroke damage via the sigma 1 receptor. Mol Brain 2018; 11:51. [PMID: 30223868 PMCID: PMC6142691 DOI: 10.1186/s13041-018-0395-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/10/2018] [Indexed: 12/18/2022] Open
Abstract
Cannabidiol (CBD), the major non-psychotomimetic compound present in the Cannabis sativa plant, exhibits therapeutic potential for various human diseases, including chronic neurodegenerative diseases, such as Alzheimer's and Parkinson's, ischemic stroke, epilepsy and other convulsive syndromes, neuropsychiatric disorders, neuropathic allodynia and certain types of cancer. CBD does not bind directly to endocannabinoid receptors 1 and 2, and despite research efforts, its specific targets remain to be fully identified. Notably, sigma 1 receptor (σ1R) antagonists inhibit glutamate N-methyl-D-aspartate acid receptor (NMDAR) activity and display positive effects on most of the aforesaid diseases. Thus, we investigated the effects of CBD on three animal models in which NMDAR overactivity plays a critical role: opioid analgesia attenuation, NMDA-induced convulsive syndrome and ischemic stroke. In an in vitro assay, CBD disrupted the regulatory association of σ1R with the NR1 subunit of NMDAR, an effect shared by σ1R antagonists, such as BD1063 and progesterone, and prevented by σ1R agonists, such as 4-IBP, PPCC and PRE084. The in vivo administration of CBD or BD1063 enhanced morphine-evoked supraspinal antinociception, alleviated NMDA-induced convulsive syndrome, and reduced the infarct size caused by permanent unilateral middle cerebral artery occlusion. These positive effects of CBD were reduced by the σ1R agonists PRE084 and PPCC, and absent in σ1R-/- mice. Thus, CBD displays antagonist-like activity toward σ1R to reduce the negative effects of NMDAR overactivity in the abovementioned experimental situations.
Collapse
Affiliation(s)
- María Rodríguez-Muñoz
- Neuropharmacology. Department of Traslational Neuroscience, Cajal Institute, CSIC, E-28002 Madrid, Spain
| | - Yara Onetti
- Neuropharmacology. Department of Traslational Neuroscience, Cajal Institute, CSIC, E-28002 Madrid, Spain
| | - Elsa Cortés-Montero
- Neuropharmacology. Department of Traslational Neuroscience, Cajal Institute, CSIC, E-28002 Madrid, Spain
| | - Javier Garzón
- Neuropharmacology. Department of Traslational Neuroscience, Cajal Institute, CSIC, E-28002 Madrid, Spain
| | - Pilar Sánchez-Blázquez
- Neuropharmacology. Department of Traslational Neuroscience, Cajal Institute, CSIC, E-28002 Madrid, Spain
| |
Collapse
|
43
|
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
|
44
|
Rodríguez-Muñoz M, Sánchez-Blázquez P, Garzón J. Fenfluramine diminishes NMDA receptor-mediated seizures via its mixed activity at serotonin 5HT2A and type 1 sigma receptors. Oncotarget 2018; 9:23373-23389. [PMID: 29805740 PMCID: PMC5955088 DOI: 10.18632/oncotarget.25169] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/03/2018] [Indexed: 10/27/2022] Open
Abstract
Fenfluramine exhibits antiepileptic properties and thus diminishes epileptiform discharges in experimental animal models of Dravet syndrome. Fenfluramine is metabolized into norfenfluramine in vivo, which shows greater affinity and agonist activity at serotonin 5HT2 receptors (5HT2R) than fenfluramine. In this study, we found that fenfluramine and norfenfluramine disrupted the regulatory association of the sigma 1 receptor (σ1R) with NR1 subunits of glutamate N-methyl-D-aspartate receptors (NMDAR), an effect that was also produced by σ1R antagonists such as S1RA and prevented by σ1R agonists such as PPCC. The antagonists removed σ1R bound to NMDAR NR1 subunits enabling calcium-regulated calmodulin (CaM) to bind to those subunits. As a result, CaM may inhibit calcium permeation through NMDARs. The serotoninergic activity of fenfluramine at 5HT2AR, and likely also at 5HT2CR, collaborated with its activity at σ1Rs to prevent the convulsive syndrome promoted by NMDAR overactivation. Notably, fenfluramine enhanced the inhibitory coupling of G protein-coupled receptors such as 5HT1AR and cannabinoid type 1 receptor with NMDARs, thus allowing the more effective restrain of NMDAR activity. Thus, fenfluramine circumvents the negative side effects of direct NMDAR antagonists and may improve the quality of life of subjects affected by such proconvulsant dysfunctions.
Collapse
Affiliation(s)
- María Rodríguez-Muñoz
- Neuropharmacology, Department of Translational Neurosciences, Cajal Institute, CSIC, Madrid E-28002, Spain
| | - Pilar Sánchez-Blázquez
- Neuropharmacology, Department of Translational Neurosciences, Cajal Institute, CSIC, Madrid E-28002, Spain
| | - Javier Garzón
- Neuropharmacology, Department of Translational Neurosciences, Cajal Institute, CSIC, Madrid E-28002, Spain
| |
Collapse
|
45
|
Tejada MÁ, Montilla-García Á, González-Cano R, Bravo-Caparrós I, Ruiz-Cantero MC, Nieto FR, Cobos EJ. Targeting immune-driven opioid analgesia by sigma-1 receptors: Opening the door to novel perspectives for the analgesic use of sigma-1 antagonists. Pharmacol Res 2018; 131:224-230. [PMID: 29454675 DOI: 10.1016/j.phrs.2018.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 02/08/2018] [Indexed: 12/16/2022]
Abstract
Immune cells have a known role in pronociception, since they release a myriad of inflammatory algogens which interact with neurons to facilitate pain signaling. However, these cells also produce endogenous opioid peptides with analgesic potential. The sigma-1 receptor is a ligand-operated chaperone that modulates neurotransmission by interacting with multiple protein partners, including the μ-opioid receptor. We recently found that sigma-1 antagonists are able to induce opioid analgesia by enhancing the action of endogenous opioid peptides of immune origin during inflammation. This opioid analgesia is seen only at the inflamed site, where immune cells naturally accumulate. In this article we review the difficulties of targeting the opioid system for selective pain relief, and discuss the dual role of immune cells in pain and analgesia. Our discussion creates perspectives for possible novel therapeutic uses of sigma-1 antagonists as agents able to maximize the analgesic potential of the immune system.
Collapse
Affiliation(s)
- Miguel Á Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neurosciences, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Ángeles Montilla-García
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neurosciences, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Rafael González-Cano
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Inmaculada Bravo-Caparrós
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neurosciences, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - M Carmen Ruiz-Cantero
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neurosciences, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Francisco R Nieto
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neurosciences, 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
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neurosciences, 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.
| |
Collapse
|
46
|
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
|
47
|
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
|
48
|
Arena E, Dichiara M, Floresta G, Parenti C, Marrazzo A, Pittalà V, Amata E, Prezzavento O. Novel Sigma-1 receptor antagonists: from opioids to small molecules: what is new? Future Med Chem 2018; 10:231-256. [PMID: 29185346 DOI: 10.4155/fmc-2017-0164] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023] Open
Abstract
Sigma-1 (σ1) receptor has been identified as a chaperone protein that interacts with other proteins, such as N-methyl-D-aspartate (NMDA) and opioid receptors, modulating their activity. σ1 receptor antagonists have been developed to obtain useful compounds for the treatment of psychoses, pain, drug abuse and cancer. Some interesting compounds such as E-5842 (5) and MS-377 (24), haloperidol and piperazine derivatives, respectively, were endowed with high affinity for σ1 receptors (Ki σ1 = 4 and 73 nM; Ki σ2 = 220 and 6900, respectively). They were developed for the treatment of psychotic disorders and 5 also underwent Phase II clinical trials suggesting interesting potential therapeutic applications. Here, σ1 receptor antagonists have been grouped based on chemical structure and reviewed according to structure-activity relationship and potential therapeutic role.
Collapse
Affiliation(s)
- Emanuela Arena
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Maria Dichiara
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Giuseppe Floresta
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
- Department of Chemical Sciences, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Carmela Parenti
- Department of Drug Sciences, Pharmacology Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Valeria Pittalà
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Emanuele Amata
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| |
Collapse
|
49
|
Rodríguez-Muñoz M, Sánchez-Blázquez P, Callado LF, Meana JJ, Garzón-Niño J. Schizophrenia and depression, two poles of endocannabinoid system deregulation. Transl Psychiatry 2017; 7:1291. [PMID: 29249810 PMCID: PMC5802629 DOI: 10.1038/s41398-017-0029-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/04/2017] [Accepted: 09/07/2017] [Indexed: 02/06/2023] Open
Abstract
The activity of certain G protein-coupled receptors (GPCRs) and of glutamate N-Methyl-D-aspartate receptors (NMDARs) is altered in both schizophrenia and depression. Using postmortem prefrontal cortex samples from subjects with schizophrenia or depression, we observed a series of opposite changes in the expression of signaling proteins that have been implicated in the cross-talk between GPCRs and NMDARs. Thus, the levels of HINT1 proteins and NMDAR NR1 subunits carrying the C1 cytosolic segment were increased in depressives and decreased in schizophrenics, respect to matched controls. The differences in NR1 C1 subunits were compensated for via altered expression of NR1 subunits lacking the C1 segment; thus, the total number of NR1 subunits was comparable among the three groups. GPCRs influence the function of NR1 C1-containing NMDARs via PKC/Src, and thus, the association of mu-opioid and dopamine 2 receptors with NR1 C1 subunits was augmented in depressives and decreased in schizophrenics. However, the association of cannabinoid 1 receptors (CB1Rs) with NR1 C1 remained nearly constant. Endocannabinoids, via CB1Rs, control the presence of NR1 C1 subunits in the neural membrane. Thus, an altered endocannabinoid system may contribute to the pathophysiology of schizophrenia and depression by modifying the HINT1-NR1 C1/GPCR ratio, thereby altering GPCR-NMDAR cross-regulation.
Collapse
Affiliation(s)
- María Rodríguez-Muñoz
- 0000 0001 2177 5516grid.419043.bNeuropharmacology, Department of Translational Neurosciences, Instituto Cajal, CSIC, Madrid, E-28002 Spain
| | - Pilar Sánchez-Blázquez
- 0000 0001 2177 5516grid.419043.bNeuropharmacology, Department of Translational Neurosciences, Instituto Cajal, CSIC, Madrid, E-28002 Spain
| | - Luis F. Callado
- grid.452310.1Department of Pharmacology, University of the Basque Country UPV/EHU, BioCruces Health Research Institute, Barakaldo, Spain
| | - J. Javier Meana
- grid.452310.1Department of Pharmacology, University of the Basque Country UPV/EHU, BioCruces Health Research Institute, Barakaldo, Spain
| | - Javier Garzón-Niño
- Neuropharmacology, Department of Translational Neurosciences, Instituto Cajal, CSIC, Madrid, E-28002, Spain.
| |
Collapse
|
50
|
HINT1 in Neuropsychiatric Diseases: A Potential Neuroplastic Mediator. Neural Plast 2017; 2017:5181925. [PMID: 29214080 PMCID: PMC5682914 DOI: 10.1155/2017/5181925] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 08/23/2017] [Accepted: 09/18/2017] [Indexed: 01/25/2023] Open
Abstract
Although many studies have investigated the functions of histidine triad nucleotide-binding protein 1 (HINT1), its roles in neurobiological processes remain to be fully elucidated. As a member of the histidine triad (HIT) enzyme superfamily, HINT1 is distributed in almost every organ and has both enzymatic and nonenzymatic activity. Accumulating clinical and preclinical evidence suggests that HINT1 may play an important role as a neuroplastic mediator in neuropsychiatric diseases, such as schizophrenia, inherited peripheral neuropathies, mood disorders, and drug addiction. Though our knowledge of HINT1 is limited, it is believed that further research on the neuropathological functions of HINT1 would eventually benefit patients with neuropsychiatric and even psychosomatic diseases.
Collapse
|