1
|
Carrel A, Yiannakas A, Roukens JJ, Reynoso-Moreno I, Orsi M, Thakkar A, Arus-Pous J, Pellegata D, Gertsch J, Reymond JL. Exploring Simple Drug Scaffolds from the Generated Database Chemical Space Reveals a Chiral Bicyclic Azepane with Potent Neuropharmacology. J Med Chem 2025; 68:9176-9201. [PMID: 40274264 PMCID: PMC12067442 DOI: 10.1021/acs.jmedchem.4c02549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025]
Abstract
To assess how much structural diversity remains unexploited in simple drug scaffolds, we investigated ring systems functionalized with amine handles. Starting from the ring systems database GDB-4c, we enumerated 1139 possible amines and diamines with up to two five-, six-, or seven-membered rings. From the 680 cases not listed in PubChem, we synthesized several unprecedented cis- and trans-fused azepanes and tested possible targets predicted using the polypharmacology browser PPB2. From this screening campaign, an N-benzylated azepane emerged as a potent inhibitor of monoamine transporters with some selectivity toward norepinephrine (NET, SLC6A2) and dopamine transporter (DAT, SLC6A3) inhibition (IC50 < 100 nM) in combination with σ-1R inhibition (IC50 ≈ 110 nM). The in vitro profile, favorable pharmacokinetic properties, and preliminary behavioral and metabolomic effects in mice suggest a potential of N-benzylated bicyclic azepanes to target neuropsychiatric disorders. These experiments highlight the potential of simple but still unexplored scaffolds for drug discovery.
Collapse
Affiliation(s)
- Aline Carrel
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, Bern 3012, Switzerland
| | - Adonis Yiannakas
- Institute
of Biochemistry and Molecular Medicine, University of Bern, Gertrud-Woker Strasse 5, Bern 3012, Switzerland
| | - Jaap-Jan Roukens
- Institute
of Biochemistry and Molecular Medicine, University of Bern, Gertrud-Woker Strasse 5, Bern 3012, Switzerland
| | - Ines Reynoso-Moreno
- Institute
of Biochemistry and Molecular Medicine, University of Bern, Gertrud-Woker Strasse 5, Bern 3012, Switzerland
| | - Markus Orsi
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, Bern 3012, Switzerland
| | - Amol Thakkar
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, Bern 3012, Switzerland
| | - Josep Arus-Pous
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, Bern 3012, Switzerland
| | - Daniele Pellegata
- Institute
of Biochemistry and Molecular Medicine, University of Bern, Gertrud-Woker Strasse 5, Bern 3012, Switzerland
| | - Jürg Gertsch
- Institute
of Biochemistry and Molecular Medicine, University of Bern, Gertrud-Woker Strasse 5, Bern 3012, Switzerland
| | - Jean-Louis Reymond
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, Bern 3012, Switzerland
| |
Collapse
|
2
|
Shokr MM, Badawi GA, Elshazly SM, Zaki HF, Mohamed AF. Sigma 1 Receptor and Its Pivotal Role in Neurological Disorders. ACS Pharmacol Transl Sci 2025; 8:47-65. [PMID: 39816800 PMCID: PMC11729429 DOI: 10.1021/acsptsci.4c00564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
Sigma 1 receptor (S1R) is a multifunctional, ligand-activated protein located in the membranes of the endoplasmic reticulum (ER). It mediates a variety of neurological disorders, including epilepsy, amyotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease. The wide neuroprotective effects of S1R agonists are achieved by a variety of pro-survival and antiapoptotic S1R-mediated signaling functions. Nonetheless, relatively little is known about the specific molecular mechanisms underlying S1R activity. Many studies on S1R protein have highlighted the importance of maintaining normal cellular homeostasis through its control of calcium and lipid exchange between the ER and mitochondria, ER-stress response, and many other mechanisms. In this review, we will discuss S1R different cellular localization and explain S1R-associated biological activity, such as its localization in the ER-plasma membrane and Mitochondrion-Associated ER Membrane interfaces. While outlining the cellular mechanisms and important binding partners involved in these processes, we also explained how the dysregulation of these pathways contributes to neurodegenerative disorders.
Collapse
Affiliation(s)
- Mustafa M. Shokr
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University−Arish Branch, Arish, 45511, Egypt
| | - Ghada A. Badawi
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University−Arish Branch, Arish, 45511, Egypt
| | - Shimaa M. Elshazly
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Hala F. Zaki
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed F. Mohamed
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Faculty
of Pharmacy, King Salman International University
(KSIU), South Sinai 46612, Egypt
| |
Collapse
|
3
|
Soldatov V, Venediktov A, Belykh A, Piavchenko G, Naimzada MD, Ogneva N, Kartashkina N, Bushueva O. Chaperones vs. oxidative stress in the pathobiology of ischemic stroke. Front Mol Neurosci 2024; 17:1513084. [PMID: 39723236 PMCID: PMC11668803 DOI: 10.3389/fnmol.2024.1513084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
As many proteins prioritize functionality over constancy of structure, a proteome is the shortest stave in the Liebig's barrel of cell sustainability. In this regard, both prokaryotes and eukaryotes possess abundant machinery supporting the quality of the proteome in healthy and stressful conditions. This machinery, namely chaperones, assists in folding, refolding, and the utilization of client proteins. The functions of chaperones are especially important for brain cells, which are highly sophisticated in terms of structural and functional organization. Molecular chaperones are known to exert beneficial effects in many brain diseases including one of the most threatening and widespread brain pathologies, ischemic stroke. However, whether and how they exert the antioxidant defense in stroke remains unclear. Herein, we discuss the chaperones shown to fight oxidative stress and the mechanisms of their antioxidant action. In ischemic stroke, during intense production of free radicals, molecular chaperones preserve the proteome by interacting with oxidized proteins, regulating imbalanced mitochondrial function, and directly fighting oxidative stress. For instance, cells recruit Hsp60 and Hsp70 to provide proper folding of newly synthesized proteins-these factors are required for early ischemic response and to refold damaged polypeptides. Additionally, Hsp70 upregulates some dedicated antioxidant pathways such as FOXO3 signaling. Small HSPs decrease oxidative stress via attenuation of mitochondrial function through their involvement in the regulation of Nrf- (Hsp22), Akt and Hippo (Hsp27) signaling pathways as well as mitophagy (Hsp27, Hsp22). A similar function has also been proposed for the Sigma-1 receptor, contributing to the regulation of mitochondrial function. Some chaperones can prevent excessive formation of reactive oxygen species whereas Hsp90 is suggested to be responsible for pro-oxidant effects in ischemic stroke. Finally, heat-resistant obscure proteins (Hero) are able to shield client proteins, thus preventing their possible over oxidation.
Collapse
Affiliation(s)
- Vladislav Soldatov
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| | - Artem Venediktov
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Andrei Belykh
- Pathophysiology Department, Kursk State Medical University, Kursk, Russia
- Research Institute of General Pathology, Kursk State Medical University, Kursk, Russia
| | - Gennadii Piavchenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Mukhammad David Naimzada
- Research Institute of Experimental Medicine, Kursk State Medical University, Kursk, Russia
- Laboratory of Public Health Indicators Analysis and Health Digitalization, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Nastasya Ogneva
- Scientific Center of Biomedical Technologies, Federal Medical and Biological Agency of Russia, Moscow, Russia
| | - Natalia Kartashkina
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Olga Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russia
| |
Collapse
|
4
|
Liu C, Chen IS, Barri M, Murrell-Lagnado R, Kubo Y. Structural determinants of M2R involved in inhibition by Sigma-1R. J Biol Chem 2024; 300:108006. [PMID: 39551139 DOI: 10.1016/j.jbc.2024.108006] [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: 05/04/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024] Open
Abstract
Sigma-1 receptor (S1R) is a multimodal chaperone protein that is implicated in various pathophysiological conditions including drug addiction, Alzheimer's disease, and amyotrophic lateral sclerosis (ALS). S1R interacts with various ion channels and receptors on the endoplasmic reticulum or plasma membrane (PM). It has been reported that S1R colocalizes with the M2-muscarinic acetylcholine receptor (M2R) on the soma of motoneurons, although a functional interaction between these two proteins has not been established. Here, we investigated the regulation of M2R signaling by S1R using electrophysiological recordings of GIRK currents in HEK293T cells. We observed that S1R strongly inhibited M2R-mediated activation of GIRK1/2, but the disease mutant linked to ALS, S1R E102Q, did not. The inhibitory effect of S1R was selective for M2R and wasn't seen when S1R was co-expressed with other Gi/o coupled receptors including M4R. Chimeric and mutant receptors of M2R and M4R were generated and analyzed, and this highlighted Ala401 in the transmembrane 6 domain (TM6) of M2R and Glu172 as well as Glu175 in the extracellular loop 2 regions of M2R, as essential for the inhibition by S1R. Co-immunoprecipitation confirmed the physical interaction between M2R and S1R. Immunocytochemical labeling of M2R and S1R expressed in HeLa cells, HEK293T cells, and cultured hippocampal neurons, showed clear PM expression of M2R throughout the cell which was decreased by coexpression with S1R but was still apparent. Taken together, our results show that S1R interacts with M2R to reduce both its PM expression and function, and this involves TM6 and the extracellular loop 2.
Collapse
Affiliation(s)
- Chang Liu
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Program of Physiological Sciences, Field of Life Science, Department of Advanced Studies, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan.
| | - I-Shan Chen
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Program of Physiological Sciences, Field of Life Science, Department of Advanced Studies, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan; Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Muruj Barri
- School of Life Sciences, University of Sussex, Brighton, UK
| | | | - Yoshihiro Kubo
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Program of Physiological Sciences, Field of Life Science, Department of Advanced Studies, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan.
| |
Collapse
|
5
|
Xu ZZ, Zhou J, Duan K, Li XT, Chang S, Huang W, Lu Q, Tao J, Xie WB. Blocking Sigmar1 exacerbates methamphetamine-induced hypertension. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167284. [PMID: 38851304 DOI: 10.1016/j.bbadis.2024.167284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/06/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
AIM Methamphetamine (METH) chronic exposure is an important risk factor for hypertension development. However, the mechanisms behind METH-induced hypertension remain unclear. Therefore, we aimed to reveal the potential mechanisms underlying METH-induced hypertension. METHODS AND RESULTS We structured the mouse hypertension model by METH, and observed that METH-treated mice have presented vascular remodeling (large-and small-size arteries) with collagen deposit around the vessel and increasing blood pressure (BP) and Sigma1 receptor (Sigmar1) in vascular tissue. We hypothesized that Sigmar1 is crucial in METH-induced hypertension and vascular remodeling. Sigmar1 knockout (KO) mice and antagonist (BD1047) pretreated mice exposed to METH for six-week showed higher BP and more collagen deposited around vessels than wild-type (WT) mice exposed to METH for six-week, in contrast, mice pretreated with Sigmar1 agonist (PRE-084) had unchanged BP and perivascular collagen despite the six-week METH exposure. Furthermore, we found that METH exposure induced vascular smooth muscle cells (VSMCs) and mesenchymal stem cells to differentiate into the myofibroblast-like cell and secrete collagen into surrounding vessels. Mechanically, Sigmar1 can suppress the COL1A1 expression by blocking the classical fibrotic TGF-β/Smad2/3 signaling pathway in METH-exposed VSMCs and mesenchymal stem cells. CONCLUSION Our results suggest that Sigmar1 is involved in METH-induced hypertension and vascular fibrosis by blocking the activation of the TGF-β/Smad2/3 signaling pathway. Accordingly, Sigmar1 may be a novel therapeutic target for METH-induced hypertension and vascular fibrosis.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Blood Pressure/drug effects
- Collagen/metabolism
- Disease Models, Animal
- Hypertension/chemically induced
- Hypertension/metabolism
- Hypertension/pathology
- Hypertension/genetics
- Mesenchymal Stem Cells/metabolism
- Methamphetamine/adverse effects
- Methamphetamine/toxicity
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Receptors, sigma/metabolism
- Receptors, sigma/genetics
- Sigma-1 Receptor
- Signal Transduction/drug effects
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Zhen-Zhen Xu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Jie Zhou
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Ke Duan
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Xiao-Ting Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Sheng Chang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Wanshan Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Qiujun Lu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Jing Tao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Wei-Bing Xie
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
6
|
Fu C, Xiao Y, Zhou X, Sun Z. Insight into binding of endogenous neurosteroid ligands to the sigma-1 receptor. Nat Commun 2024; 15:5619. [PMID: 38965213 PMCID: PMC11224282 DOI: 10.1038/s41467-024-49894-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
The sigma-1 receptor (σ1R) is a non-opioid membrane receptor, which responds to a diverse array of synthetic ligands to exert various pharmacological effects. Meanwhile, candidates for endogenous ligands of σ1R have also been identified. However, how endogenous ligands bind to σ1R remains unknown. Here, we present crystal structures of σ1R from Xenopus laevis (xlσ1R) bound to two endogenous neurosteroid ligands, progesterone (a putative antagonist) and dehydroepiandrosterone sulfate (DHEAS) (a putative agonist), at 2.15-3.09 Å resolutions. Both neurosteroids bind to a similar location in xlσ1R mainly through hydrophobic interactions, but surprisingly, with opposite binding orientations. DHEAS also forms hydrogen bonds with xlσ1R, whereas progesterone interacts indirectly with the receptor through water molecules near the binding site. Binding analyses are consistent with the xlσ1R-neurosteroid complex structures. Furthermore, molecular dynamics simulations and structural data reveal a potential water entry pathway. Our results provide insight into binding of two endogenous neurosteroid ligands to σ1R.
Collapse
Affiliation(s)
- Chunting Fu
- Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Xiao
- Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoming Zhou
- Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Ziyi Sun
- Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
7
|
Barbaraci C, di Giacomo V, Maruca A, Patamia V, Rocca R, Dichiara M, Di Rienzo A, Cacciatore I, Cataldi A, Balaha M, Rapino M, Zagni C, Zampieri D, Pasquinucci L, Parenti C, Amata E, Rescifina A, Alcaro S, Marrazzo A. Discovery of first novel sigma/HDACi dual-ligands with a potent in vitro antiproliferative activity. Bioorg Chem 2023; 140:106794. [PMID: 37659146 DOI: 10.1016/j.bioorg.2023.106794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 09/04/2023]
Abstract
Designing and discovering compounds for dual-target inhibitors is challenging to synthesize new, safer, and more efficient drugs than single-target drugs, especially to treat multifactorial diseases such as cancer. The simultaneous regulation of multiple targets might represent an alternative synthetic approach to optimize patient compliance and tolerance, minimizing the risk of target-based drug resistance due to the modulation of a few targets. To this end, we conceived for the first time the design and synthesis of dual-ligands σR/HDACi to evaluate possible employment as innovative candidates to address this complex disease. Among all synthesized compounds screened for several tumoral cell lines, compound 6 (Kiσ1R = 38 ± 3.7; Kiσ2R = 2917 ± 769 and HDACs IC50 = 0.59 µM) is the most promising candidate as an antiproliferative agent with an IC50 of 0.9 µM on the HCT116 cell line and no significant toxicity to normal cells. Studies of molecular docking, which confirmed the affinity over σ1R and a pan-HDACs inhibitory behavior, support a possible balanced affinity and activity between both targets.
Collapse
Affiliation(s)
- Carla Barbaraci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Viviana di Giacomo
- Department of Pharmacy, University "G. d'Annunzio", Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Annalisa Maruca
- Net4science academic spinoff srl, Università degli Studi "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Viale Europa, 88100, Catanzaro, Italy
| | - Vincenzo Patamia
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Roberta Rocca
- Net4science academic spinoff srl, Università degli Studi "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Viale Europa, 88100, Catanzaro, Italy
| | - Maria Dichiara
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Annalisa Di Rienzo
- Department of Pharmacy, University "G. d'Annunzio", Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Ivana Cacciatore
- Department of Pharmacy, University "G. d'Annunzio", Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Amelia Cataldi
- Department of Pharmacy, University "G. d'Annunzio", Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Marwa Balaha
- Department of Pharmacy, University "G. d'Annunzio", Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafr El Sheikh 33516, Egypt
| | - Monica Rapino
- Genetic Molecular Institute of CNR, Unit of Chieti, "G. d' Annunzio" University, Via dei Vestini 31, 66100 Chieti-Pescara, Italy
| | - Chiara Zagni
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Daniele Zampieri
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Giorgieri 1, 34127 Trieste, Italy
| | - Lorella Pasquinucci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Carmela Parenti
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Emanuele Amata
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Antonio Rescifina
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Stefano Alcaro
- Net4science academic spinoff srl, Università degli Studi "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Viale Europa, 88100, Catanzaro, Italy; Dipartimento di Scienze della Salute, Università degli Studi "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Viale Europa, 88100, Catanzaro, Italy.
| | - Agostino Marrazzo
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy.
| |
Collapse
|
8
|
Robinson TS, Osman MA. An Emerging Role for Sigma Receptor 1 in Personalized Treatment of Breast Cancer. Cancers (Basel) 2023; 15:3464. [PMID: 37444574 PMCID: PMC10340381 DOI: 10.3390/cancers15133464] [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: 05/07/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
Despite the major progress in treating breast cancer, recurrence remains a problem and types such as triple-negative breast cancer still lack targeted medicine. The orphan Sigma receptor1 (SigmaR1) has emerged as a target in breast cancer, but its mechanism of action is unclear and hinders clinical utility. SigmaR1 is widely expressed in organ tissues and localized to various sub-cellular compartments, particularly the endoplasmic reticulum (ER), the mitochondrial-associated membranes (MAMs) and the nuclear envelope. As such, it involves diverse cellular functions, including protein quality control/ER stress, calcium signaling, cholesterol homeostasis, mitochondrial integrity and energy metabolism. Consequently, SigmaR1 has been implicated in a number of cancers and degenerative diseases and thus has been intensively pursued as a therapeutic target. Because SigmaR1 binds a number of structurally unrelated ligands, it presents an excellent context-dependent therapeutic target. Here, we review its role in breast cancer and the current therapies that have been considered based on its known functions. As SigmaR1 is not classified as an oncoprotein, we propose a model in which it serves as an oligomerization adaptor in key cellular pathways, which may help illuminate its association with variable diseases and pave the way for clinical utility in personalized medicine.
Collapse
Affiliation(s)
| | - Mahasin A. Osman
- Department of Medicine, Division of Oncology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA;
| |
Collapse
|
9
|
Zhao F, Yang T, Zhou L, Li R, Liu J, Zhao J, Jia R. Sig1R activates extracellular matrix-induced bladder cancer cell proliferation and angiogenesis by combing β-integrin. Aging (Albany NY) 2023; 15:204721. [PMID: 37199665 DOI: 10.18632/aging.204721] [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: 10/25/2022] [Accepted: 04/15/2023] [Indexed: 05/19/2023]
Abstract
The extracellular matrix (ECM) regulates many biological functions involved in tumorigenesis and tumor development; however, the underlying mechanism remains unknown. Sigma 1 receptor (Sig1R), a stress-activated chaperone, regulates the crosstalk between the ECM and tumor cells and is related to the malignant characteristics of several tumors. However, the link between Sig1R overexpression and ECM during malignancy has not been established in bladder cancer (BC). Here, we analyzed the interaction of Sig1R and β-integrin in BC cells and its role in ECM-mediated cell proliferation and angiogenesis. We found that Sig1R forms a complex with β-integrin to promote ECM-mediated BC cell proliferation and angiogenesis, which enhances the aggressiveness of the tumor cells. This leads to poor survival. Our research revealed that Sig1R mediates the cross-talk between BC cells and their ECM microenvironment, thereby driving the progression of BC. Promisingly, targeting an ion channel function through Sig1R inhibition may serve as a potential approach for BC treatment.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Tianli Yang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Rongfei Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jun Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
10
|
Morató X, Fernández-Dueñas V, Pérez-Villamor P, Valle-León M, Vela JM, Merlos M, Burgueño J, Ciruela F. Development of a Novel σ 1 Receptor Biosensor Based on Its Heterodimerization with Binding Immunoglobulin Protein in Living Cells. ACS Chem Neurosci 2023. [PMID: 37191585 DOI: 10.1021/acschemneuro.3c00206] [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: 05/17/2023] Open
Abstract
The σ1 receptor (S1R) is a ligand-regulated non-opioid intracellular receptor involved in several pathological conditions. The development of S1R-based drugs as therapeutic agents is a challenge due to the lack of simple functional assays to identify and classify S1R ligands. We have developed a novel nanoluciferase binary technology (NanoBiT) assay based on the ability of S1R to heteromerize with the binding immunoglobulin protein (BiP) in living cells. The S1R-BiP heterodimerization biosensor allows for rapid and accurate identification of S1R ligands by monitoring the dynamics of association-dissociation of S1R and BiP. Acute treatment of cells with the S1R agonist PRE-084 produced rapid and transient dissociation of the S1R-BiP heterodimer, which was blocked by haloperidol. The effect of PRE-084 was enhanced by calcium depletion, leading to a higher reduction in heterodimerization even in the presence of haloperidol. Prolonged incubation of cells with S1R antagonists (haloperidol, NE-100, BD-1047, and PD-144418) increased the formation of S1R-BiP heteromers, while agonists (PRE-084, 4-IBP, and pentazocine) did not alter heterodimerization under the same experimental conditions. The newly developed S1R-BiP biosensor is a simple and effective tool for exploring S1R pharmacology in an easy cellular setting. This biosensor is suitable for high-throughput applications and a valuable resource in the researcher's toolkit.
Collapse
Affiliation(s)
- Xavier Morató
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, 08908 L'Hospitalet de Llobregat, Spain
| | - Víctor Fernández-Dueñas
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, 08908 L'Hospitalet de Llobregat, Spain
| | | | - Marta Valle-León
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, 08908 L'Hospitalet de Llobregat, Spain
| | - José Miguel Vela
- Welab Barcelona, Parc Científic Barcelona, 08028 Barcelona, Spain
| | - Manuel Merlos
- Welab Barcelona, Parc Científic Barcelona, 08028 Barcelona, Spain
| | - Javier Burgueño
- Welab Barcelona, Parc Científic Barcelona, 08028 Barcelona, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, 08908 L'Hospitalet de Llobregat, Spain
| |
Collapse
|
11
|
Cai L, Liow JS, Morse CL, Telu S, Davies R, Manly LS, Zoghbi SS, Chin FT, Innis RB, Pike VW. Candidate 3-benzazepine-1-ol type GluN2B receptor radioligands ( 11C-NR2B-Me enantiomers) have high binding in cerebellum but not to σ1 receptors. EJNMMI Res 2023; 13:28. [PMID: 37017827 PMCID: PMC10076467 DOI: 10.1186/s13550-023-00975-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/12/2023] [Indexed: 04/06/2023] Open
Abstract
INTRODUCTION We recently reported 11C-NR2B-SMe ([S-methyl-11C](R,S)-7-thiomethoxy-3-(4-(4-methyl-phenyl)butyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-ol) and its enantiomers as candidate radioligands for imaging the GluN2B subunit within rat N-methyl-D-aspartate receptors. However, these radioligands gave unexpectedly high and displaceable binding in rat cerebellum, possibly due to cross-reactivity with sigma-1 (σ1) receptors. This study investigated 11C-labeled enantiomers of a close analogue (7-methoxy-3-(4-(p-tolyl)butyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-ol; NR2B-Me) of 11C-NR2B-SMe as new candidate GluN2B radioligands. PET was used to evaluate these radioligands in rats and to assess potential cross-reactivity to σ1 receptors. METHODS NR2B-Me was assayed for binding affinity and selectivity to GluN2B in vitro. 11C-NR2B-Me and its enantiomers were prepared by Pd-mediated treatment of boronic ester precursors with 11C-iodomethane. Brain PET scans were conducted after radioligand intravenous injection into rats. Various ligands for GluN2B receptors or σ1 receptors were administered at set doses in pre-blocking or displacement experiments to assess their impact on imaging data. 18F-FTC146 and enantiomers of 11C-NR2B-SMe were used for comparison. Radiometabolites from brain and plasma were measured ex vivo and in vitro. RESULTS NR2B-Me enantiomers showed high GluN2B affinity and selectivity in vitro. 11C-NR2B-Me enantiomers gave high early whole rat brain uptake of radioactivity, including high uptake in cerebellum, followed by slower decline. Radioactivity in brain at 30 min ex vivo was virtually all unchanged radioligand. Only less lipophilic radiometabolites appeared in plasma. When 11C-(R)-NR2B-Me was used, three high-affinity GluN2B ligands-NR2B-SMe, Ro25-6981, and CO101,244-showed increasing pre-block of whole brain radioactivity retention with increasing dose. Two σ1 receptor antagonists, FTC146 and BD1407, were ineffective pre-blocking agents. Together, these results strongly resemble those obtained with 11C-NR2B-SMe enantiomers, except that 11C-NR2B-Me enantiomers showed faster reversibility of binding. When 18F-FTC146 was used as a radioligand, FTC146 and BD1407 showed strong pre-blocking effects whereas GluN2B ligands showed only weak blocking effects. CONCLUSION 11C-NR2B-Me enantiomers showed specific binding to GluN2B receptors in rat brain in vivo. High unexpected specific binding in cerebellum was not due to σ1 receptors. Additional investigation is needed to identify the source of the high specific binding.
Collapse
Affiliation(s)
- Lisheng Cai
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA.
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Riley Davies
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Lester S Manly
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Frederick T Chin
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 1201 Welch Road, Rm. PS049, Stanford, CA, 94305-584, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| |
Collapse
|
12
|
Rapetti-Mauss R, Nigri J, Berenguier C, Finetti P, Tubiana SS, Labrum B, Allegrini B, Pellissier B, Efthymiou G, Hussain Z, Bousquet C, Dusetti N, Bertucci F, Guizouarn H, Melnyk P, Borgese F, Tomasini R, Soriani O. SK2 channels set a signalling hub bolstering CAF-triggered tumourigenic processes in pancreatic cancer. Gut 2023; 72:722-735. [PMID: 36882214 DOI: 10.1136/gutjnl-2021-326610] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 08/12/2022] [Indexed: 03/09/2023]
Abstract
OBJECTIVE Intercellular communication within pancreatic ductal adenocarcinoma (PDAC) dramatically contributes to metastatic processes. The underlying mechanisms are poorly understood, resulting in a lack of targeted therapy to counteract stromal-induced cancer cell aggressiveness. Here, we investigated whether ion channels, which remain understudied in cancer biology, contribute to intercellular communication in PDAC. DESIGN We evaluated the effects of conditioned media from patient-derived cancer-associated fibroblasts (CAFs) on electrical features of pancreatic cancer cells (PCC). The molecular mechanisms were deciphered using a combination of electrophysiology, bioinformatics, molecular and biochemistry techniques in cell lines and human samples. An orthotropic mouse model where CAF and PCC were co-injected was used to evaluate tumour growth and metastasis dissemination. Pharmacological studies were carried out in the Pdx1-Cre, Ink4afl/fl LSL-KrasG12D (KICpdx1) mouse model. RESULTS We report that the K+ channel SK2 expressed in PCC is stimulated by CAF-secreted cues (8.84 vs 2.49 pA/pF) promoting the phosphorylation of the channel through an integrin-epidermal growth factor receptor (EGFR)-AKT (Protein kinase B) axis. SK2 stimulation sets a positive feedback on the signalling pathway, increasing invasiveness in vitro (threefold) and metastasis formation in vivo. The CAF-dependent formation of the signalling hub associating SK2 and AKT requires the sigma-1 receptor chaperone. The pharmacological targeting of Sig-1R abolished CAF-induced activation of SK2, reduced tumour progression and extended the overall survival in mice (11.7 weeks vs 9.5 weeks). CONCLUSION We establish a new paradigm in which an ion channel shifts the activation level of a signalling pathway in response to stromal cues, opening a new therapeutic window targeting the formation of ion channel-dependent signalling hubs.
Collapse
Affiliation(s)
| | - Jérémy Nigri
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS UMR7258, Université Aix-Marseille, Marseille, France
| | | | - Pascal Finetti
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS UMR7258, Université Aix-Marseille, Marseille, France
| | - Sarah Simha Tubiana
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS UMR7258, Université Aix-Marseille, Marseille, France
| | - Bonnie Labrum
- Université Côte d'azur, CNRS, Inserm, iBV, Nice, France
| | | | | | - Georgios Efthymiou
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS UMR7258, Université Aix-Marseille, Marseille, France
| | - Zainab Hussain
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS UMR7258, Université Aix-Marseille, Marseille, France
| | - Corinne Bousquet
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM Unité Mixte de Recherche UMR-1037, CNRS Equipe de Recherche Labellisée ERL5294, Equipe de Recherche Labellisée "Ligue Contre le Cancer" & "LabEx Toucan", Université de Toulouse, Toulouse, France
| | - Nelson Dusetti
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS UMR7258, Université Aix-Marseille, Marseille, France
| | - François Bertucci
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS UMR7258, Université Aix-Marseille, Marseille, France
| | | | - Patricia Melnyk
- Lille Neuroscience and Cognition Research Center UMR-S 1172, University of Lille, INSERM, CHU Lille, Lille, France
| | | | - Richard Tomasini
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS UMR7258, Université Aix-Marseille, Marseille, France
| | | |
Collapse
|
13
|
Pergolizzi J, Varrassi G, Coleman M, Breve F, Christo DK, Christo PJ, Moussa C. The Sigma Enigma: A Narrative Review of Sigma Receptors. Cureus 2023; 15:e35756. [PMID: 37020478 PMCID: PMC10069457 DOI: 10.7759/cureus.35756] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/04/2023] [Indexed: 03/07/2023] Open
Abstract
The sigma-1 and sigma-2 receptors were first discovered in the 1960s and were thought to be a form of opioid receptors initially. Over time, more was gradually learned about these receptors, which are actually protein chaperones, and many of their unique or unusual properties can contribute to a range of important new therapeutic applications. These sigma receptors translocate in the body and regulate calcium homeostasis and mitochondrial bioenergetics and they also have neuroprotective effects. The ligands to which these sigma receptors respond are several and dissimilar, including neurosteroids, neuroleptics, and cocaine. There is controversy as to their endogenous ligands. Sigma receptors are also involved in the complex processes of cholesterol homeostasis and protein folding. While previous work on this topic has been limited, research has been conducted in multiple disease states, such as addiction, aging. Alzheimer's disease, cancer, psychiatric disorders, pain and neuropathic pain, Parkinson's disease, and others. There is currently increasing interest in sigma-1 and sigma-2 receptors as they provide potential therapeutic targets for many disease indications.
Collapse
|
14
|
Li J, Satyshur KA, Guo LW, Ruoho AE. Sphingoid Bases Regulate the Sigma-1 Receptor-Sphingosine and N, N'-Dimethylsphingosine Are Endogenous Agonists. Int J Mol Sci 2023; 24:3103. [PMID: 36834510 PMCID: PMC9962145 DOI: 10.3390/ijms24043103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Both bioactive sphingolipids and Sigma-1 receptor (S1R) chaperones occur ubiquitously in mammalian cell membranes. Endogenous compounds that regulate the S1R are important for controlling S1R responses to cellular stress. Herein, we interrogated the S1R in intact Retinal Pigment Epithelial cells (ARPE-19) with the bioactive sphingoid base, sphingosine (SPH), or the pain-provoking dimethylated SPH derivative, N,N'-dimethylsphingosine (DMS). As informed by a modified native gel approach, the basal and antagonist (BD-1047)-stabilized S1R oligomers dissociated to protomeric forms in the presence of SPH or DMS (PRE-084 as control). We, thus, posited that SPH and DMS are endogenous S1R agonists. Consistently, in silico docking of SPH and DMS to the S1R protomer showed strong associations with Asp126 and Glu172 in the cupin beta barrel and extensive van der Waals interactions of the C18 alkyl chains with the binding site including residues in helices 4 and 5. Mean docking free energies were 8.73-8.93 kcal/mol for SPH and 8.56-8.15 kcal/mol for DMS, and calculated binding constants were ~40 nM for SPH and ~120 nM for DMS. We hypothesize that SPH, DMS, and similar sphingoid bases access the S1R beta barrel via a membrane bilayer pathway. We further propose that the enzymatic control of ceramide concentrations in intracellular membranes as the primary sources of SPH dictates availability of endogenous SPH and DMS to the S1R and the subsequent control of S1R activity within the same cell and/or in cellular environments.
Collapse
Affiliation(s)
- Jing Li
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Kenneth A. Satyshur
- Small Molecule Screening Facility, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
| | - Lian-Wang Guo
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Arnold E. Ruoho
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
| |
Collapse
|
15
|
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
|
16
|
Sigma-1 receptor agonist PRE-084 confers protection against TAR DNA-binding protein-43 toxicity through NRF2 signalling. Redox Biol 2022; 58:102542. [PMID: 36442393 PMCID: PMC9706169 DOI: 10.1016/j.redox.2022.102542] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting upper and lower motor neurons. As a consequence, ALS patients display a locomotor disorder related to muscle weakness and progressive paralysis. Pathological mechanisms that participate in ALS involve deficient unfolded protein response, mitochondrial dysfunction and oxidative stress, among others. Finding a therapeutic target to break the vicious circle is particularly challenging. Sigma-1 receptor (S1R) is an endoplasmic reticulum (ER) chaperone that may be one of those targets. We here address and decipher the efficiency of S1R activation on a key ALS gene, TDP43, in zebrafish vertebrate model. While expression of mutant TDP43 (TDP43G348C) led to locomotor defects, treatment with the reference S1R agonist PRE-084 rescued motor performances in a zebrafish model. Treatment with the agonist ameliorated maximal mitochondrial respiration in the TDP43 context. We observed that TDP43G348C exacerbated ER stress induced by tunicamycin, resulting in increased levels of ER stress chaperone BiP and pro-apoptotic factor CHOP. Importantly, PRE-084 treatment in the same condition further heightened BiP levels but also EIF2α/ATF4 and NRF2 signalling cascades, both known to promote antioxidant protection during ER stress. Moreover, we showed that increasing NRF2 levels directly or by sulforaphane treatment rescued locomotor defects of TDP43G348C zebrafish. For the first time, we here provide the proof of concept that PRE-084 prevents mutant TDP43 toxicity by boosting ER stress response and antioxidant cascade through NRF2 signalling.
Collapse
|
17
|
Wang YM, Xia CY, Jia HM, He J, Lian WW, Yan Y, Wang WP, Zhang WK, Xu JK. Sigma-1 receptor: A potential target for the development of antidepressants. Neurochem Int 2022; 159:105390. [PMID: 35810915 DOI: 10.1016/j.neuint.2022.105390] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/10/2022] [Accepted: 07/05/2022] [Indexed: 10/17/2022]
Abstract
Though a great many of studies on the development of antidepressants for the therapy of major depression disorder (MDD) and the development of antidepressants have been carried out, there still lacks an efficient approach in clinical practice. The involvement of Sigma-1 receptor in the pathological process of MDD has been verified. In this review, recent research focusing on the role of Sigma-1 receptor in the etiology of MDD were summarized. Preclinical studies and clinical trials have found that stress induce the variation of Sigma-1 receptor in the blood, brain and heart. Dysfunction and absence of Sigma-1 receptor result in depressive-like behaviors in rodent animals. Agonists of Sigma-1 receptor show not only antidepressant-like activities but also therapeutical effects in complications of depression. The mechanisms underlying antidepressant-like effects of Sigma-1 receptor may include suppressing neuroinflammation, regulating neurotransmitters, ameliorating brain-derived neurotrophic factor and N-Methyl-D-Aspartate receptor, and alleviating the endoplasmic reticulum stress and mitochondria damage during stress. Therefore, Sigma-1 receptor represents a potential target for antidepressants development.
Collapse
Affiliation(s)
- Yu-Ming Wang
- School of Life Sciences & School of Chinese Medicine Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China; Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Cong-Yuan Xia
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Hong-Mei Jia
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, PR China
| | - Jun He
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wen-Wen Lian
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Yu Yan
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wen-Ping Wang
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wei-Ku Zhang
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China.
| | - Jie-Kun Xu
- School of Life Sciences & School of Chinese Medicine Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| |
Collapse
|
18
|
Huang L, Xiao H, Xie X, Hu F, Tang F, Smith SB, Gan L. Generation of Sigmar1 conditional knockout mouse using CRISPR-Cas9 gene targeting. Genesis 2022; 60:e23487. [PMID: 35633570 DOI: 10.1002/dvg.23487] [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: 12/17/2021] [Revised: 04/01/2022] [Accepted: 04/25/2022] [Indexed: 11/08/2022]
Abstract
The Sigma 1 receptor (SIGMAR1) is a transmembrane protein located in the mitochondria-associated endoplasmic reticulum membrane, and plays an important role in cell survival as a pluripotent modulator of a variety of signaling pathways related to neurodegeneration. Though SIGMAR1 is a potential target for neurodegenerative diseases, the specific role of SIGMAR1 in different tissue and cell types remains unclear. Here we reported the generation of Sigmar1 conditional knockout (Sigmar1loxP ) mice using CRISPR-Cas9 method to insert loxP sites into the 5'- and 3'-untranslated regions of Sigmar1. We showed that the insertion of loxP sequences did not affect the expression of Sigmar1 and that Sigmar1loxP/loxP mice exhibited no detectable visual defects compared with wild-type mice at the early adult stage. By crossing Sigmar1loxP mice with retina-specific Six3-Cre and ubiquitous CMV-Cre mice, we confirmed the deletion of Sigmar1 coding regions of exons 1-4, and the retina-specific and global loss of SIGMAR1 expression, respectively. Thus, Sigmar1loxP mice provide a valuable tool for unraveling the tissue and cell-type-specific role of Sigmar1.
Collapse
Affiliation(s)
- Liang Huang
- Department of Neuroscience and Regenerative Medicine, Augusta University Medical College of Georgia, Augusta, Georgia, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, USA
| | - Haiyan Xiao
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, USA.,Department of Cellular Biology and Anatomy, Augusta University Medical College of Georgia, Augusta, Georgia, USA
| | - Xiaoling Xie
- Department of Neuroscience and Regenerative Medicine, Augusta University Medical College of Georgia, Augusta, Georgia, USA
| | - Fang Hu
- Department of Neuroscience and Regenerative Medicine, Augusta University Medical College of Georgia, Augusta, Georgia, USA
| | - Fulei Tang
- Department of Neuroscience and Regenerative Medicine, Augusta University Medical College of Georgia, Augusta, Georgia, USA
| | - Sylvia B Smith
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, USA.,Department of Cellular Biology and Anatomy, Augusta University Medical College of Georgia, Augusta, Georgia, USA
| | - Lin Gan
- Department of Neuroscience and Regenerative Medicine, Augusta University Medical College of Georgia, Augusta, Georgia, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
19
|
Milenina LS, Krutetskaya ZI, Antonov VG, Krutetskaya NI. Sigma-1 Receptor Ligands Chlorpromazine and Trifluoperazine Attenuate Ca 2+ Responses in Rat Peritoneal Macrophages. CELL AND TISSUE BIOLOGY 2022; 16:233-244. [PMID: 35668825 PMCID: PMC9136207 DOI: 10.1134/s1990519x22030075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/23/2022]
Abstract
Sigma-1 receptors are ubiquitous multifunctional ligand-regulated molecular chaperones in the endoplasmic reticulum membrane with a unique history, structure, and pharmacological profile. Sigma-1 receptors bind ligands of different chemical structure and pharmacological action and modulate a wide range of cellular processes in health and disease, including Ca2+ signaling. To elucidate the involvement of sigma-1 receptors in the processes of Ca2+ signaling in macrophages we studied the effect of sigma-1 receptor ligands, phenothiazine neuroleptics chlorpromazine and trifluoperazine, on Ca2+ responses induced by inhibitors of endoplasmic Ca2+-ATPases thapsigargin and cyclopiazonic acid, as well as by disulfide-containing immunomodulators Glutoxim and Molixan in rat peritoneal macrophages. Using Fura-2AM microfluorimetry we showed for the first time that chlorpromazine and trifluoperazine inhibit both phases of Ca2+ responses induced by Glutoxim, Molixan, thapsigargin, and cyclopiazonic acid in rat peritoneal macrophages. The data obtained indicate the participation of sigma-1 receptors in a complex signaling cascade caused by Glutoxim or Molixan and leading to an increase in intracellular Ca2+ concentration in macrophages. The results also indicate the involvement of sigma-1 receptors in the regulation of store-dependent Ca2+entry in macrophages.
Collapse
Affiliation(s)
- L. S. Milenina
- Department of Biophysics, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Z. I. Krutetskaya
- Department of Biophysics, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - V. G. Antonov
- Department of Clinical Biochemistry and Laboratory Diagnostics, Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - N. I. Krutetskaya
- Department of Biophysics, St. Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
20
|
Meng F, Xiao Y, Ji Y, Sun Z, Zhou X. An open-like conformation of the sigma-1 receptor reveals its ligand entry pathway. Nat Commun 2022; 13:1267. [PMID: 35273182 PMCID: PMC8913746 DOI: 10.1038/s41467-022-28946-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/15/2022] [Indexed: 02/08/2023] Open
Abstract
The sigma-1 receptor (σ1R) is a non-opioid transmembrane receptor which has been implicated in many diseases, including neurodegenerative disorders and cancer. After more than forty years of research, substantial progress has been made in understanding this unique receptor, yet the molecular mechanism of its ligand entry pathway remains uncertain. Published structures of human σ1R reveal its homotrimeric organization of a cupin-fold β-barrel body that contains the ligand binding site, a carboxy-terminal V-shaped two-helix bundle, and a single amino-terminal transmembrane helix, while simulation studies have suggested a ligand entry pathway that is generated by conformational rearrangements of the cupin-fold domain. Here, we present multiple crystal structures, including an open-like conformation, of σ1R from Xenopus laevis. Together with functional binding analysis our data suggest that access to the σ1R ligand binding site is likely achieved by protein conformational changes that involve the carboxy-terminal two-helix bundle, rather than structural changes in the cupin-fold domain.
Collapse
Affiliation(s)
- Fuhui Meng
- Department of Integrated Traditional Chinese and Western Medicine, Rare Diseases Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yang Xiao
- Department of Integrated Traditional Chinese and Western Medicine, Rare Diseases Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yujia Ji
- Department of Integrated Traditional Chinese and Western Medicine, Rare Diseases Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ziyi Sun
- Department of Integrated Traditional Chinese and Western Medicine, Rare Diseases Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xiaoming Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Rare Diseases Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
21
|
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: 21] [Impact Index Per Article: 5.3] [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
|
22
|
Rossino G, Rui M, Linciano P, Rossi D, Boiocchi M, Peviani M, Poggio E, Curti D, Schepmann D, Wünsch B, González-Avendaño M, Vergara-Jaque A, Caballero J, Collina S. Bitopic Sigma 1 Receptor Modulators to Shed Light on Molecular Mechanisms Underpinning Ligand Binding and Receptor Oligomerization. J Med Chem 2021; 64:14997-15016. [PMID: 34624193 DOI: 10.1021/acs.jmedchem.1c00886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The sigma 1 receptor (S1R) is an enigmatic ligand-operated chaperone involved in many important biological processes, and its functions are not fully understood yet. Herein, we developed a novel series of bitopic S1R ligands as versatile tools to investigate binding processes, allosteric modulation, and the oligomerization mechanism. These molecules have been prepared in the enantiopure form and subjected to a preliminary biological evaluation, while in silico investigations helped to rationalize the results. Compound 7 emerged as the first bitopic S1R ligand endowed with low nanomolar affinity (Ki = 2.6 nM) reported thus far. Computational analyses suggested that 7 may stabilize the open conformation of the S1R by simultaneously binding the occluded primary binding site and a peripheral site on the cytosol-exposed surface. These findings pave the way to new S1R ligands with enhanced activity and/or selectivity, which could also be used as probes for the identification of a potential allosteric site.
Collapse
Affiliation(s)
- Giacomo Rossino
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Marta Rui
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Pasquale Linciano
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Daniela Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Massimo Boiocchi
- Centro Grandi Strumenti, University of Pavia, via Bassi 21, 27100 Pavia, Italy
| | - Marco Peviani
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
| | - Elena Poggio
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
| | - Daniela Curti
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
| | - Dirk Schepmann
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Correnstraße 48, 48149 Münster, Germany
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Correnstraße 48, 48149 Münster, Germany
| | - Mariela González-Avendaño
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
| | - Ariela Vergara-Jaque
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
| | - Julio Caballero
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| |
Collapse
|
23
|
Jabłońska M, Grzelakowska K, Wiśniewski B, Mazur E, Leis K, Gałązka P. Pridopidine in the treatment of Huntington's disease. Rev Neurosci 2021; 31:441-451. [PMID: 32083454 DOI: 10.1515/revneuro-2019-0085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/06/2019] [Indexed: 01/26/2023]
Abstract
Huntington's disease (HD) is a highly common inherited monogenic neurodegenerative disease, and the gene responsible for its development is located in the 4p16.3 chromosome. The product of that gene mutation is an abnormal huntingtin (Htt) protein that disrupts the neural conduction, thus leading to motor and cognitive disorders. The disease progresses to irreversible changes in the central nervous system (CNS). Although only a few drugs are available to symptomatic treatment, 'dopamine stabilizers' (as represented by the pridopidine) may be the new treatment options. The underlying causes of HD are dopaminergic conduction disorders. Initially, the disease is hyperkinetic (chorea) until it eventually reaches the hypokinetic phase. Studies confirmed a correlation between the amount of dopamine in the CNS and the stage of the disease. Pridopidine has the capacity to be a dopamine buffer, which could increase or decrease the dopamine content depending on the disease phase. A research carried out on animal models demonstrated the protective effect of pridopidine on nerve cells thanks to its ability to alter the cortical glutamatergic signaling through the N-methyl-D-aspartate (NMDA) receptors. Studies on dopamine stabilizers also reported that pridopidine has a 100-fold greater affinity for the sigma-1 receptor than for the D2 receptor. Disturbances in the activity of sigma-1 receptors occur in neurodegenerative diseases, including HD. Their interaction with pridopidine results in the neuroprotective effect, which is manifested as an increase in the plasticity of synaptic neurons and prevention of their atrophy within the striatum. To determine the effectiveness of pridopidine in the treatment of HD, large multicenter randomized studies such as HART, MermaiHD, and PRIDE-HD were carried out.
Collapse
Affiliation(s)
- Magdalena Jabłońska
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Klaudyna Grzelakowska
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Bartłomiej Wiśniewski
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Ewelina Mazur
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Kamil Leis
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Przemysław Gałązka
- Department of General and Oncological Pediatric Surgery, Antoni Jurasz University Hospital No. 1 in Bydgoszcz, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| |
Collapse
|
24
|
Wang M, Wan C, He T, Han C, Zhu K, Waddington JL, Zhen X. Sigma-1 receptor regulates mitophagy in dopaminergic neurons and contributes to dopaminergic protection. Neuropharmacology 2021; 196:108360. [PMID: 33122030 DOI: 10.1016/j.neuropharm.2020.108360] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 12/26/2022]
Abstract
Mitochondria are essential for neuronal survival and function, and mitochondrial dysfunction plays a critical role in the pathological development of Parkinson's disease (PD). Mitochondrial quality control is known to contribute to the survival of dopaminergic (DA) neurons, with mitophagy being a key regulator of the quality control system. In this study, we show that mitophagy is impaired in the substantia nigra pars compacta (SNc) of the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. Treatment with the sigma-1 receptor (Sig 1R) agonist 2-morpholin-4-ylethyl 1-phenylcyclohexane-1-carboxylate (PRE-084) reduced loss of DA neurons, restored motor ability and MPTP-induced damage to mitophagy activity in the SNc of PD-like mice. Additionally, knockdown of Sig 1R in SH-SY5Y DA cells inhibited mitophagy and enhanced 1-methyl-4-phenylpyridinium ion (MPP+) neurotoxicity, whereas application of the Sig 1R selective agonist SKF10047 promoted clearance of damaged mitochondria. Moreover, knockdown of Sig 1R in SH-SY5Y cells resulted in decreased levels of p-ULK1 (Unc-51 Like Autophagy Activating Kinase 1) (Ser555), p-TBK1 (TANK Binding Kinase 1) (Ser172), p-ubiquitin (Ub) (Ser65), Parkin recruitment, and stabilization of PTEN-induced putative kinase 1 (PINK1) in mitochondria. The present data provide the first evidence for potential roles of PINK1/Parkin in Sig 1R-modulated mitophagy in DA neurons.
Collapse
Affiliation(s)
- Mingmei Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Chunlei Wan
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Tao He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Chaojun Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Kailian Zhu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - John L Waddington
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China.
| |
Collapse
|
25
|
Dvorácskó S, Lázár L, Fülöp F, Palkó M, Zalán Z, Penke B, Fülöp L, Tömböly C, Bogár F. Novel High Affinity Sigma-1 Receptor Ligands from Minimal Ensemble Docking-Based Virtual Screening. Int J Mol Sci 2021; 22:8112. [PMID: 34360878 PMCID: PMC8347176 DOI: 10.3390/ijms22158112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023] Open
Abstract
Sigma-1 receptor (S1R) is an intracellular, multi-functional, ligand operated protein that also acts as a chaperone. It is considered as a pluripotent drug target in several pathologies. The publication of agonist and antagonist bound receptor structures has paved the way for receptor-based in silico drug design. However, recent studies on this subject payed no attention to the structural differences of agonist and antagonist binding. In this work, we have developed a new ensemble docking-based virtual screening protocol utilizing both agonist and antagonist bound S1R structures. This protocol was used to screen our in-house compound library. The S1R binding affinities of the 40 highest ranked compounds were measured in competitive radioligand binding assays and the sigma-2 receptor (S2R) affinities of the best S1R binders were also determined. This way three novel high affinity S1R ligands were identified and one of them exhibited a notable S1R/S2R selectivity.
Collapse
Affiliation(s)
- Szabolcs Dvorácskó
- Biological Research Centre, Institute of Biochemistry, Eötvös Loránd Research Network (ELKH), H-6726 Szeged, Hungary; (S.D.); (C.T.)
| | - László Lázár
- Institute of Pharmaceutical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (L.L.); (F.F.); (M.P.); (Z.Z.)
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (L.L.); (F.F.); (M.P.); (Z.Z.)
| | - Márta Palkó
- Institute of Pharmaceutical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (L.L.); (F.F.); (M.P.); (Z.Z.)
| | - Zita Zalán
- Institute of Pharmaceutical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (L.L.); (F.F.); (M.P.); (Z.Z.)
| | - Botond Penke
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Hungary;
| | - Lívia Fülöp
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Hungary;
| | - Csaba Tömböly
- Biological Research Centre, Institute of Biochemistry, Eötvös Loránd Research Network (ELKH), H-6726 Szeged, Hungary; (S.D.); (C.T.)
| | - Ferenc Bogár
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Hungary;
- MTA-SZTE Biomimetic Systems Research Group, Eötvös Loránd Research Network (ELKH), H-6720 Szeged, Hungary
| |
Collapse
|
26
|
Aishwarya R, Abdullah CS, Morshed M, Remex NS, Bhuiyan MS. Sigmar1's Molecular, Cellular, and Biological Functions in Regulating Cellular Pathophysiology. Front Physiol 2021; 12:705575. [PMID: 34305655 PMCID: PMC8293995 DOI: 10.3389/fphys.2021.705575] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The Sigma 1 receptor (Sigmar1) is a ubiquitously expressed multifunctional inter-organelle signaling chaperone protein playing a diverse role in cellular survival. Recessive mutation in Sigmar1 have been identified as a causative gene for neuronal and neuromuscular disorder. Since the discovery over 40 years ago, Sigmar1 has been shown to contribute to numerous cellular functions, including ion channel regulation, protein quality control, endoplasmic reticulum-mitochondrial communication, lipid metabolism, mitochondrial function, autophagy activation, and involved in cellular survival. Alterations in Sigmar1’s subcellular localization, expression, and signaling has been implicated in the progression of a wide range of diseases, such as neurodegenerative diseases, ischemic brain injury, cardiovascular diseases, diabetic retinopathy, cancer, and drug addiction. The goal of this review is to summarize the current knowledge of Sigmar1 biology focusing the recent discoveries on Sigmar1’s molecular, cellular, pathophysiological, and biological functions.
Collapse
Affiliation(s)
- Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Md Shenuarin Bhuiyan
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States.,Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| |
Collapse
|
27
|
Sharma M, Naura AS, Singla SK. A deleterious interplay between endoplasmic reticulum stress and its functional linkage to mitochondria in nephrolithiasis. Free Radic Biol Med 2021; 168:70-80. [PMID: 33798617 DOI: 10.1016/j.freeradbiomed.2021.03.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/15/2021] [Accepted: 03/24/2021] [Indexed: 12/18/2022]
Abstract
Hyperoxaluria is one of the leading causes of calcium oxalate stone formation in the kidney. Since hyperoxaluria produces Endoplasmic Reticulum (ER) stress in the kidney, it is thus likely that the adaptive unfolded protein response might affect the mitochondrial population as ER and mitochondria share close physical and functional interactions mandatory for several biological processes. Thus this work was designed to study the putative effects of endoplasmic reticulum stress on the renal mitochondria during hyperoxaluria-induced nephrolithiasis. The results showed that hyperoxaluria induced an ER stress led to the unfolded protein response in the renal tissue of experimental rats. Hampered mitochondrion functioning was detected with decreased mitochondrial membrane potential and upsurged mitochondria calcium. These changes in the mitochondria function and ER stress are preceded by apoptosis. The expression of Sigma-1 receptor protein found in the Mitochondria associated ER membranes, the connecting link between ER and mitochondria was found to decrease in the hyperoxaluric rats. Inhibition of ER stress by 4-Phenylbutyric acid prevented the decrease in mitochondria membrane potential and increase in mitochondria calcium observed in hyperoxaluric rats. Also, it restored the protein expression of the sigma-1 receptor protein. On the other hand, N-acetyl cysteine had a nominal impact on the reduction of the ER stress-induced mitochondrial dysfunction. In conclusion, our data showed that hyperoxaluria induces renal ER stress which triggers mitochondria dysfunction, might be via alteration in the sigma-1 receptor protein in the mitochondria-associated ER membranes, which leads to apoptosis, renal injury, and calcium oxalate crystal deposition.
Collapse
Affiliation(s)
- Minu Sharma
- Department of Biochemistry, Panjab University, Chandigarh, India.
| | - Amarjit S Naura
- Department of Biochemistry, Panjab University, Chandigarh, India.
| | - S K Singla
- Department of Biochemistry, Panjab University, Chandigarh, India.
| |
Collapse
|
28
|
Grieb P, Swiatkiewicz M, Prus K, Rejdak K. Hypoxia may be a determinative factor in COVID-19 progression. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100030. [PMID: 34870146 PMCID: PMC8106824 DOI: 10.1016/j.crphar.2021.100030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/21/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
The disease which develops following SARS-CoV-2 virus infection, known as COVID-19, in most affected countries displays mortality from 1.5% to 9.8%. When leukocytosis due to granulocytosis, thrombocytopenia, and increased level of D-dimers are detected early during the disease course, they are accurate predictors of mortality. Based on the published observations that each of the aforementioned disturbances by itself may appear as a consequence of hypoxia, a hypothesis is presented that early hypoxia consequential to sleep apnea and/or blunted respiratory response to chemical stimuli is an early determinant of COVID-19 progression to the severe and critical stage. Further, it is noted that host-directed therapies which may counteract hypoxia and its early downstream effects are initiated only upon hospitalization of COVID-19 patients, which is too late to be fully effective. An example is anticoagulation treatment with low molecular weight heparin. Repurposing drugs which could counteract some early posthypoxic events, such as fluvoxamine, amantadine and N-acetylcysteine, for post-exposure prophylaxis of SARS-CoV-2 infection and early prehospital treatment of COVID-19, is indicated.
Collapse
Affiliation(s)
- Pawel Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Maciej Swiatkiewicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Prus
- Department of Neurology, Medical University of Lublin, Lublin, Poland
| | - Konrad Rejdak
- Department of Neurology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
29
|
Zhemkov V, Geva M, Hayden MR, Bezprozvanny I. Sigma-1 Receptor (S1R) Interaction with Cholesterol: Mechanisms of S1R Activation and Its Role in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:4082. [PMID: 33920913 PMCID: PMC8071319 DOI: 10.3390/ijms22084082] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
The sigma-1 receptor (S1R) is a 223 amino acid-long transmembrane endoplasmic reticulum (ER) protein. The S1R modulates the activity of multiple effector proteins, but its signaling functions are poorly understood. S1R is associated with cholesterol, and in our recent studies we demonstrated that S1R association with cholesterol induces the formation of S1R clusters. We propose that these S1R-cholesterol interactions enable the formation of cholesterol-enriched microdomains in the ER membrane. We hypothesize that a number of secreted and signaling proteins are recruited and retained in these microdomains. This hypothesis is consistent with the results of an unbiased screen for S1R-interacting partners, which we performed using the engineered ascorbate peroxidase 2 (APEX2) technology. We further propose that S1R agonists enable the disassembly of these cholesterol-enriched microdomains and the release of accumulated proteins such as ion channels, signaling receptors, and trophic factors from the ER. This hypothesis may explain the pleotropic signaling functions of the S1R, consistent with previously observed effects of S1R agonists in various experimental systems.
Collapse
Affiliation(s)
- Vladimir Zhemkov
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Michal Geva
- Prilenia Therapeutics Development LTD, Herzliya 4673304, Israel; (M.G.); (M.R.H.)
| | - Michael R. Hayden
- Prilenia Therapeutics Development LTD, Herzliya 4673304, Israel; (M.G.); (M.R.H.)
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V6H 3V5, Canada
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
- Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg State Polytechnic University, 195251 St. Petersburg, Russia
| |
Collapse
|
30
|
Maurice T. Bi-phasic dose response in the preclinical and clinical developments of sigma-1 receptor ligands for the treatment of neurodegenerative disorders. Expert Opin Drug Discov 2021; 16:373-389. [PMID: 33070647 DOI: 10.1080/17460441.2021.1838483] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022]
Abstract
Introduction: The sigma-1 receptor (S1R) is attracting much attention for disease-modifying therapies in neurodegenerative diseases. It is a conserved protein, present in plasma and endoplasmic reticulum (ER) membranes and enriched in mitochondria-associated ER membranes (MAMs). It modulates ER-mitochondria Ca2+ transfer and ER stress pathways. Mitochondrial and MAM dysfunctions contribute to neurodegenerative processes in diseases such as Alzheimer, Parkinson, Huntington or Amyotrophic Lateral Sclerosis. Interestingly, the S1R can be activated by small druggable molecules and accumulating preclinical data suggest that S1R agonists are effective protectants in these neurodegenerative diseases.Area covered: In this review, we will present the data showing the high therapeutic potential of S1R drugs for the treatment of neurodegenerative diseases, focusing on pridopidine as a potent and selective S1R agonist under clinical development. Of particular interest is the bi-phasic (bell-shaped) dose-response effect, representing a common feature of all S1R agonists and described in numerous preclinical models in vitro, in vivo and in clinical trials.Expert opinion: S1R agonists modulate inter-organelles communication altered in neurodegenerative diseases and activate intracellular survival pathways. Research will continue growing in the future. The particular cellular nature of this chaperone protein must be better understood to facilitate the clinical developement of promising molecules.
Collapse
Affiliation(s)
- Tangui Maurice
- MMDN, Univ Montpellier, EPHE, INSERM, UMR_S1198, Montpellier, France
| |
Collapse
|
31
|
Casanovas M, Reyes-Resina I, Lillo A, Lillo J, López-Arnau R, Camarasa J, Escubedo E, Navarro G, Franco R. Methamphetamine Blocks Adenosine A 2A Receptor Activation via Sigma 1 and Cannabinoid CB 1 Receptors. Int J Mol Sci 2021; 22:2743. [PMID: 33803075 PMCID: PMC7963146 DOI: 10.3390/ijms22052743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 03/01/2021] [Indexed: 11/16/2022] Open
Abstract
Methamphetamine is, worldwide, one of the most consumed drugs of abuse. One important side effect is neurodegeneration leading to a decrease in life expectancy. The aim of this paper was to check whether the drug affects one of the receptors involved in neurodegeneration/neuroprotection events, namely the adenosine A2A receptor (A2AR). First, we noticed that methamphetamine does not affect A2A functionality if the receptor is expressed in a heterologous system. However, A2AR becomes sensitive to the drug upon complexes formation with the cannabinoid CB1 receptor (CB1R) and the sigma 1 receptor (σ1R). Signaling via both adenosine A2AR and cannabinoid CB1R was affected by methamphetamine in cells co-expressing the two receptors. In striatal primary cultures, the A2AR-CB1R heteromer complex was detected and methamphetamine not only altered its expression but completely blocked the A2AR- and the CB1R-mediated activation of the mitogen activated protein kinase (MAPK) pathway. In conclusion, methamphetamine, with the participation of σ1R, alters the expression and function of two interacting receptors, A2AR, which is a therapeutic target for neuroprotection, and CB1R, which is the most abundant G protein-coupled receptor (GPCR) in the brain.
Collapse
Affiliation(s)
- Mireia Casanovas
- Biology School, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (M.C.); (I.R.-R.); (J.L.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Irene Reyes-Resina
- Biology School, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (M.C.); (I.R.-R.); (J.L.)
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain;
| | - Jaume Lillo
- Biology School, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (M.C.); (I.R.-R.); (J.L.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Raul López-Arnau
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain; (R.L.-A.); (J.C.); (E.E.)
| | - Jorge Camarasa
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain; (R.L.-A.); (J.C.); (E.E.)
| | - Elena Escubedo
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain; (R.L.-A.); (J.C.); (E.E.)
| | - Gemma Navarro
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain;
| | - Rafael Franco
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Chemistry School, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
32
|
Agha H, McCurdy CR. In vitro and in vivo sigma 1 receptor imaging studies in different disease states. RSC Med Chem 2021; 12:154-177. [PMID: 34046607 PMCID: PMC8127618 DOI: 10.1039/d0md00186d] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/02/2020] [Indexed: 12/11/2022] Open
Abstract
The sigma receptor system has been classified into two distinct subtypes, sigma 1 (σ1R) and sigma 2 (σ2R). Sigma 1 receptors (σ1Rs) are involved in many neurodegenerative diseases and different central nervous system disorders such as Alzheimer's disease, Parkinson's disease, schizophrenia, and drug addiction, and pain. This makes them attractive targets for developing radioligands as tools to gain a better understanding of disease pathophysiology and clinical diagnosis. Over the years, several σ1R radioligands have been developed to image the changes in σ1R distribution and density providing insights into their role in disease development. Moreover, the involvement of both σ1Rs and σ2Rs with cancer make these ligands, especially those that are σ2R selective, great tools for imaging different types of tumors. This review will discuss the principles of molecular imaging using PET and SPECT, known σ1R radioligands and their applications for labelling σ1Rs under different disease conditions. Furthermore, this review will highlight σ1R radioligands that have demonstrated considerable potential as biomarkers, and an opportunity to fulfill the ultimate goal of better healthcare outcomes and improving human health.
Collapse
Affiliation(s)
- Hebaalla Agha
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida Gainesville FL 32610 USA +(352) 273 7705 +1 (352) 294 8691
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida Gainesville FL 32610 USA +(352) 273 7705 +1 (352) 294 8691
- UF Translational Drug Development Core, University of Florida Gainesville FL 32610 USA
| |
Collapse
|
33
|
Battista T, Pascarella G, Staid DS, Colotti G, Rosati J, Fiorillo A, Casamassa A, Vescovi AL, Giabbai B, Semrau MS, Fanelli S, Storici P, Squitieri F, Morea V, Ilari A. Known Drugs Identified by Structure-Based Virtual Screening Are Able to Bind Sigma-1 Receptor and Increase Growth of Huntington Disease Patient-Derived Cells. Int J Mol Sci 2021; 22:1293. [PMID: 33525510 PMCID: PMC7865886 DOI: 10.3390/ijms22031293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/15/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Huntington disease (HD) is a devastating and presently untreatable neurodegenerative disease characterized by progressively disabling motor and mental manifestations. The sigma-1 receptor (σ1R) is a protein expressed in the central nervous system, whose 3D structure has been recently determined by X-ray crystallography and whose agonists have been shown to have neuroprotective activity in neurodegenerative diseases. To identify therapeutic agents against HD, we have implemented a drug repositioning strategy consisting of: (i) Prediction of the ability of the FDA-approved drugs publicly available through the ZINC database to interact with σ1R by virtual screening, followed by computational docking and visual examination of the 20 highest scoring drugs; and (ii) Assessment of the ability of the six drugs selected by computational analyses to directly bind purified σ1R in vitro by Surface Plasmon Resonance and improve the growth of fibroblasts obtained from HD patients, which is significantly impaired with respect to control cells. All six of the selected drugs proved able to directly bind purified σ1R in vitro and improve the growth of HD cells from both or one HD patient. These results support the validity of the drug repositioning procedure implemented herein for the identification of new therapeutic tools against HD.
Collapse
Affiliation(s)
- Theo Battista
- Institute of Molecular Biology and Pathology, National Research Council of Italy, 00185 Rome, Italy; (T.B.); (G.P.); (D.S.S.); (G.C.)
- Department of Biochemical Sciences “A. Rossi Fanelli”, “Sapienza” University, 00185 Rome, Italy;
| | - Gianmarco Pascarella
- Institute of Molecular Biology and Pathology, National Research Council of Italy, 00185 Rome, Italy; (T.B.); (G.P.); (D.S.S.); (G.C.)
- Department of Biochemical Sciences “A. Rossi Fanelli”, “Sapienza” University, 00185 Rome, Italy;
| | - David Sasah Staid
- Institute of Molecular Biology and Pathology, National Research Council of Italy, 00185 Rome, Italy; (T.B.); (G.P.); (D.S.S.); (G.C.)
- Department of Biochemical Sciences “A. Rossi Fanelli”, “Sapienza” University, 00185 Rome, Italy;
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, National Research Council of Italy, 00185 Rome, Italy; (T.B.); (G.P.); (D.S.S.); (G.C.)
| | - Jessica Rosati
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (J.R.); (A.C.); (A.L.V.)
| | - Annarita Fiorillo
- Department of Biochemical Sciences “A. Rossi Fanelli”, “Sapienza” University, 00185 Rome, Italy;
| | - Alessia Casamassa
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (J.R.); (A.C.); (A.L.V.)
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy
| | - Angelo Luigi Vescovi
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (J.R.); (A.C.); (A.L.V.)
| | - Barbara Giabbai
- Protein Facility, Structural Biology Lab, Elettra Sincrotrone Trieste, 34149 Basovizza, Italy; (B.G.); (M.S.S.); (P.S.)
| | - Marta Stefania Semrau
- Protein Facility, Structural Biology Lab, Elettra Sincrotrone Trieste, 34149 Basovizza, Italy; (B.G.); (M.S.S.); (P.S.)
- Department of Cellular, Computational and Integrative Biology—CIBIO, University of Trento, 38123 Trento, Italy
| | - Sergio Fanelli
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (S.F.); (F.S.)
| | - Paola Storici
- Protein Facility, Structural Biology Lab, Elettra Sincrotrone Trieste, 34149 Basovizza, Italy; (B.G.); (M.S.S.); (P.S.)
| | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (S.F.); (F.S.)
| | - Veronica Morea
- Institute of Molecular Biology and Pathology, National Research Council of Italy, 00185 Rome, Italy; (T.B.); (G.P.); (D.S.S.); (G.C.)
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, National Research Council of Italy, 00185 Rome, Italy; (T.B.); (G.P.); (D.S.S.); (G.C.)
| |
Collapse
|
34
|
Lara A, Esperante I, Meyer M, Liere P, Di Giorgio N, Schumacher M, Guennoun R, Gargiulo-Monachelli G, De Nicola AF, Gonzalez Deniselle MC. Neuroprotective Effects of Testosterone in Male Wobbler Mouse, a Model of Amyotrophic Lateral Sclerosis. Mol Neurobiol 2021; 58:2088-2106. [PMID: 33411236 DOI: 10.1007/s12035-020-02209-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/12/2020] [Indexed: 11/30/2022]
Abstract
Patients suffering of amyotrophic lateral sclerosis (ALS) present motoneuron degeneration leading to muscle atrophy, dysphagia, and dysarthria. The Wobbler mouse, an animal model of ALS, shows a selective loss of motoneurons, astrocytosis, and microgliosis in the spinal cord. The incidence of ALS is greater in men; however, it increases in women after menopause, suggesting a role of sex steroids in ALS. Testosterone is a complex steroid that exerts its effects directly via androgen (AR) or Sigma-1 receptors and indirectly via estrogen receptors (ER) after aromatization into estradiol. Its reduced-metabolite 5α-dihydrotestosterone acts via AR. This study analyzed the effects of testosterone in male symptomatic Wobblers. Controls or Wobblers received empty or testosterone-filled silastic tubes for 2 months. The cervical spinal cord from testosterone-treated Wobblers showed (1) similar androgen levels to untreated control and (2) increased levels of testosterone, and its 5α-reduced metabolites, 5α- dihydrotestosterone, and 3β-androstanediol, but (3) undetectable levels of estradiol compared to untreated Wobblers. Testosterone-treated controls showed comparable steroid concentrations to its untreated counterpart. In testosterone- treated Wobblers a reduction of AR, ERα, and aromatase and high levels of Sigma-1 receptor mRNAs was demonstrated. Testosterone treatment increased ChAT immunoreactivity and the antiinflammatory mediator TGFβ, while it lessened vacuolated motoneurons, GFAP+ astrogliosis, the density of IBA1+ microgliosis, proinflammatory mediators, and oxidative/nitrosative stress. Clinically, testosterone treatment in Wobblers slowed the progression of paw atrophy and improved rotarod performance. Collectively, our findings indicate an antiinflammatory and protective effect of testosterone in the degenerating spinal cord. These results coincided with a high concentration of androgen-reduced derivatives after testosterone treatment suggesting that the steroid profile may have a beneficial role on disease progression.
Collapse
Affiliation(s)
- Agustina Lara
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428, Buenos Aires, Argentina
| | - Iván Esperante
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428, Buenos Aires, Argentina
| | - Maria Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428, Buenos Aires, Argentina
| | - Philippe Liere
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, Le Kremlin-Bicêtre, 94276, France
| | - Noelia Di Giorgio
- Laboratory of Neuroendocrinology, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, Buenos Aires, 1428, Argentina
| | - Michael Schumacher
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, Le Kremlin-Bicêtre, 94276, France
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, Le Kremlin-Bicêtre, 94276, France
| | - Gisella Gargiulo-Monachelli
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428, Buenos Aires, Argentina
| | - Alejandro Federico De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428, Buenos Aires, Argentina.,Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1121, Buenos Aires, Argentina
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428, Buenos Aires, Argentina. .,Department of Physiology, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, Buenos Aires, 1121, Argentina.
| |
Collapse
|
35
|
Shinoda Y, Haga Y, Akagawa K, Fukunaga K. Wildtype σ1 receptor and the receptor agonist improve ALS-associated mutation-induced insolubility and toxicity. J Biol Chem 2020; 295:17573-17587. [PMID: 33453999 PMCID: PMC7762949 DOI: 10.1074/jbc.ra120.015012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/10/2020] [Indexed: 12/12/2022] Open
Abstract
Genetic mutations related to ALS, a progressive neurological disease, have been discovered in the gene encoding σ-1 receptor (σ1R). We previously reported that σ1RE102Q elicits toxicity in cells. The σ1R forms oligomeric states that are regulated by ligands. Nevertheless, little is known about the effect of ALS-related mutations on oligomer formation. Here, we transfected NSC-34 cells, a motor neuronal cell line, and HEK293T cells with σ1R-mCherry (mCh), σ1RE102Q-mCh, or nontagged forms to investigate detergent solubility and subcellular distribution using immunocytochemistry and fluorescence recovery after photobleaching. The oligomeric state was determined using crosslinking procedure. σ1Rs were soluble to detergents, whereas the mutants accumulated in the insoluble fraction. Within the soluble fraction, peak distribution of mutants appeared in higher sucrose density fractions. Mutants formed intracellular aggregates that were co-stained with p62, ubiquitin, and phosphorylated pancreatic eukaryotic translation initiation factor-2-α kinase in NSC-34 cells but not in HEK293T cells. The aggregates had significantly lower recovery in fluorescence recovery after photobleaching. Acute treatment with σ1R agonist SA4503 failed to improve recovery, whereas prolonged treatment for 48 h significantly decreased σ1RE102Q-mCh insolubility and inhibited apoptosis. Whereas σ1R-mCh formed monomers and dimers, σ1RE102Q-mCh also formed trimers and tetramers. SA4503 reduced accumulation of the four types in the insoluble fraction and increased monomers in the soluble fraction. The σ1RE102Q insolubility was diminished by σ1R-mCh co-expression. These results suggest that the agonist and WT σ1R modify the detergent insolubility, toxicity, and oligomeric state of σ1RE102Q, which may lead to promising new treatments for σ1R-related ALS.
Collapse
Affiliation(s)
- Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yudai Haga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Koichiro Akagawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
36
|
Fluorinated 2-Arylcyclopropan-1-amines - A new class of sigma receptor ligands. Bioorg Med Chem 2020; 28:115726. [PMID: 33007549 DOI: 10.1016/j.bmc.2020.115726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 11/20/2022]
Abstract
Stereoisomeric 2-aryl-2-fluoro-cyclopropan-1-amines have been discovered as a new class of σ receptor ligands showing different selectivity for the two subtypes of the receptor. Generally, compounds substituted in 4-position are much more active than corresponding 3-substituted isomers. trans-2-Fluoro-2-(4-methoxyphenyl)cyclopropan-1-amine (19a) was the most potent (Ki = 4.8 nM) σ1 receptor ligand, while cis-2-fluoro-2-(4-trifluoromethylphenyl)cyclopropan-1-amine (20b) was the most potent (Ki = 95 nM) σ2 receptor ligand.
Collapse
|
37
|
Grachev ID, Meyer PM, Becker GA, Bronzel M, Marsteller D, Pastino G, Voges O, Rabinovich L, Knebel H, Zientek F, Rullmann M, Sattler B, Patt M, Gerhards T, Strauss M, Kluge A, Brust P, Savola JM, Gordon MF, Geva M, Hesse S, Barthel H, Hayden MR, Sabri O. Sigma-1 and dopamine D2/D3 receptor occupancy of pridopidine in healthy volunteers and patients with Huntington disease: a [ 18F] fluspidine and [ 18F] fallypride PET study. Eur J Nucl Med Mol Imaging 2020; 48:1103-1115. [PMID: 32995944 PMCID: PMC8041674 DOI: 10.1007/s00259-020-05030-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/07/2020] [Indexed: 11/24/2022]
Abstract
PURPOSE Pridopidine is an investigational drug for Huntington disease (HD). Pridopidine was originally thought to act as a dopamine stabilizer. However, pridopidine shows highest affinity to the sigma-1 receptor (S1R) and enhances neuroprotection via the S1R in preclinical studies. Using [18F] fluspidine and [18F] fallypride PET, the purpose of this study was to assess in vivo target engagement/receptor occupancy of pridopidine to the S1R and dopamine D2/D3 receptor (D2/D3R) at clinical relevant doses in healthy volunteers (HVs) and as proof-of-concept in a small number of patients with HD. METHODS Using [18F] fluspidine PET (300 MBq, 0-90 min), 11 male HVs (pridopidine 0.5 to 90 mg; six dose groups) and three male patients with HD (pridopidine 90 mg) were investigated twice, without and 2 h after single dose of pridopidine. Using [18F] fallypride PET (200 MBq, 0-210 min), four male HVs were studied without and 2 h following pridopidine administration (90 mg). Receptor occupancy was analyzed by the Lassen plot. RESULTS S1R occupancy as function of pridopidine dose (or plasma concentration) in HVs could be described by a three-parameter Hill equation with a Hill coefficient larger than one. A high degree of S1R occupancy (87% to 91%) was found throughout the brain at pridopidine doses ranging from 22.5 to 90 mg. S1R occupancy was 43% at 1 mg pridopidine. In contrast, at 90 mg pridopidine, the D2/D3R occupancy was only minimal (~ 3%). CONCLUSIONS Our PET findings indicate that at clinically relevant single dose of 90 mg, pridopidine acts as a selective S1R ligand showing near to complete S1R occupancy with negligible occupancy of the D2/D3R. The dose S1R occupancy relationship suggests cooperative binding of pridopidine to the S1R. Our findings provide significant clarification about pridopidine's mechanism of action and support further use of the 45-mg twice-daily dose to achieve full and selective targeting of the S1R in future clinical trials of neurodegenerative disorders. Clinical Trials.gov Identifier: NCT03019289 January 12, 2017; EUDRA-CT-Nr. 2016-001757-41.
Collapse
Affiliation(s)
- Igor D Grachev
- Teva Branded Pharmaceutical Products R&D, Inc, Malvern, PA, 19355, USA.,Guide Pharmaceutical Consulting, LLC, Millstone, NJ, 08535, USA
| | - Philipp M Meyer
- Department of Nuclear Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Georg A Becker
- Department of Nuclear Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Marcus Bronzel
- ABX-CRO Advanced Pharmaceutical Services Forschungsgesellschaft mbH, Dresden, Germany
| | - Doug Marsteller
- Teva Branded Pharmaceutical Products R&D, Inc, Frazer, PA, 19355, USA
| | - Gina Pastino
- Teva Branded Pharmaceutical Products R&D, Inc, Frazer, PA, 19355, USA
| | - Ole Voges
- ABX-CRO Advanced Pharmaceutical Services Forschungsgesellschaft mbH, Dresden, Germany
| | - Laura Rabinovich
- Teva Branded Pharmaceutical Products R&D, Inc, Frazer, PA, 19355, USA
| | - Helena Knebel
- Teva Branded Pharmaceutical Products R&D, Inc, Frazer, PA, 19355, USA
| | - Franziska Zientek
- Department of Nuclear Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Michael Rullmann
- Department of Nuclear Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Bernhard Sattler
- Department of Nuclear Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Thilo Gerhards
- Department of Nuclear Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Maria Strauss
- Department of Psychiatry and Psychotherapy, University of Leipzig Medical Center, Leipzig, Germany
| | - Andreas Kluge
- ABX-CRO Advanced Pharmaceutical Services Forschungsgesellschaft mbH, Dresden, Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Leipzig, Germany
| | - Juha-Matti Savola
- Teva Branded Pharmaceutical Products R&D, Inc, Frazer, PA, 19355, USA
| | - Mark F Gordon
- Teva Branded Pharmaceutical Products R&D, Inc, Frazer, PA, 19355, USA
| | - Michal Geva
- Prilenia Therapeutics Development Ltd., Herzliya, Israel
| | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | | | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
38
|
Voronin MV, Vakhitova YV, Seredenin SB. Chaperone Sigma1R and Antidepressant Effect. Int J Mol Sci 2020; 21:E7088. [PMID: 32992988 PMCID: PMC7582751 DOI: 10.3390/ijms21197088] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
This review analyzes the current scientific literature on the role of the Sigma1R chaperone in the pathogenesis of depressive disorders and pharmacodynamics of antidepressants. As a result of ligand activation, Sigma1R is capable of intracellular translocation from the endoplasmic reticulum (ER) into the region of nuclear and cellular membranes, where it interacts with resident proteins. This unique property of Sigma1R provides regulation of various receptors, ion channels, enzymes, and transcriptional factors. The current review demonstrates the contribution of the Sigma1R chaperone to the regulation of molecular mechanisms involved in the antidepressant effect.
Collapse
Affiliation(s)
- Mikhail V. Voronin
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia;
| | | | - Sergei B. Seredenin
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia;
| |
Collapse
|
39
|
Almási N, Török S, Dvorácskó S, Tömböly C, Csonka Á, Baráth Z, Murlasits Z, Valkusz Z, Pósa A, Varga C, Kupai K. Lessons on the Sigma-1 Receptor in TNBS-Induced Rat Colitis: Modulation of the UCHL-1, IL-6 Pathway. Int J Mol Sci 2020; 21:E4046. [PMID: 32516975 PMCID: PMC7312485 DOI: 10.3390/ijms21114046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/27/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammatory Bowel Disease (IBD) is an autoimmune ailment of the gastrointestinal (GI) tract, which is characterized by enhanced activation of proinflammatory cytokines. It is suggested that the sigma-1 receptor (σ1R) confers anti-inflammatory effects. As the exact pathogenesis of IBD is still unknown and treatment options are limited, we aimed to investigate the effects of σ1R in 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced experimental colitis. To this end, male Wistar-Harlan rats were used to model colitic inflammation through the administration of TNBS. To investigate the effects of σ1R, Fluvoxamine (FLV, σ1R agonist) and BD1063 (σ1R antagonist) were applied via intracolonic administration to the animals once a day for three days. Our radioligand binding studies indicated the existence of σ1Rs as [3H](+)-pentazocine binding sites, and FLV treatment increased the reduced σ1R maximum binding capacity in TNBS-induced colitis. Furthermore, FLV significantly attenuated the colonic damage, the effect of which was abolished by the administration of BD1063. Additionally, FLV potentially increased the expression of ubiquitin C-terminal hydrolase ligase-1 (UCHL-1) and the levels of endothelial nitric oxide synthase (eNOS), and decreased the levels of interleukin-6 (IL-6) and inducible NOS (iNOS) expression. In summary, our study offers evidence for the anti-inflammatory potential of FLV and σ1R in experimental colitis, and our results present a promising approach to the development of new σ1R-targeted treatment options against IBD.
Collapse
Affiliation(s)
- Nikoletta Almási
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, H-6726 Szeged, Hungary; (N.A.); (S.T.); (A.P.); (C.V.)
| | - Szilvia Török
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, H-6726 Szeged, Hungary; (N.A.); (S.T.); (A.P.); (C.V.)
| | - Szabolcs Dvorácskó
- Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre, H-6726 Szeged, Hungary; (S.D.); (C.T.)
- Department of Medical Chemistry, University of Szeged, H-6725 Szeged, Hungary
| | - Csaba Tömböly
- Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre, H-6726 Szeged, Hungary; (S.D.); (C.T.)
| | - Ákos Csonka
- Department of Traumatology, University of Szeged, H-6725 Szeged, Hungary;
| | - Zoltán Baráth
- Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, University of Szeged, H-6720 Szeged, Hungary;
| | - Zsolt Murlasits
- Laboratory Animals Research Center, Qatar University, Doha 2713, Qatar;
| | - Zsuzsanna Valkusz
- 1st Department of Medicine, Medical Faculty, Albert Szent-Györgyi Clinical Center, University of Szeged, H-6720 Szeged, Hungary;
| | - Anikó Pósa
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, H-6726 Szeged, Hungary; (N.A.); (S.T.); (A.P.); (C.V.)
| | - Csaba Varga
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, H-6726 Szeged, Hungary; (N.A.); (S.T.); (A.P.); (C.V.)
| | - Krisztina Kupai
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, H-6726 Szeged, Hungary; (N.A.); (S.T.); (A.P.); (C.V.)
| |
Collapse
|
40
|
Lavender E, Hirasawa-Fujita M, Domino EF. Ketamine's dose related multiple mechanisms of actions: Dissociative anesthetic to rapid antidepressant. Behav Brain Res 2020; 390:112631. [PMID: 32437885 DOI: 10.1016/j.bbr.2020.112631] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 03/19/2020] [Accepted: 03/29/2020] [Indexed: 01/15/2023]
Abstract
Ketamine induces safe and effective anesthesia and displays unusual cataleptic properties that gave rise to the term dissociative anesthesia. Since 1970, clinicians only utilized the drug as an anesthetic or analgesic for decades, but ketamine was found to have rapid acting antidepressant effects in 1990s. Accumulated evidence exhibits NMDAR antagonism may not be the only mechanism of ketamine. The contributions of AMPA receptor, mTor signal pathway, monoaminergic system, sigma-1 receptor, cholinergic, opioid and cannabinoid systems, as well as voltage-gated calcium channels and hyperpolarization cyclic nucleotide gated channels are discussed for the antidepressant effects. Also the effects of ketamine's enantiomers and metabolites are reviewed. Furthermore ketamine's anesthetic and analgesic mechanisms are briefly revisited. Overall, pharmacology of ketamine, its enantiomers and metabolites is very unique. Insight into multiple mechanisms of action will provide further development and desirable clinical effects of ketamine.
Collapse
Affiliation(s)
- Eli Lavender
- University of Michigan Medical School, Department of Pharmacology, 1150 W Medical Center Dr, Ann Arbor, MI 48109, USA
| | - Mika Hirasawa-Fujita
- University of Michigan Medical School, Department of Pharmacology, 1150 W Medical Center Dr, Ann Arbor, MI 48109, USA
| | - Edward F Domino
- University of Michigan Medical School, Department of Pharmacology, 1150 W Medical Center Dr, Ann Arbor, MI 48109, USA.
| |
Collapse
|
41
|
Toussaint M, Deuther-Conrad W, Kranz M, Fischer S, Ludwig FA, Juratli TA, Patt M, Wünsch B, Schackert G, Sabri O, Brust P. Sigma-1 Receptor Positron Emission Tomography: A New Molecular Imaging Approach Using ( S)-(-)-[ 18F]Fluspidine in Glioblastoma. Molecules 2020; 25:E2170. [PMID: 32384802 PMCID: PMC7248975 DOI: 10.3390/molecules25092170] [Citation(s) in RCA: 5] [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: 04/13/2020] [Revised: 04/30/2020] [Accepted: 05/02/2020] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most devastating primary brain tumour characterised by infiltrative growth and resistance to therapies. According to recent research, the sigma-1 receptor (sig1R), an endoplasmic reticulum chaperone protein, is involved in signaling pathways assumed to control the proliferation of cancer cells and thus could serve as candidate for molecular characterisation of GBM. To test this hypothesis, we used the clinically applied sig1R-ligand (S)-(-)-[18F]fluspidine in imaging studies in an orthotopic mouse model of GBM (U87-MG) as well as in human GBM tissue. A tumour-specific overexpression of sig1R in the U87-MG model was revealed in vitro by autoradiography. The binding parameters demonstrated target-selective binding according to identical KD values in the tumour area and the contralateral side, but a higher density of sig1R in the tumour. Different kinetic profiles were observed in both areas, with a slower washout in the tumour tissue compared to the contralateral side. The translational relevance of sig1R imaging in oncology is reflected by the autoradiographic detection of tumour-specific expression of sig1R in samples obtained from patients with glioblastoma. Thus, the herein presented data support further research on sig1R in neuro-oncology.
Collapse
Affiliation(s)
- Magali Toussaint
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
| | - Mathias Kranz
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
- PET Imaging Center, University Hospital of North Norway (UNN), 9009 Tromsø, Norway
- Nuclear Medicine and Radiation Biology Research Group, The Arctic University of Norway, 9009 Tromsø, Norway
| | - Steffen Fischer
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
| | - Friedrich-Alexander Ludwig
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
| | - Tareq A. Juratli
- Department of Neurosurgery, Technische Universität Dresden (TUD), University Hospital Carl Gustav Carus, 01307 Dresden, Germany; (T.A.J.); (G.S.)
| | - Marianne Patt
- Department of Nuclear Medicine, University Hospital Leipzig, 04318 Leipzig, Germany; (M.P.); (O.S.)
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, 48149 Münster, Germany;
| | - Gabriele Schackert
- Department of Neurosurgery, Technische Universität Dresden (TUD), University Hospital Carl Gustav Carus, 01307 Dresden, Germany; (T.A.J.); (G.S.)
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, 04318 Leipzig, Germany; (M.P.); (O.S.)
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
| |
Collapse
|
42
|
Hong WC. Distinct Regulation of σ 1 Receptor Multimerization by Its Agonists and Antagonists in Transfected Cells and Rat Liver Membranes. J Pharmacol Exp Ther 2020; 373:290-301. [PMID: 32060048 DOI: 10.1124/jpet.119.262790] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 02/04/2020] [Indexed: 11/22/2022] Open
Abstract
Extensive studies have shown that the σ 1 receptor (σ 1R) interacts with and modulates the activity of multiple proteins with important biological functions. Recent crystal structures of σ 1R as a homotrimer differ from a dimer-tetramer model postulated earlier. It remains inconclusive whether ligand binding regulates σ 1R oligomerization. Here, novel nondenaturing gel methods and mutational analysis were used to examine σ 1R oligomerization. In transfected cells, σ 1R exhibited as multimers, dimers, and monomers. Overall, σ 1R agonists decreased, whereas σ 1R antagonists increased σ 1R multimers, suggesting that agonists and antagonists differentially affect the stability of σ 1R multimers. Endogenous σ 1R in rat liver membranes also showed similar regulation of oligomerization as in cells. Mutations at key residues lining the trimerization interface (Arg119, Asp195, Phe191, Trp136, and Gly91) abolished multimerization without disrupting dimerization. Intriguingly, truncation of the N terminus reduced σ 1R to apparent monomer. These results demonstrate that multiple domains play crucial roles in coordinating high-order quaternary organization of σ 1R. The E102Q σ 1R mutant implicated in juvenile amyotrophic lateral sclerosis formed dimers only, suggesting that dysregulation of σ 1R multimeric assembly may impair its function. Interestingly, oligomerization of σ 1R was pH-dependent and correlated with changes in [3H](+)-pentazocine binding affinity and Bmax Combined with mutational analysis, it is reasoned that σ 1R multimers possess high-affinity and high-capacity [3H](+)-pentazocine binding, whereas monomers likely lack binding. These results suggest that σ 1R may exist in interconvertible oligomeric states in a dynamic equilibrium. Further exploration of ligand-regulated σ 1R multimerization may provide novel approaches to modulate the function of σ 1R and its interacting proteins. SIGNIFICANCE STATEMENT: The σ 1 receptor (σ 1R) modulates the activities of various partner proteins. Recently, crystal structures of σ 1R were elucidated as homotrimers. This study used novel nondenaturing gel methods to examine σ1R oligomerization in transfected cells and rat liver membranes. Overall, agonist binding decreased, whereas antagonist binding increased σ 1R multimers, which comprised trimers and larger units. σ 1R multimers were shown to bind [3H](+)-pentazocine with high affinity and high capacity. Furthermore, mutational analysis revealed a crucial role of its N-terminal domain in σ 1R multimerization.
Collapse
Affiliation(s)
- Weimin Conrad Hong
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana
| |
Collapse
|
43
|
Cai L, Liow JS, Morse CL, Telu S, Davies R, Frankland MP, Zoghbi SS, Cheng K, Hall MD, Innis RB, Pike VW. Evaluation of 11C-NR2B-SMe and Its Enantiomers as PET Radioligands for Imaging the NR2B Subunit Within the NMDA Receptor Complex in Rats. J Nucl Med 2020; 61:1212-1220. [PMID: 31924728 DOI: 10.2967/jnumed.119.235143] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/19/2019] [Indexed: 01/01/2023] Open
Abstract
[S-methyl-11C](±)-7-methoxy-3-(4-(4-(methylthio)phenyl)butyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-ol (11C-NR2B-SMe) and its enantiomers were synthesized as candidates for imaging the NR2B subunit within the N-methyl-d-aspartate receptor with PET. Methods: Brains were scanned with PET for 90 min after intravenous injection of one of the candidate radioligands into rats. To detect any NR2B-specific binding of radioligand in brain, various preblocking or displacing agents were evaluated for their impact on the PET brain imaging data. Radiometabolites from brain and other tissues were measured ex vivo and in vitro. Results: Each radioligand gave high early whole-brain uptake of radioactivity, followed by a brief fast decline and then a slow final decline. 11C-(S)-NR2B-SMe was studied extensively. Ex vivo measurements showed that radioactivity in rat brain at 30 min after radioligand injection was virtually unchanged radioligand. Only less lipophilic radiometabolites appeared in plasma. High-affinity NR2B ligands, Ro-25-6981, ifenprodil, and CO101244, showed increasing preblocking of whole-brain radioactivity retention with increasing dose (0.01-3.00 mg/kg, intravenously). Five σ1 antagonists (FTC146, BD1407, F3, F4, and NE100) and 4 σ1 agonists ((+)-pentazocine, (±)-PPCC, PRE-084, and (+)-SKF10047) were ineffective preblocking agents, except FTC146 and F4 at a high dose. Two potent σ1 receptor agonists, TC1 and SA4503, showed dose-dependent preblocking effects in the presence or absence of pharmacologic σ1 receptor blockade with FTC146. Conclusion: 11C-(S)-NR2B-SMe has adequate NR2B-specific PET signal in rat brain to warrant further evaluation in higher species. TC1 and SA4503 likely have off-target binding to NR2B in vivo.
Collapse
Affiliation(s)
- Lisheng Cai
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Riley Davies
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Michael P Frankland
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Ken Cheng
- NCATS Chemical Genomics Center, National Institutes of Health, Rockville, Maryland
| | - Matthew D Hall
- NCATS Chemical Genomics Center, National Institutes of Health, Rockville, Maryland
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| |
Collapse
|
44
|
Delint-Ramirez I, Garcia-Oscos F, Segev A, Kourrich S. Cocaine engages a non-canonical, dopamine-independent, mechanism that controls neuronal excitability in the nucleus accumbens. Mol Psychiatry 2020; 25:680-691. [PMID: 29880884 PMCID: PMC7042730 DOI: 10.1038/s41380-018-0092-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 04/03/2018] [Accepted: 04/13/2018] [Indexed: 11/25/2022]
Abstract
Drug-induced enhanced dopamine (DA) signaling in the brain is a canonical mechanism that initiates addiction processes. However, indirect evidence suggests that cocaine also triggers non-canonical, DA-independent, mechanisms that contribute to behavioral responses to cocaine, including psychomotor sensitization and cocaine self-administration. Identifying these mechanisms and determining how they are initiated is fundamental to further our understanding of addiction processes. Using physiologically relevant in vitro tractable models, we found that cocaine-induced hypoactivity of nucleus accumbens shell (NAcSh) medium spiny neurons (MSNs), one hallmark of cocaine addiction, is independent of DA signaling. Combining brain slice studies and site-directed mutagenesis in HEK293T cells, we found that cocaine binding to intracellular sigma-1 receptor (σ1) initiates this mechanism. Subsequently, σ1 binds to Kv1.2 potassium channels, followed by accumulation of Kv1.2 in the plasma membrane, thereby depressing NAcSh MSNs firing. This mechanism is specific to D1 receptor-expressing MSNs. Our study uncovers a mechanism for cocaine that bypasses DA signaling and leads to addiction-relevant neuroadaptations, thereby providing combinatorial strategies for treating stimulant abuse.
Collapse
Affiliation(s)
- Ilse Delint-Ramirez
- 0000 0000 9482 7121grid.267313.2Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Francisco Garcia-Oscos
- 0000 0000 9482 7121grid.267313.2Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Amir Segev
- 0000 0000 9482 7121grid.267313.2Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Saïd Kourrich
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
45
|
Rossino G, Orellana I, Caballero J, Schepmann D, Wünsch B, Rui M, Rossi D, González-Avendaño M, Collina S, Vergara-Jaque A. New Insights into the Opening of the Occluded Ligand-Binding Pocket of Sigma1 Receptor: Binding of a Novel Bivalent RC-33 Derivative. J Chem Inf Model 2019; 60:756-765. [DOI: 10.1021/acs.jcim.9b00649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Giacomo Rossino
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Ivana Orellana
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
| | - Julio Caballero
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
| | - Dirk Schepmann
- Institute of Pharmaceutical and Medicinal Chemistry, University of Muenster, Correnstrasse 48, 48149 Münster, Germany
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, University of Muenster, Correnstrasse 48, 48149 Münster, Germany
| | - Marta Rui
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Daniela Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Mariela González-Avendaño
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Ariela Vergara-Jaque
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
- Multidisciplinary Scientific Nucleus, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
- Millennium Nucleus of Ion Channels-associated Diseases (MiNICAD), Santiago, Chile
| |
Collapse
|
46
|
Soriani O, Kourrich S. The Sigma-1 Receptor: When Adaptive Regulation of Cell Electrical Activity Contributes to Stimulant Addiction and Cancer. Front Neurosci 2019; 13:1186. [PMID: 31780884 PMCID: PMC6861184 DOI: 10.3389/fnins.2019.01186] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022] Open
Abstract
The sigma-1 receptor (σ1R) is an endoplasmic reticulum (ER)-resident chaperone protein that acts like an inter-organelle signaling modulator. Among its several functions such as ER lipid metabolisms/transports and indirect regulation of genes transcription, one of its most intriguing feature is the ability to regulate the function and trafficking of a variety of functional proteins. To date, and directly relevant to the present review, σ1R has been found to regulate both voltage-gated ion channels (VGICs) belonging to distinct superfamilies (i.e., sodium, Na+; potassium, K+; and calcium, Ca2+ channels) and non-voltage-gated ion channels. This regulatory function endows σ1R with a powerful capability to fine tune cells’ electrical activity and calcium homeostasis—a regulatory power that appears to favor cell survival in pathological contexts such as stroke or neurodegenerative diseases. In this review, we present the current state of knowledge on σ1R’s role in the regulation of cellular electrical activity, and how this seemingly adaptive function can shift cell homeostasis and contribute to the development of very distinct chronic pathologies such as psychostimulant abuse and tumor cell growth in cancers.
Collapse
Affiliation(s)
| | - Saïd Kourrich
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC, Canada.,Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
47
|
Bunse P, Schlepphorst C, Glorius F, Kitamura M, Wünsch B. Short and Atom-Economic Enantioselective Synthesis of the σ 1-Receptor Ligands ( S)- and ( R)-Fluspidine-Important Tools for Positron Emission Tomography Studies. J Org Chem 2019; 84:13744-13754. [PMID: 31523971 DOI: 10.1021/acs.joc.9b01882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aryl bromides 2a and 2b bearing an alkynyl substituent in the o-position reacted with n-butyllithium and 1-benzylpiperidin-4-one in a one-pot Domino reaction to form ester 3 and aldehyde 5, respectively. Enantiomeric alcohols (R)-8 and (S)-8 were obtained by conjugate NaBH4 reduction of α,β-unsaturated ester 3 in the presence of chiral cocomplexes (R,R)-10 and (S,S)-10. Starting from orthoester 2a, the precursors (R)-8 and (S)-8 for the synthesis of fluspidine enantiomers (R)-1/[18F](R)-1 and (S)-1/[18F](S)-1 were obtained in only two reaction steps without additional steps for N-protection in an atom-economic manner in 95.6% ee and 97.2% ee, respectively.
Collapse
Affiliation(s)
- Paul Bunse
- Institut für Pharmazeutische und Medizinische Chemie der Westfälischen Wilhelms-Universität Münster , Corrensstraße 48 , D-48149 Münster , Germany
| | - Christoph Schlepphorst
- Organisch-Chemisches Institut der Westfälischen Wilhelms-Universität Münster , Corrensstraße 40 , D-48149 Münster , Germany
| | - Frank Glorius
- Organisch-Chemisches Institut der Westfälischen Wilhelms-Universität Münster , Corrensstraße 40 , D-48149 Münster , Germany
| | - Masato Kitamura
- Graduate School of Pharmaceutical Sciences and Research Center for Materials Science , Nagoya University , Chikusa, Nagoya 464-8601 , Japan
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie der Westfälischen Wilhelms-Universität Münster , Corrensstraße 48 , D-48149 Münster , Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003-CiM) , Westfälische Wilhelms-Universität Münster , D-48149 Münster , Germany
| |
Collapse
|
48
|
Schmidt HR, Kruse AC. The Molecular Function of σ Receptors: Past, Present, and Future. Trends Pharmacol Sci 2019; 40:636-654. [PMID: 31387763 PMCID: PMC6748033 DOI: 10.1016/j.tips.2019.07.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/03/2019] [Accepted: 07/10/2019] [Indexed: 10/26/2022]
Abstract
The σ1 and σ2 receptors are enigmatic proteins that have attracted attention for decades due to the chemical diversity and therapeutic potential of their ligands. However, despite ongoing clinical trials with σ receptor ligands for multiple conditions, relatively little is known regarding the molecular function of these receptors. In this review, we revisit past research on σ receptors and discuss the interpretation of these data in light of recent developments. We provide a synthesis of emerging structural and genetic data on the σ1 receptor and discuss the recent cloning of the σ2 receptor. Finally, we discuss the major questions that remain in the study of σ receptors.
Collapse
Affiliation(s)
- Hayden R Schmidt
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
49
|
Yang H, Shen H, Li J, Guo LW. SIGMAR1/Sigma-1 receptor ablation impairs autophagosome clearance. Autophagy 2019; 15:1539-1557. [PMID: 30871407 PMCID: PMC6693456 DOI: 10.1080/15548627.2019.1586248] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 01/30/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022] Open
Abstract
Autophagosome-lysosome fusion is a common critical step in various forms of macroautophagy/autophagy including mitophagy, the selective degradation of mitochondria. Regulations of this fusion process remain poorly defined. Here we have determined the role of SIGMAR1, a unique endoplasmic reticulum membrane protein. Knockout of Sigmar1 impaired mitochondrial clearance without altering the PINK1-PRKN/Parkin signaling, in mouse retinal explants and cultured cells treated with carbonyl cyanide m-chlorophenyl hydrazone (CCCP) for induction of mitophagy. SIGMAR1 depletion also caused accumulation of autophagosome markers LC3-II and SQSTM1, but did not change the levels of BECN1 and ATG7, proteins associated with autophagosome biogenesis. Lysosomal pH and protease activities were not negatively affected. However, sigmar1 knockout partially compromised autophagosome-lysosome fusion in CCCP-treated NSC34 cells, as revealed by reduced GFP fluorescence quenching of GFP-RFP-LC3-II puncta and co-localization of lysosomes with mitochondria. Furthermore, SIGMAR1 co-immunoprecipitated with ATG14, STX17, and VAMP8 (but not SNAP29), proteins key to autophagosome-lysosome membrane fusion. Re-expressing SIGMAR1 in the null background rescued clearance of mitochondria and autophagosomes. In summary, we started out finding that sigmar1 knockout impaired the clearance of mitochondria and autophagosomes, and then narrowed down the SIGMAR1 modulation to the autophagosome-lysosome fusion step. This study may shed new light on understanding autophagy-associated cyto-protection and disease mechanisms. Abbreviations: APEX2, a genetically engineered peroxidase; BiFC, bimolecule fluorescence complementation; CCCP, a mitophagy inducing compound; CRISPR, clustered regularly interspaced short palindromic repeats; EM, electron microscopy; ER, endoplasmic reticulum; MAP1LC3/LC3, microtubule-associated protein 1 light chain 3; SIGMAR1, sigma non-opioid intracellular receptor 1.
Collapse
Affiliation(s)
- Huan Yang
- Department of Surgery and Departemnt of Physiology & Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH USA
| | - Hongtao Shen
- Department of Surgery and Departemnt of Physiology & Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH USA
| | - Jing Li
- Department of Surgery and Departemnt of Physiology & Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH USA
| | - Lian-Wang Guo
- Department of Surgery and Departemnt of Physiology & Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI
| |
Collapse
|
50
|
Torres B, Tyler JS, Satyshur KA, Ruoho AE. Human indole(ethyl)amine-N-methyltransferase (hINMT) catalyzed methylation of tryptamine, dimethylsulfide and dimethylselenide is enhanced under reducing conditions - A comparison between 254C and 254F, two common hINMT variants. PLoS One 2019; 14:e0219664. [PMID: 31310642 PMCID: PMC6634407 DOI: 10.1371/journal.pone.0219664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 06/28/2019] [Indexed: 11/18/2022] Open
Abstract
Phenylalanine and cysteine comprise common miss-sense variants (i.e., single nucleotide polymorphisms [SNPs]) at amino acid position 254 of the human indole(ethyl)amine-N-methyltransferase (hINMT). The phenylalanine variant, which occurs in linkage disequilibrium with two 3’ UTR SNPs, has been reported to associate with elevated urine levels of trimethylselenonium (TMSe), the Se-methylated product of volatile dimethylselenide. hINMT allozymes expressing either cysteine (254C) or phenylalanine (254F) at position 254 were compared for enzyme activity (i.e., Km and Vmax) towards the INMT substrates tryptamine, dimethylsulfide (DMS) and dimethylselenide (DMSe) in vitro. The SNP 254C had a higher Vmax for DMS and tryptamine in the presence of reducing agent than in its absence. Conversely, Vmax for 254F was insensitive to the presence or absence of reducing agent for these substrates. SNP 254F showed a lower Km for tryptamine in the absence of reducing agent than 254C. No statistically significant difference in Vmax or Km was observed between 254C and 254F allozymes in the presence of reducing agent for DMSe, The Km values for DMSe methylation were about 10-fold (254C) or 6-fold (254F) more favorable than for tryptamine methylation with reducing agent present. These findings indicated that: 1) That phenylalanine at position 254 renders hINMT methylation of substrates DMS and tryptamine insensitive to a non reducing environment. 2) That human INMT harbors significant thioether-S-methyltransferase (TEMT) activity with a higher affinity for DMSe than tryptamine, 3) The reduction of a 44C/254C disulfide bond in hINMT that increases Vmax is proposed.
Collapse
Affiliation(s)
- Brian Torres
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - James S. Tyler
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Kenneth A. Satyshur
- Small Molecule Screening Facility, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Arnold E. Ruoho
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|