1
|
Kecskés S, Mészáros M, Dvorácskó S, Szabó Í, Porkoláb G, Barna L, Harazin A, Szecskó A, Menyhárt Á, Bari F, Deli MA, Penke B, Farkas E, Veszelka S. The impact of the novel σ 1 receptor ligand (S)-L1 on brain endothelial cells and cerebrovascular reactivity challenged by ischemia. Eur J Pharmacol 2025; 1000:177724. [PMID: 40348322 DOI: 10.1016/j.ejphar.2025.177724] [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: 02/13/2025] [Revised: 04/14/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Intracellular sigma-1 receptors (σ1 receptors) have a versatile function through the regulation of lipid rafts, neuroreceptors and ion channels, and can influence signal transduction and neuronal plasticity. Since decreased activity of σ1 receptors is a common pathological feature in the early stages of many neurological diseases, σ1 receptor agonists may represent a promising therapeutic tool for the treatment of these disorders. In this study, we aimed to comprehensively investigate the potential protective effects of the novel synthetic σ1 receptor agonist (S)-L1 against endothelial endoplasmic reticulum (ER) stress and cerebral ischemia. In binding affinity experiments, we showed that (S)-L1 has a high affinity and selectivity for σ1 receptor with virtually no affinity for any of the other receptors tested. Next, (S)-L1 exerted protection against endoplasmic reticulum stress in human brain endothelial cells, consistent with the localization of σ1 receptors in endothelial cells. Furthermore, (S)-L1 penetration was demonstrated across the cell culture model of the blood-brain barrier, providing a rationale for neuronal action in addition to endothelial protection. Finally, (S)-L1 inhibited spreading depolarization, suppressed apoptosis and rescued astrocytes in a rat model of cerebral ischemia. Based on our results, (S)-L1 exerts a protective effect on both brain endothelial cells and neural tissue. Moreover, since these experiments revealed no affinity for serotonergic receptors, the compound holds promise as an adjuvant therapy for the treatment of cerebrovascular disease without potential psychedelic side effects.
Collapse
Affiliation(s)
- Szilvia Kecskés
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Cerebral Blood Flow and Metabolism Research Group, Somogyi u. 4, Szeged, 6720, Hungary; Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi School of Medicine and Faculty of Science and Informatics, University of Szeged, Somogyi u. 4, Szeged, 6720, Hungary
| | - Mária Mészáros
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary; One Health Institute, Faculty of Health Sciences, University of Debrecen, Nagyerdei krt. 98, Debrecen, 4032, Hungary
| | - Szabolcs Dvorácskó
- Laboratory of Biomolecular Structure and Pharmacology, Institute of Biochemistry, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Írisz Szabó
- Department of Medical Physics and Informatics, Albert Szent-Györgyi School of Medicine and Faculty of Science and Informatics, University of Szeged, Korányi fasor 9, Szeged, 6720, Hungary; Lendület Laboratory of Systems Neuroscience, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Gergő Porkoláb
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary; Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Lilla Barna
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
| | - András Harazin
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Anikó Szecskó
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary; Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Ákos Menyhárt
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Cerebral Blood Flow and Metabolism Research Group, Somogyi u. 4, Szeged, 6720, Hungary; Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi School of Medicine and Faculty of Science and Informatics, University of Szeged, Somogyi u. 4, Szeged, 6720, Hungary
| | - Ferenc Bari
- Department of Medical Physics and Informatics, Albert Szent-Györgyi School of Medicine and Faculty of Science and Informatics, University of Szeged, Korányi fasor 9, Szeged, 6720, Hungary
| | - Mária A Deli
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Botond Penke
- Department of Medical Chemistry, Albert Szent-Györgyi School of Medicine, University of Szeged, Dóm tér 8, Szeged, 6720, Hungary
| | - Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Cerebral Blood Flow and Metabolism Research Group, Somogyi u. 4, Szeged, 6720, Hungary; Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi School of Medicine and Faculty of Science and Informatics, University of Szeged, Somogyi u. 4, Szeged, 6720, Hungary.
| | - Szilvia Veszelka
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary.
| |
Collapse
|
2
|
Haddad M, Dieckmann LHJ, Viola TW, de Araújo MR, da Silva NR, Mari JDJ. The Efficacy of Fluvoxamine in Anxiety Disorders and Obsessive-Compulsive Disorder: An Overview of Systematic Reviews and Meta-Analyses. Pharmaceuticals (Basel) 2025; 18:353. [PMID: 40143130 PMCID: PMC11944676 DOI: 10.3390/ph18030353] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/14/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Objective: This systematic review aims to evaluate the efficacy of fluvoxamine in the treatment of anxiety disorders and obsessive-compulsive disorder (OCD) by synthesizing evidence from systematic reviews and meta-analyses. Methods: We conducted a literature search in PubMed and the Cochrane Central Register of Controlled Trials, focusing on fluvoxamine's efficacy in generalized anxiety disorder (GAD), social anxiety disorder (SAD), panic disorder (PD), and OCD. We included systematic reviews and meta-analyses of randomized controlled trials (RCTs) comparing fluvoxamine to a placebo or other drugs. The quality of evidence from the included reviews was assessed using A Measurement Tool to Assess Systematic Reviews-version 2 (AMSTAR-2). Results: The study included fourteen systematic reviews (five for OCD, three for SAD, and six for PD), covering thirty-seven RCTs (sixteen for OCD, six for SAD, and fifteen for PD), with a total of 3621 patients (1745 with OCD, 1034 with SAD, and 842 with PD). A high-quality systematic review demonstrated that fluvoxamine is superior to a placebo in improving symptoms and the response rates for OCD. Three meta-analyses comparing fluvoxamine to clomipramine in OCD found no significant differences in efficacy regarding symptom improvement. Two additional systematic reviews, both rated as high quality, confirmed the superiority of fluvoxamine in reducing symptom severity and improving the response rates in patients with SAD compared to a placebo. However, the findings for PD were inconsistent. A meta-analysis, also rated as high quality, found that while fluvoxamine showed better response rates than a placebo, the difference was not statistically significant. Conclusions: Overall, the efficacy of fluvoxamine in the treatment of OCD and SAD was demonstrated. While some reviews highlighted its potential in alleviating GAD, its impact on panic-specific outcomes remained inconsistent.
Collapse
Affiliation(s)
- Michel Haddad
- Department of Psychiatry, Brazilian Clinical Research Institute, São Paulo 01404-000, Brazil; (M.H.); (L.H.J.D.); (M.R.d.A.); (N.R.d.S.)
| | - Luiz Henrique Junqueira Dieckmann
- Department of Psychiatry, Brazilian Clinical Research Institute, São Paulo 01404-000, Brazil; (M.H.); (L.H.J.D.); (M.R.d.A.); (N.R.d.S.)
| | - Thiago Wendt Viola
- Brain Institute of Rio Grande do Sul, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, Brazil;
| | - Melissa Ribeiro de Araújo
- Department of Psychiatry, Brazilian Clinical Research Institute, São Paulo 01404-000, Brazil; (M.H.); (L.H.J.D.); (M.R.d.A.); (N.R.d.S.)
| | - Naielly Rodrigues da Silva
- Department of Psychiatry, Brazilian Clinical Research Institute, São Paulo 01404-000, Brazil; (M.H.); (L.H.J.D.); (M.R.d.A.); (N.R.d.S.)
| | - Jair de Jesus Mari
- Department of Psychiatry, Universidade Federal de São Paulo, São Paulo 04017-030, Brazil
| |
Collapse
|
3
|
Yang HM. Mitochondrial Dysfunction in Neurodegenerative Diseases. Cells 2025; 14:276. [PMID: 39996748 PMCID: PMC11853439 DOI: 10.3390/cells14040276] [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: 01/26/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Mitochondrial dysfunction represents a pivotal characteristic of numerous neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. These conditions, distinguished by unique clinical and pathological features, exhibit shared pathways leading to neuronal damage, all of which are closely associated with mitochondrial dysfunction. The high metabolic requirements of neurons make even minor mitochondrial deficiencies highly impactful, driving oxidative stress, energy deficits, and aberrant protein processing. Growing evidence from genetic, biochemical, and cellular investigations associates impaired electron transport chain activity and disrupted quality-control mechanisms, such as mitophagy, with the initial phases of disease progression. Furthermore, the overproduction of reactive oxygen species and persistent neuroinflammation can establish feedforward cycles that exacerbate neuronal deterioration. Recent clinical research has increasingly focused on interventions aimed at enhancing mitochondrial resilience-through antioxidants, small molecules that modulate the balance of mitochondrial fusion and fission, or gene-based therapeutic strategies. Concurrently, initiatives to identify dependable mitochondrial biomarkers seek to detect pathological changes prior to the manifestation of overt symptoms. By integrating the current body of knowledge, this review emphasizes the critical role of preserving mitochondrial homeostasis as a viable therapeutic approach. It also addresses the complexities of translating these findings into clinical practice and underscores the potential of innovative strategies designed to delay or potentially halt neurodegenerative processes.
Collapse
Affiliation(s)
- Han-Mo Yang
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| |
Collapse
|
4
|
Lukkahatai N, Ong IL, Benjasirisan C, Saligan LN. Brain-Derived Neurotrophic Factor (BDNF) as a Marker of Physical Exercise or Activity Effectiveness in Fatigue, Pain, Depression, and Sleep Disturbances: A Scoping Review. Biomedicines 2025; 13:332. [PMID: 40002745 PMCID: PMC11853410 DOI: 10.3390/biomedicines13020332] [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: 12/02/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Brain-derived neurotrophic factor (BDNF) has been investigated as a potential mechanistic marker or therapeutic target to manage symptoms such as fatigue, pain, depression, and sleep disturbances. However, the variability in BDNF response to exercise or physical activity (exercise/PA) and its clinical relevance in symptom management remains unclear. This scoping review assesses existing studies exploring the relationships between exercise/PA, symptoms, and BDNF levels, specifically focusing on fatigue, pain, depression, and sleep disturbances in adults. Methods: Relevant studies indexed in PubMed and CINAHL were identified. Using systematic review software, two reviewers independently screened and evaluated full texts, based on the following criteria: human studies reporting BDNF levels in adults, using exercise/PA interventions, assessing symptoms (pain, fatigue, depression, and/or sleep disturbance) as outcomes, and published in English. Results: Of 950 records, 35 records met the inclusion criteria. While exercise/PA is broadly supported for managing symptoms, 74.3% (n = 26) of studies reported increased BDNF levels, and only 40% (n = 14) showed significant increases following exercise/PA. Only 14% (n = 5) of studies demonstrated a significant relationship between changes in BDNF and symptoms. No significant differences in BDNF levels and symptoms were observed between different types of exercise (e.g., aerobic vs. strength vs. flexibility/stretching) and PA. Conclusions: The current literature provides insufficient evidence to confirm BDNF as a marker for exercise/PA effectiveness on symptoms. Further clinical investigations are needed to validate its potential as a therapeutic target.
Collapse
Affiliation(s)
- Nada Lukkahatai
- School of Nursing, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Irvin L. Ong
- Research Development and Innovation Center, Our Lady of Fatima University, Valenzuela City 1440, Philippines;
- Department of Nursing and Health Sciences, Elmhurst University, Elmhurst, IL 60126, USA
| | | | - Leorey N. Saligan
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD 20892, USA;
| |
Collapse
|
5
|
Winge T, Schepmann D, Schmidt J, Wünsch B. Synthesis and structure-affinity relationships of spirocyclic σ 1 receptor ligands with tetrahydropyran scaffold. Eur J Med Chem 2025; 281:117002. [PMID: 39547080 DOI: 10.1016/j.ejmech.2024.117002] [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: 09/06/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024]
Abstract
The σ1 receptor plays a key role in the regulation of various processes in the human body; it is involved in the development of neurodegenerative and neuropsychiatric diseases and is overexpressed in several human tumors rendering it an important target for potential drug candidates. In this project, spirocyclic σ1 receptor ligands with different substituents in 4- and 9-position were synthesized and investigated for their σ1 receptor affinity and selectivity over related targets. The σ1 affinity of the ligands was correlated with their lipophilicity (logD7.4 value) giving insight into their lipophilic ligand efficiency (LLE). The (pyridin-3-yl)methyl derivative 5i showed a promising balance of high σ1 affinity (Ki(σ1) = 3.9 nM) and selectivity (>250-fold) as well as high LLE of 5.8. 5i has a high plasma protein binding (89 %) and promising metabolic stability in the presence of mouse liver microsomes and NADPH (83 % intact after 90 min). Increasing the size of the piperidine ring of the spirocyclic ligands 5 to an azepane ring led to considerably increased σ1 affinity (Ki(5a) = 1.2 nM, Ki(23a) = 0.42 nM) and selectivity over σ2 receptors (5a: 45-fold, 23a: 150-fold).
Collapse
Affiliation(s)
- Tobias Winge
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149, Münster, Germany
| | - Dirk Schepmann
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149, Münster, Germany
| | - Judith Schmidt
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149, Münster, Germany
| | - Bernhard Wünsch
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149, Münster, Germany.
| |
Collapse
|
6
|
Sastri KT, Gupta NV, Kannan A, Dutta S, Ali M Osmani R, V B, Ramkishan A, S S. The next frontier in multiple sclerosis therapies: Current advances and evolving targets. Eur J Pharmacol 2024; 985:177080. [PMID: 39491741 DOI: 10.1016/j.ejphar.2024.177080] [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: 05/28/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Recent advancements in research have significantly enhanced our comprehension of the intricate immune components that contribute to multiple sclerosis (MS) pathogenesis. By conducting an in-depth analysis of complex molecular interactions involved in the immunological cascade of the disease, researchers have successfully identified novel therapeutic targets, leading to the development of innovative therapies. Leveraging pioneering technologies in proteomics, genomics, and the assessment of environmental factors has expedited our understanding of the vulnerability and impact of these factors on the progression of MS. Furthermore, these advances have facilitated the detection of significant biomarkers for evaluating disease activity. By integrating these findings, researchers can design novel molecules to identify new targets, paving the way for improved treatments and enhanced patient care. Our review presents recent discoveries regarding the pathogenesis of MS, highlights their genetic implications, and proposes an insightful approach for engaging with newer therapeutic targets in effectively managing this debilitating condition.
Collapse
Affiliation(s)
- K Trideva Sastri
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India.
| | - N Vishal Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India.
| | - Anbarasu Kannan
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Suman Dutta
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - Balamuralidhara V
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - A Ramkishan
- Deputy Drugs Controller (India), Central Drugs Standard Control Organization, Directorate General of Health Services, Ministry of Health & Family Welfare, Government of India, India
| | | |
Collapse
|
7
|
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
|
8
|
Schreihofer DA, Dalwadi D, Kim S, Metzger D, Oppong-Gyebi A, Das-Earl P, Schetz JA. Treatment of Stroke at a Delayed Timepoint with a Repurposed Drug Targeting Sigma 1 Receptors. Transl Stroke Res 2024; 15:1035-1049. [PMID: 37704905 DOI: 10.1007/s12975-023-01193-x] [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: 10/27/2022] [Revised: 08/04/2023] [Accepted: 09/01/2023] [Indexed: 09/15/2023]
Abstract
Sigma 1 receptors are intracellular chaperone proteins that have been explored as a subacute treatment to enhance post-stroke recovery. We recently identified the antitussive oxeladin as a selective sigma 1 receptor agonist with the ability to stimulate the release of brain-derived neurotrophic factor from neurons in vitro. In this study, we hypothesized that oral oxeladin citrate would stimulate BDNF secretion and improve stroke outcomes when administered to male rats starting 48 h after transient middle cerebral artery occlusion. Oxeladin did not alter blood clotting and crossed the blood brain barrier within 30 min of oral administration. Rats underwent 90 min of transient middle cerebral artery occlusion. Forty-eight hours later rats began receiving daily oxeladin (135 mg/kg) for 11 days. Oxeladin significantly improved neurological function on days 3, 7, and 14 following MCAO. Infarct size was not altered by a single dose, but the final extent of infarct after 14 days was decreased. However, there was no significant reduction in astrogliosis or microgliosis compared to vehicle-treated control rats. In agreement with in vitro studies, oxeladin increased the amount of mature BDNF in the cerebral cortex 2, 6, and 24 h after single oral dose. However, the increase in BDNF did not result in increases in cellular proliferation in the subventricular zone or dentate gyrus when compared to vehicle-treated controls. These results suggest that oxeladin may reduce the extent of infarct expansion in the subacute phase of stroke, although this action does not appear to involve a reduction in inflammation or increased cell proliferation.
Collapse
Affiliation(s)
- Derek A Schreihofer
- Department of Pharmacology and Neuroscience, University of North Texas Helath Science Center, Fort Worth, Texas, 76107, USA.
| | | | - Seongcheol Kim
- Department of Cellular and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Daniel Metzger
- Department of Pharmacology and Neuroscience, University of North Texas Helath Science Center, Fort Worth, Texas, 76107, USA
| | - Anthony Oppong-Gyebi
- Department of Pharmacology and Neuroscience, University of North Texas Helath Science Center, Fort Worth, Texas, 76107, USA
- Cognizant Technology Solutions, 300 Frank W. Burr Blvd, Teaneck, NJ, 07666, USA
| | - Paromita Das-Earl
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, 76107, USA
| | - John A Schetz
- Department of Pharmacology and Neuroscience, University of North Texas Helath Science Center, Fort Worth, Texas, 76107, USA
| |
Collapse
|
9
|
Qian B, Li TY, Zheng ZX, Zhang HY, Xu WQ, Mo SM, Cui JJ, Chen WJ, Lin YC, Lin ZN. The involvement of SigmaR1 K142 degradation mediated by ERAD in neural senescence linked with CdCl 2 exposure. JOURNAL OF HAZARDOUS MATERIALS 2024; 472:134466. [PMID: 38718507 DOI: 10.1016/j.jhazmat.2024.134466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/16/2024] [Accepted: 04/27/2024] [Indexed: 05/30/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Due to its uncertain pathogenesis, there is currently no treatment available for AD. Increasing evidences have linked cellular senescence to AD, although the mechanism triggering cellular senescence in AD requires further exploration. To investigate the involvement of cellular senescence in AD, we explored the effects of cadmium chloride (CdCl2) exposure, one of the potential environmental risk factors for AD, on neuron senescence in vivo and in vitro. β-amyloid (Aβ) and tubulin-associated protein (tau) pathologies were found to be enhanced by CdCl2 exposure in the in vitro models, while p53/p21/Rb cascade-related neuronal senescence pathways were activated. Conversely, the use of melatonin, a cellular senescence inhibitor, or a cadmium ion chelator suppressed CdCl2-induced neuron senescence, along with the Aβ and tau pathologies. Mechanistically, CdCl2 exposure activated the suppressor enhancer Lin-12/Notch 1-like (SEL1L)/HMG-CoA reductase degradation 1 (HRD1)-regulated endoplasmic reticulum-associated degradation (ERAD), which enhanced the ubiquitin degradation of sigma-1 receptor (SigmaR1) by specifically recognizing its K142 site, resulting in the activation of the p53/p21/Rb pathway via the induction of Ca2+ dyshomeostasis and mitochondrial dysfunction. In the in vivo models, the administration of the SigmaR1 agonist ANAVEX2-73 rescues neurobehavioral inhibition and alleviates cellular senescence and AD-like pathology in the brain tissue of CdCl2-exposed mice. Consequently, the present study revealed a novel senescence-associated regulatory route for the SEL1L/HRD1/SigmaR1 axis that affects the pathological progression of CdCl2 exposure-associated AD. CdCl2 exposure activated SEL1L/HRD1-mediated ERAD and promoted the ubiquitinated degradation of SigmaR1, activating p53/p21/Rb pathway-regulated neuronal senescence. The results of the present study suggest that SigmaR1 may function as a neuroprotective biomarker of neuronal senescence, and pharmacological activation of SigmaR1 could be a promising intervention strategy for AD therapy.
Collapse
Affiliation(s)
- Bo Qian
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Ting-Yu Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhao-Xuan Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Han-Yu Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wen-Qi Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Su-Min Mo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jia-Jia Cui
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wei-Jie Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yu-Chun Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Zhong-Ning Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
10
|
Flores R, Iqbal S, Sikazwe D. Phenylacetyl-/Trolox- Amides: Synthesis, Sigma-1, HDAC-6, and Antioxidant Activities. Int J Mol Sci 2023; 24:15295. [PMID: 37894975 PMCID: PMC10607876 DOI: 10.3390/ijms242015295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/07/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
In search of novel multi-mechanistic approaches for treating Alzheimer's disease (AD), we have embarked on synthesizing single small molecules for probing contributory roles of the following combined disease targets: sigma-1 (σ-1), class IIb histone deacetylase-6 (HDAC-6), and oxidative stress (OS). Herein, we report the synthesis and partial evaluation of 20 amides (i.e., phenylacetic and Trolox or 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid derivatives). Target compounds were conveniently synthesized via amidation by either directly reacting acyl chlorides with amines or condensing acids with amines in the presence of coupling agents 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo [4,5-b] pyridinium 3-oxide hexafluorophosphate (HATU) or 1,1'-carbonyldiimidazole (CDI). Overall, this project afforded compound 8 as a promising lead with σ-1 affinity (Ki = 2.1 μM), HDAC-6 (IC50 = 17 nM), and antioxidant (1.92 Trolox antioxidant equivalents or TEs) activities for optimization in ensuing structure-activity relationship (SAR) studies.
Collapse
Affiliation(s)
| | | | - Donald Sikazwe
- Pharmaceutical Sciences Department, Feik School of Pharmacy, University of the Incarnate Word, San Antonio, TX 78209, USA; (R.F.); (S.I.)
| |
Collapse
|
11
|
Siddiqui T, Bhatt LK. Targeting Sigma-1 Receptor: A Promising Strategy in the Treatment of Parkinson's Disease. Neurochem Res 2023; 48:2925-2935. [PMID: 37259012 PMCID: PMC10231286 DOI: 10.1007/s11064-023-03960-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
Parkinson's disease is a neurodegenerative disease affecting mainly the elderly population. It is characterized by the loss of dopaminergic neurons of the substantia nigra pars compacta region. Parkinson's disease patients exhibit motor symptoms like tremors, rigidity, bradykinesia/hypokinesia, and non-motor symptoms like depression, cognitive decline, delusion, and pain. Major pathophysiological factors which contribute to neuron loss include excess/misfolded alpha-synuclein aggregates, microglial cell-mediated neuroinflammation, excitotoxicity, oxidative stress, and defective mitochondrial function. Sigma-1 receptors are molecular chaperones located at mitochondria-associated ER membrane. Their activation (by endogenous ligands or agonists) has shown neuroprotective and neurorestorative effects in various diseases. This review discusses the roles of activated Sig-1 receptors in modulating various pathophysiological features of Parkinson's disease like alpha-synuclein aggregates, neuroinflammation, excitotoxicity, and oxidative stress.
Collapse
Affiliation(s)
- Talha Siddiqui
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|
12
|
Wang J, Barwick SR, Xiao H, Smith SB. Evaluation of the role of Sigma 1 receptor and Cullin3 in retinal photoreceptor cells. Free Radic Biol Med 2023; 205:214-223. [PMID: 37328017 PMCID: PMC10527355 DOI: 10.1016/j.freeradbiomed.2023.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/24/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
Sigma 1 receptor (Sig1R), a pluripotent modulator of cell survival, is neuroprotective in models of retinal degeneration when activated by the high-affinity, high-specificity ligand (+)-pentazocine ((+)-PTZ). The molecular mechanisms of Sig1R-mediated retinal neuroprotection are under investigation. We previously reported that the antioxidant regulatory transcription factor Nrf2 may be involved in Sig1R-mediated retinal photoreceptor cell (PRC) rescue. Cullin 3 (Cul3) is a component of the Nrf2-Keap1 antioxidant pathway and facilitates Nrf2 ubiquitination. Our earlier transcriptome analysis revealed decreased Cul3 in retinas lacking Sig1R. Here, we asked whether Sig1R activation can modulate Cul3 expression in 661 W cone PRCs. Proximity ligation and co-immunoprecipitation (co-IP) showed that Cul3 resides closely to and co-IPs with Sig1R. Activation of Sig1R using (+)-PTZ significantly increased Cul3 at the gene/protein level; silencing Sig1R decreased Cul3 gene/protein levels. Experiments in which Cul3 was silenced in cells exposed to tBHP resulted in increased oxidative stress, which was not attenuated with Sig1R activation by (+)-PTZ, whereas cells transfected with scrambled siRNA (and incubated with tBHP) responded to (+)-PTZ treatment by decreasing levels of oxidative stress. Assessment of mitochondrial respiration and glycolysis revealed significantly improved maximal respiration, spare capacity and glycolytic capacity in oxidatively-stressed cells transfected with scrambled siRNA and treated with (+)-PTZ, but not in (+)-PTZ treated, oxidatively-stressed cells in which Cul3 had been silenced. The data provide the first evidence that Sig1R co-localizes/interacts with Cul3, a key player in the Nrf2-Keap1 antioxidant pathway. The data suggest that the preservation of mitochondrial respiration/glycolytic function and reduction of oxidative stress observed upon activation of Sig1R occur in part in a Cul3-dependent manner.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Shannon R Barwick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
13
|
Wang T, Jia H. The Sigma Receptors in Alzheimer's Disease: New Potential Targets for Diagnosis and Therapy. Int J Mol Sci 2023; 24:12025. [PMID: 37569401 PMCID: PMC10418732 DOI: 10.3390/ijms241512025] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 08/13/2023] Open
Abstract
Sigma (σ) receptors are a class of unique proteins with two subtypes: the sigma-1 (σ1) receptor which is situated at the mitochondria-associated endoplasmic reticulum (ER) membrane (MAM), and the sigma-2 (σ2) receptor, located in the ER-resident membrane. Increasing evidence indicates the involvement of both σ1 and σ2 receptors in the pathogenesis of Alzheimer's disease (AD), and thus these receptors represent two potentially effective biomarkers for emerging AD therapies. The availability of optimal radioligands for positron emission tomography (PET) neuroimaging of the σ1 and σ2 receptors in humans will provide tools to monitor AD progression and treatment outcomes. In this review, we first summarize the significance of both receptors in the pathophysiology of AD and highlight AD therapeutic strategies related to the σ1 and σ2 receptors. We then survey the potential PET radioligands, with an emphasis on the requirements of optimal radioligands for imaging the σ1 or σ2 receptors in humans. Finally, we discuss current challenges in the development of PET radioligands for the σ1 or σ2 receptors, and the opportunities for neuroimaging to elucidate the σ1 and σ2 receptors as novel biomarkers for early AD diagnosis, and for monitoring of disease progression and AD drug efficacy.
Collapse
Affiliation(s)
- Tao Wang
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China;
- Department of Nuclear Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Hongmei Jia
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China;
| |
Collapse
|
14
|
Pergolizzi J, Varrassi G. The Emerging Role of Sigma Receptors in Pain Medicine. Cureus 2023; 15:e42626. [PMID: 37641763 PMCID: PMC10460634 DOI: 10.7759/cureus.42626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023] Open
Abstract
Sigma receptors are protein chaperones with the unexpected characteristic of being activated by ligand binding. As such, they represent intriguing new targets for potential drug development. As a protein chaperone, these "receptors" escort proteins from the endoplasmic reticulum to their destinations and act to correct misfolded proteins. The two subtypes of sigma receptors, named σ1 and σ2, are markedly distinct from each other. Agonists and antagonists at these receptors show promise as new drug targets, addressing a range of diseases including neurodegenerative disorders, cancer, and cardiac disorders, and may also be analgesic agents and rehabilitation drugs for opioid use disorder. As an analgesic, sigma receptors seem to be more effective in treating neuropathic than nociceptive pain. New bifunctional compounds are being developed with opioids, because agents targeting sigma receptors may have an opioid-sparing effect. The pipeline of agents based on the sigma receptors is long and may treat things from Fragile X syndrome to Parkinson's disease to Huntington's disease to cancer. A novel agent ADV502 acts as a high-affinity σ1 antagonist and partial agonist at the µ-opioid receptor and may be an important agent both for the treatment of neuropathic cancer pain and for rehabilitation of opioid use disorder. Since there has been little recent innovation in pain medicine regarding new compounds and drug targets, drugs that affect the sigma receptor system seem promising and encouraging.
Collapse
|
15
|
Alemany-Navarro M, Diz-de Almeida S, Cruz R, Riancho JA, Rojas-Martínez A, Lapunzina P, Flores C, Carracedo A. Psychiatric polygenic risk as a predictor of COVID-19 risk and severity: insight into the genetic overlap between schizophrenia and COVID-19. Transl Psychiatry 2023; 13:189. [PMID: 37280221 DOI: 10.1038/s41398-023-02482-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/24/2023] [Accepted: 05/23/2023] [Indexed: 06/08/2023] Open
Abstract
Despite the high contagion and mortality rates that have accompanied the coronavirus disease-19 (COVID-19) pandemic, the clinical presentation of the syndrome varies greatly from one individual to another. Potential host factors that accompany greater risk from COVID-19 have been sought and schizophrenia (SCZ) patients seem to present more severe COVID-19 than control counterparts, with certain gene expression similarities between psychiatric and COVID-19 patients reported. We used summary statistics from the last SCZ, bipolar disorder (BD), and depression (DEP) meta-analyses available on the Psychiatric Genomics Consortium webpage to calculate polygenic risk scores (PRSs) for a target sample of 11,977 COVID-19 cases and 5943 subjects with unknown COVID-19 status. Linkage disequilibrium score (LDSC) regression analysis was performed when positive associations were obtained from the PRS analysis. The SCZ PRS was a significant predictor in the case/control, symptomatic/asymptomatic, and hospitalization/no hospitalization analyses in the total and female samples; and of symptomatic/asymptomatic status in men. No significant associations were found for the BD or DEP PRS or in the LDSC regression analysis. SNP-based genetic risk for SCZ, but not for BD or DEP, may be associated with higher risk of SARS-CoV-2 infection and COVID-19 severity, especially among women; however, predictive accuracy barely exceeded chance level. We believe that the inclusion of sexual loci and rare variations in the analysis of genomic overlap between SCZ and COVID-19 will help to elucidate the genetic commonalities between these conditions.
Collapse
Affiliation(s)
- M Alemany-Navarro
- IBIS (Universidad de Sevilla, HUVR, Junta de Andalucia, CSIC), Sevilla, Spain.
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain.
- Fundación Pública Galega de Medicina Xenómica, Sistema Galego de Saúde (SERGAS) Santiago de Compostela, Santiago de Compostela, Spain.
- Grupo de Genética. Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain.
| | - S Diz-de Almeida
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), Instituto de Salud Carlos III, Madrid, Spain
| | - R Cruz
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), Instituto de Salud Carlos III, Madrid, Spain
| | - J A Riancho
- IDIVAL, Cantabria, Spain
- Universidad de Cantabria, Cantabria, Spain
- Hospital U M Valdecilla, Cantabria, Spain
| | - A Rojas-Martínez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| | - P Lapunzina
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Genética Médica y Molecular (INGEMM) del Hospital Universitario La Paz, Madrid, Spain
- ERN-ITHACA-European Reference Network, Santa Cruz de Tenerife, Canarias, Spain
| | - C Flores
- Research Unit, Hospital Universitario N.S. de Candelaria, Santa Cruz de Tenerife, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- Department of Clinical Sciences, University Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - A Carracedo
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica, Sistema Galego de Saúde (SERGAS) Santiago de Compostela, Santiago de Compostela, Spain
- Grupo de Genética. Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
16
|
Dalenogare DP, Souza Monteiro de Araújo D, Landini L, Titiz M, De Siena G, De Logu F, Geppetti P, Nassini R, Trevisan G. Neuropathic-like Nociception and Spinal Cord Neuroinflammation Are Dependent on the TRPA1 Channel in Multiple Sclerosis Models in Mice. Cells 2023; 12:1511. [PMID: 37296632 PMCID: PMC10252670 DOI: 10.3390/cells12111511] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/08/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Background: Transient receptor potential ankyrin 1 (TRPA1) activation is implicated in neuropathic pain-like symptoms. However, whether TRPA1 is solely implicated in pain-signaling or contributes to neuroinflammation in multiple sclerosis (MS) is unknown. Here, we evaluated the TRPA1 role in neuroinflammation underlying pain-like symptoms using two different models of MS. Methods: Using a myelin antigen, Trpa1+/+ or Trpa1-/- female mice developed relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE) (Quil A as adjuvant) or progressive experimental autoimmune encephalomyelitis (PMS)-EAE (complete Freund's adjuvant). The locomotor performance, clinical scores, mechanical/cold allodynia, and neuroinflammatory MS markers were evaluated. Results: Mechanical and cold allodynia detected in RR-EAE, or PMS-EAE Trpa1+/+ mice, were not observed in Trpa1-/- mice. The increased number of cells labeled for ionized calcium-binding adapter molecule 1 (Iba1) or glial fibrillary acidic protein (GFAP), two neuroinflammatory markers in the spinal cord observed in both RR-EAE or PMS-EAE Trpa1+/+ mice, was reduced in Trpa1-/- mice. By Olig2 marker and luxol fast blue staining, prevention of the demyelinating process in Trpa1-/- induced mice was also detected. Conclusions: Present results indicate that the proalgesic role of TRPA1 in EAE mouse models is primarily mediated by its ability to promote spinal neuroinflammation and further strengthen the channel inhibition to treat neuropathic pain in MS.
Collapse
Affiliation(s)
- Diéssica Padilha Dalenogare
- Graduated Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, RS, Brazil;
| | - Daniel Souza Monteiro de Araújo
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Lorenzo Landini
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Mustafa Titiz
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Gaetano De Siena
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Francesco De Logu
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Pierangelo Geppetti
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Romina Nassini
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Gabriela Trevisan
- Graduated Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, RS, Brazil;
| |
Collapse
|
17
|
Vavers E, Zvejniece L, Dambrova M. Sigma-1 receptor and seizures. Pharmacol Res 2023; 191:106771. [PMID: 37068533 PMCID: PMC10176040 DOI: 10.1016/j.phrs.2023.106771] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 04/19/2023]
Abstract
Over the last decade, sigma-1 receptor (Sig1R) has been recognized as a valid target for the treatment of seizure disorders and seizure-related comorbidities. Clinical trials with Sig1R ligands are underway testing therapies for the treatment of drug-resistant seizures, developmental and epileptic encephalopathies, and photosensitive epilepsy. However, the direct molecular mechanism by which Sig1R modulates seizures and the balance between excitatory and inhibitory pathways has not been fully elucidated. This review article aims to summarize existing knowledge of Sig1R and its involvement in seizures by focusing on the evidence obtained from Sig1R knockout animals and the anti-seizure effects of Sig1R ligands. In addition, this review article includes a discussion of the advantages and disadvantages of the use of existing compounds and describes the challenges and future perspectives on the use of Sig1R as a target for the treatment of seizure disorders.
Collapse
Affiliation(s)
- Edijs Vavers
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles 21, LV-1006, Riga, Latvia; University of Tartu, Faculty of Science and Technology, Institute of Chemistry, Ravila 14a, 50411, Tartu, Estonia.
| | - Liga Zvejniece
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles 21, LV-1006, Riga, Latvia
| | - Maija Dambrova
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles 21, LV-1006, Riga, Latvia; Riga Stradiņš University, Faculty of Pharmacy, Konsula 21, LV-1007, Riga, Latvia
| |
Collapse
|
18
|
Pascarella G, Antonelli L, Narzi D, Battista T, Fiorillo A, Colotti G, Guidoni L, Morea V, Ilari A. Investigation of the Entry Pathway and Molecular Nature of σ1 Receptor Ligands. Int J Mol Sci 2023; 24:ijms24076367. [PMID: 37047338 PMCID: PMC10094450 DOI: 10.3390/ijms24076367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
The σ1 receptor (σ1-R) is an enigmatic endoplasmic reticulum resident transmembrane protein implicated in a variety of central nervous system disorders and whose agonists have neuroprotective activity. In spite of σ1-R’s physio-pathological and pharmacological importance, two of the most important features required to fully understand σ1-R function, namely the receptor endogenous ligand(s) and the molecular mechanism of ligand access to the binding site, have not yet been unequivocally determined. In this work, we performed molecular dynamics (MD) simulations to help clarify the potential route of access of ligand(s) to the σ1-R binding site, on which discordant results had been reported in the literature. Further, we combined computational and experimental procedures (i.e., virtual screening (VS), electron density map fitting and fluorescence titration experiments) to provide indications about the nature of σ1-R endogenous ligand(s). Our MD simulations on human σ1-R suggested that ligands access the binding site through a cavity that opens on the protein surface in contact with the membrane, in agreement with previous experimental studies on σ1-R from Xenopus laevis. Additionally, steroids were found to be among the preferred σ1-R ligands predicted by VS, and 16,17-didehydroprogesterone was shown by fluorescence titration to bind human σ1-R, with significantly higher affinity than the prototypic σ1-R ligand pridopidine in the same essay. These results support the hypothesis that steroids are among the most important physiological σ1-R ligands.
Collapse
Affiliation(s)
- Gianmarco Pascarella
- Institute of Molecular Biology and Pathology (IBPM), National Research Council of Italy (CNR), 00185 Rome, Italy (A.I.)
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Roma, Italy
| | - Lorenzo Antonelli
- Institute of Molecular Biology and Pathology (IBPM), National Research Council of Italy (CNR), 00185 Rome, Italy (A.I.)
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Roma, Italy
| | - Daniele Narzi
- Department of Physical and Chemical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Correspondence: (D.N.); (V.M.)
| | - Theo Battista
- Protein Production Facility, Structural Biology Laboratory, Elettra Sincrotrone Trieste, 34149 Basovizza, Italy
| | - Annarita Fiorillo
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Roma, Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology (IBPM), National Research Council of Italy (CNR), 00185 Rome, Italy (A.I.)
| | - Leonardo Guidoni
- Department of Physical and Chemical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Veronica Morea
- Institute of Molecular Biology and Pathology (IBPM), National Research Council of Italy (CNR), 00185 Rome, Italy (A.I.)
- Correspondence: (D.N.); (V.M.)
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology (IBPM), National Research Council of Italy (CNR), 00185 Rome, Italy (A.I.)
| |
Collapse
|
19
|
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
|
20
|
Thapa R, Flores R, Cheng KH, Mochona B, Sikazwe D. Design and Synthesis of New Acyl Urea Analogs as Potential σ1R Ligands. Molecules 2023; 28:2319. [PMID: 36903567 PMCID: PMC10005056 DOI: 10.3390/molecules28052319] [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: 02/07/2023] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
In search of synthetically accessible open-ring analogs of PD144418 or 5-(1-propyl-1,2,5,6-tetrahydropyridin-3-yl)-3-(p-tolyl)isoxazole, a highly potent sigma-1 receptor (σ1R) ligand, we herein report the design and synthesis of sixteen arylated acyl urea derivatives. Design aspects included modeling the target compounds for drug-likeness, docking at σ1R crystal structure 5HK1, and contrasting the lower energy molecular conformers with that of the receptor-embedded PD144418-a molecule we opined that our compounds could mimic pharmacologically. Synthesis of our acyl urea target compounds was achieved in two facile steps which involved first generating the N-(phenoxycarbonyl) benzamide intermediate and then coupling it with the appropriate amines weakly to strongly nucleophilic amines. Two potential leads (compounds 10 and 12, with respective in vitro σ1R binding affinities of 2.18 and 9.54 μM) emerged from this series. These leads will undergo further structure optimization with the ultimate goal of developing novel σ1R ligands for testing in neurodegeneration models of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Rajesh Thapa
- Pharmaceutical Sciences Department, Feik School of Pharmacy, University of the Incarnate Word, San Antonio, TX 78209, USA
| | - Rafael Flores
- Pharmaceutical Sciences Department, Feik School of Pharmacy, University of the Incarnate Word, San Antonio, TX 78209, USA
| | - Kwan H. Cheng
- Department of Physics and Astronomy and Neuroscience Program, Trinity University, San Antonio, TX 78212, USA
| | - Bereket Mochona
- Department of Chemistry, Florida A&M University, Tallahassee, FL 32307, USA
| | - Donald Sikazwe
- Pharmaceutical Sciences Department, Feik School of Pharmacy, University of the Incarnate Word, San Antonio, TX 78209, USA
| |
Collapse
|
21
|
Piechal A, Jakimiuk A, Pyrzanowska J, Blecharz-Klin K, Joniec-Maciejak I, Wiercińska-Drapało A, Mirowska-Guzel D, Widy-Tyszkiewicz E. Long-term Administration of 3-Di-O-Tolylguanidine Modulates Spatial Learning and Memory in Rats and Causes Transition in the Concentration of Neurotransmitters in the Hippocampus, Prefrontal Cortex and Striatum. Neuroscience 2023; 510:129-146. [PMID: 36493909 DOI: 10.1016/j.neuroscience.2022.11.036] [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: 07/31/2022] [Revised: 11/14/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
The sigma-1 and sigma-2 (σ1 and σ2) receptors are found in high concentrations in the brain, and their altered expression leads to a variety of neuropsychiatric disorders. 3-di-tolylguanidine (DTG) stimulates the activity of both of these receptors. We assessed the effects of administering DTG to adult male Sprague Dawley rats on learning and memory consolidation processes and on the levels of neurotransmitters in selected brain structures. Spatial learning and memory were evaluated in the water maze test. The DTG was administered orally at daily doses of 3 mg/kg (DTG3), 10 mg/kg (DTG10) or 30 mg/kg (DTG30) for 10 weeks before and during the water-maze test. After completion of the experiment, the concentration of monoamines and their metabolites as well as amino acids in structures involved in cognitive performance - the hippocampus, prefrontal cortex, and striatum - were determined using high performance liquid chromatography (HPLC). The DTG10 group showed an improvement in memory processes related to the "new" platform location, whereas the DTG30 group was worse at finding the "old" platform location. Since the administration of DTG led to differences in dopaminergic transmission, it was assumed to influence memory processes in this way. Changes in histidine, serine, alanine, taurine, and glutamic acid levels in selected structures of the brains of rats with memory impairment were also observed. We conclude that long-term administration of DTG modulates spatial learning and memory in rats and changes the concentrations of neurotransmitters in the hippocampus, prefrontal cortex, and striatum..
Collapse
Affiliation(s)
- Agnieszka Piechal
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Alicja Jakimiuk
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Justyna Pyrzanowska
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland.
| | - Kamilla Blecharz-Klin
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Alicja Wiercińska-Drapało
- Department of Infectious and Tropical Diseases and Hepatology, Medical University of Warsaw, Wolska 37, 01-201 Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Ewa Widy-Tyszkiewicz
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| |
Collapse
|
22
|
Munguia-Galaviz FJ, Miranda-Diaz AG, Cardenas-Sosa MA, Echavarria R. Sigma-1 Receptor Signaling: In Search of New Therapeutic Alternatives for Cardiovascular and Renal Diseases. Int J Mol Sci 2023; 24:ijms24031997. [PMID: 36768323 PMCID: PMC9916216 DOI: 10.3390/ijms24031997] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Cardiovascular and renal diseases are among the leading causes of death worldwide, and regardless of current efforts, there is a demanding need for therapeutic alternatives to reduce their progression to advanced stages. The stress caused by diseases leads to the activation of protective mechanisms in the cell, including chaperone proteins. The Sigma-1 receptor (Sig-1R) is a ligand-operated chaperone protein that modulates signal transduction during cellular stress processes. Sig-1R interacts with various ligands and proteins to elicit distinct cellular responses, thus, making it a potential target for pharmacological modulation. Furthermore, Sig-1R ligands activate signaling pathways that promote cardioprotection, ameliorate ischemic injury, and drive myofibroblast activation and fibrosis. The role of Sig-1R in diseases has also made it a point of interest in developing clinical trials for pain, neurodegeneration, ischemic stroke, depression in patients with heart failure, and COVID-19. Sig-1R ligands in preclinical models have significantly beneficial effects associated with improved cardiac function, ventricular remodeling, hypertrophy reduction, and, in the kidney, reduced ischemic damage. These basic discoveries could inform clinical trials for heart failure (HF), myocardial hypertrophy, acute kidney injury (AKI), and chronic kidney disease (CKD). Here, we review Sig-1R signaling pathways and the evidence of Sig-1R modulation in preclinical cardiac and renal injury models to support the potential therapeutic use of Sig-1R agonists and antagonists in these diseases.
Collapse
Affiliation(s)
- Francisco Javier Munguia-Galaviz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Division de Ciencias de la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzman 49000, Jalisco, Mexico
| | - Alejandra Guillermina Miranda-Diaz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Miguel Alejandro Cardenas-Sosa
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Raquel Echavarria
- CONACYT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
- Correspondence:
| |
Collapse
|
23
|
Atakpa-Adaji P, Ivanova A. IP 3R at ER-Mitochondrial Contact Sites: Beyond the IP 3R-GRP75-VDAC1 Ca 2+ Funnel. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2023; 6:25152564231181020. [PMID: 37426575 PMCID: PMC10328019 DOI: 10.1177/25152564231181020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/23/2023] [Indexed: 07/11/2023]
Abstract
Membrane contact sites (MCS) circumvent the topological constraints of functional coupling between different membrane-bound organelles by providing a means of communication and exchange of materials. One of the most characterised contact sites in the cell is that between the endoplasmic reticulum and the mitochondrial (ERMCS) whose function is to couple cellular Ca2+ homeostasis and mitochondrial function. Inositol 1,4,5-trisphosphate receptors (IP3Rs) on the ER, glucose-regulated protein 75 (GRP 75) and voltage-dependent anion channel 1 (VDAC1) on the outer mitochondrial membrane are the canonical component of the Ca2+ transfer unit at ERMCS. These are often reported to form a Ca2+ funnel that fuels the mitochondrial low-affinity Ca2+ uptake system. We assess the available evidence on the IP3R subtype selectivity at the ERMCS and consider if IP3Rs have other roles at the ERMCS beyond providing Ca2+. Growing evidence suggests that all three IP3R subtypes can localise and regulate Ca2+ signalling at ERMCS. Furthermore, IP3Rs may be structurally important for assembly of the ERMCS in addition to their role in providing Ca2+ at these sites. Evidence that various binding partners regulate the assembly and Ca2+ transfer at ERMCS populated by IP3R-GRP75-VDAC1, suggesting that cells have evolved mechanisms that stabilise these junctions forming a Ca2+ microdomain that is required to fuel mitochondrial Ca2+ uptake.
Collapse
Affiliation(s)
- Peace Atakpa-Adaji
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Adelina Ivanova
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| |
Collapse
|
24
|
Huang P, Li Z, Peng T, Yang J, Bi L, Huang G, Qiu Y, Yang M, Ye P, Huang M, Jin H, Sun L. Evaluation of [ 18F]F-TZ3108 for PET Imaging of Metabolic-Associated Fatty Liver Disease. Mol Imaging Biol 2022; 24:909-919. [PMID: 35705779 DOI: 10.1007/s11307-022-01740-2] [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: 01/24/2022] [Revised: 04/20/2022] [Accepted: 05/06/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE Sigma-1 receptor (Sig-1R), a chaperone that resides at the mitochondrion-associated endoplasmic reticulum (ER) membrane, is an ER stress biomarker. It is thought that ER stress plays a critical role in the progression of metabolic-associated fatty liver disease (MAFLD). The aim of this study was to evaluate a positron emission tomography (PET) tracer [18F]F-TZ3108 targeting Sig-1R for MAFLD. PROCEDURES The mouse model of MAFLD was established by feeding high-fat diet (HFD) for 12 weeks. Dynamic (0-60 min) PET/CT scans were performed after intravenous injection of 2-deoxy-2[18F]fluoro-D-glucose ([18F]-FDG) and [18F]F-TZ3108. Tracer kinetic modeling was performed for quantification of the PET/CT imaging of the liver. Post-PET biodistribution, the liver tissue western blotting (WB), and immunofluorescence (IF) were performed to compare the expression of Sig-1R levels in the organs harvested from both MAFLD and age-matched control mice. RESULTS The micro PET/CT imaging revealed a significantly decreased uptake of [18F]F-TZ3108 in the livers of the MAFLD group compared to the healthy controls, while the uptake of [18F]-FDG in the livers was not significantly different between the two groups. Based on the tracer kinetic modeling, the binding disassociate rate (k4) for [18F]F-TZ3108 was significantly increased in MAFLD group compared to healthy controls. The volume distribution (VT), and the non-displacement binding potential (BPND) revealed significantly decrease in MAFLD compared to healthy controls respectively. The post-PET biodistribution (%ID/g) of [18F]F-TZ3108 in the livers of MAFLD mice was significantly reduced nearly twofold than that in the livers of control mice. WB and IF experiments further confirmed the reduction of Sig-1R expression in the MAFLD group. CONCLUSIONS The expression of Sig-1R in the liver, measured by the PET tracer, [18F]F-TZ3108, was significantly decreased in mouse model of MAFLD. The [18F]F-TZ3108 PET/CT imaging may provide a novel means of visualization for ER stress in MAFLD or other diseases in vivo.
Collapse
Affiliation(s)
- Peiyi Huang
- Department of Endocrinology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Zhijun Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Tukang Peng
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Jihua Yang
- Department of Endocrinology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Lei Bi
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Guolong Huang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Yifan Qiu
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Min Yang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Peizhen Ye
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Mingxing Huang
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Hongjun Jin
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China.
| | - Liao Sun
- Department of Endocrinology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China.
| |
Collapse
|
25
|
Alfonsetti M, d’Angelo M, Castelli V. Neurotrophic factor-based pharmacological approaches in neurological disorders. Neural Regen Res 2022; 18:1220-1228. [PMID: 36453397 PMCID: PMC9838155 DOI: 10.4103/1673-5374.358619] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Aging is a physiological event dependent on multiple pathways that are linked to lifespan and processes leading to cognitive decline. This process represents the major risk factor for aging-related diseases such as Alzheimer's disease, Parkinson's disease, and ischemic stroke. The incidence of all these pathologies increases exponentially with age. Research on aging biology has currently focused on elucidating molecular mechanisms leading to the development of those pathologies. Cognitive deficit and neurodegeneration, common features of aging-related pathologies, are related to the alteration of the activity and levels of neurotrophic factors, such as brain-derived neurotrophic factor, nerve growth factor, and glial cell-derived neurotrophic factor. For this reason, treatments that modulate neurotrophin levels have acquired a great deal of interest in preventing neurodegeneration and promoting neural regeneration in several neurological diseases. Those treatments include both the direct administration of neurotrophic factors and the induced expression with viral vectors, neurotrophins' binding with biomaterials or other molecules to increase their bioavailability but also cell-based therapies. Considering neurotrophins' crucial role in aging pathologies, here we discuss the involvement of several neurotrophic factors in the most common brain aging-related diseases and the most recent therapeutic approaches that provide direct and sustained neurotrophic support.
Collapse
Affiliation(s)
- Margherita Alfonsetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy,Correspondence to: Vanessa Castelli, .
| |
Collapse
|
26
|
Jin J, Arbez N, Sahn JJ, Lu Y, Linkens KT, Hodges TR, Tang A, Wiseman R, Martin SF, Ross CA. Neuroprotective Effects of σ 2R/TMEM97 Receptor Modulators in the Neuronal Model of Huntington's Disease. ACS Chem Neurosci 2022; 13:2852-2862. [PMID: 36108101 PMCID: PMC9547941 DOI: 10.1021/acschemneuro.2c00274] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Huntington's disease (HD) is a genetic neurodegenerative disease caused by an expanded CAG repeat in the Huntingtin (HTT) gene that encodes for an expanded polyglutamine (polyQ) repeat in exon-1 of the human mutant huntingtin (mHTT) protein. The presence of this polyQ repeat results in neuronal degeneration, for which there is no cure or treatment that modifies disease progression. In previous studies, we have shown that small molecules that bind selectively to σ2R/TMEM97 can have significant neuroprotective effects in models of Alzheimer's disease, traumatic brain injury, and several other neurodegenerative diseases. In the present work, we extend these investigations and show that certain σ2R/TMEM97-selective ligands decrease mHTT-induced neuronal toxicity. We first synthesized a set of compounds designed to bind to σ2R/TMEM97 and determined their binding profiles (Ki values) for σ2R/TMEM97 and other proteins in the central nervous system. Modulators with high affinity and selectivity for σ2R/TMEM97 were then tested in our HD cell model. Primary cortical neurons were cultured in vitro for 7 days and then co-transfected with either a normal HTT construct (Htt N-586-22Q/GFP) or the mHTT construct Htt-N586-82Q/GFP. Transfected neurons were treated with either σ2R/TMEM97 or σ1R modulators for 48 h. After treatment, neurons were fixed and stained with Hoechst, and condensed nuclei were quantified to assess cell death in the transfected neurons. Significantly, σ2R/TMEM97 modulators reduce the neuronal toxicity induced by mHTT, and their neuroprotective effects are not blocked by NE-100, a selective σ1R antagonist known to block neuroprotection by σ1R ligands. These results indicate for the first time that σ2R/TMEM97 modulators can protect neurons from mHTT-induced neuronal toxicity, suggesting that targeting σ2R/TMEM97 may lead to a novel therapeutic approach to treat patients with HD.
Collapse
Affiliation(s)
- Jing Jin
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore Maryland, 21287, United States
| | - Nicolas Arbez
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore Maryland, 21287, United States
- Cellular Sciences Department, IdRS, Croissy-sur-Seine, France
| | - James J. Sahn
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, United States
| | - Yan Lu
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, United States
| | - Kathryn T. Linkens
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, United States
| | - Timothy R. Hodges
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, United States
| | - Anthony Tang
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore Maryland, 21287, United States
| | - Robyn Wiseman
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore Maryland, 21287, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, United States
| | - Stephen F. Martin
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, United States
| | - Christopher A. Ross
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore Maryland, 21287, United States
- Departments of Neurology, Neuroscience and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, United States
| |
Collapse
|
27
|
Turgutalp B, Bhattarai P, Ercetin T, Luise C, Reis R, Gurdal EE, Isaak A, Biriken D, Dinter E, Sipahi H, Schepmann D, Junker A, Wünsch B, Sippl W, Gulcan HO, Kizil C, Yarim M. Discovery of Potent Cholinesterase Inhibition-Based Multi-Target-Directed Lead Compounds for Synaptoprotection in Alzheimer's Disease. J Med Chem 2022; 65:12292-12318. [PMID: 36084304 DOI: 10.1021/acs.jmedchem.2c01003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Drug development efforts that focused on single targets failed to provide effective treatment for Alzheimer's disease (AD). Therefore, we designed cholinesterase inhibition (ChEI)-based multi-target-directed ligands (MTDLs) to simultaneously target AD-related receptors. We built a library of 70 compounds, sequentially screened for ChEI, and determined σ1R, σ2R, NMDAR-GluN2B binding affinities, and P2X7R antagonistic activities. Nine fulfilled in silico drug-likeness criteria and did not display toxicity in three cell lines. Seven displayed cytoprotective activity in two stress-induced cellular models. Compared to donepezil, six showed equal/better synaptic protection in a zebrafish model of acute amyloidosis-induced synaptic degeneration. Two P2X7R antagonists alleviated the activation state of microglia in vivo. Permeability studies were performed, and four did not inhibit CYP450 3A4, 2D6, and 2C9. Therefore, four ChEI-based lead MTDLs are promising drug candidates for synaptic integrity protection and could serve as disease-modifying AD treatment. Our study also proposes zebrafish as a useful preclinical tool for drug discovery and development.
Collapse
Affiliation(s)
- Bengisu Turgutalp
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, 34755 Istanbul, Turkey.,German Centre for Neurodegenerative Diseases (DZNE), Helmholtz Association, 01307 Dresden, Germany
| | - Prabesh Bhattarai
- German Centre for Neurodegenerative Diseases (DZNE), Helmholtz Association, 01307 Dresden, Germany.,Department of Neurology, Columbia University Irving Medical Center, 10032 New York, United States
| | - Tugba Ercetin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Eastern Mediterranean University, TRNC, via Mersin 10, 99628 Famagusta, Turkey
| | - Chiara Luise
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-Universität Halle-Wittenberg, 6099 Halle (Saale), Germany
| | - Rengin Reis
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, 34755 Istanbul, Turkey.,Department of Toxicology, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, 34758 Istanbul, Turkey
| | - Enise Ece Gurdal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, 34755 Istanbul, Turkey.,Institute of Chemistry, Martin-Luther-Universität Halle-Wittenberg, 06120 Halle, Germany
| | - Andreas Isaak
- European Institute for Molecular Imaging (EIMI), der Westfälischen Wilhelms-Universität, D-48149 Münster, Germany
| | - Derya Biriken
- German Centre for Neurodegenerative Diseases (DZNE), Helmholtz Association, 01307 Dresden, Germany.,Department of Medical Microbiology, Ankara University Faculty of Medicine, 06620 Ankara, Turkey
| | - Elisabeth Dinter
- German Centre for Neurodegenerative Diseases (DZNE), Helmholtz Association, 01307 Dresden, Germany.,Department of Neurology, University Clinic, TU Dresden, 01307 Dresden, Germany
| | - Hande Sipahi
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, 34755 Istanbul, Turkey
| | - Dirk Schepmann
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, D-48149 Münster, Germany
| | - Anna Junker
- European Institute for Molecular Imaging (EIMI), der Westfälischen Wilhelms-Universität, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, D-48149 Münster, Germany
| | - Wolfgang Sippl
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-Universität Halle-Wittenberg, 6099 Halle (Saale), Germany
| | - Hayrettin Ozan Gulcan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Eastern Mediterranean University, TRNC, via Mersin 10, 99628 Famagusta, Turkey
| | - Caghan Kizil
- German Centre for Neurodegenerative Diseases (DZNE), Helmholtz Association, 01307 Dresden, Germany.,Department of Neurology, Columbia University Irving Medical Center, 10032 New York, United States
| | - Mine Yarim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, 34755 Istanbul, Turkey
| |
Collapse
|
28
|
Seo HI, Kwon SC, Kwak JY. Protective effects of sigma 1 receptor agonist PRE084 on 2,4,6-trinitrobenzene sulfonic acid-induced experimental colitis in mice. Ann Surg Treat Res 2022; 103:160-168. [PMID: 36128036 PMCID: PMC9478428 DOI: 10.4174/astr.2022.103.3.160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose We aimed to investigate the protective effect of sigma 1 receptor agonist and antagonist, PRE084 and BD1047, respectively, on 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice. Methods Thirty male ICR mice were randomly divided into 5 groups: control, 50% ethanol, colitis, PRE084 + colitis, and combined (PRE084 + BD1047 + colitis). Colitis was induced by intrarectal administration of TNBS. PRE084 and BD1047 were injected daily, starting 3 days before colitis induction. Distal colon tissue was excised for histopathological evaluation, and levels of glutathione (GSH), superoxide dismutase (SOD), myeloperoxidase (MPO), and lipid peroxidation were determined. Results Colitis caused weight loss, mucosal damage, upregulation of tumor necrosis factor-α, interleukin (IL)-1β, IL-6, MPO, and thiobarbituric acid reactive substance activities, and downregulation of GSH and SOD activities. These changes caused by TNBS-induced colitis were significantly ameliorated by PRE084 pretreatment. However, the combined pretreatment with BD1047 significantly attenuated the protective effect of PRE084, thereby reverting to the colitis-induced state. Conclusion We conclude that the sigma 1 receptor agonist PRE084 exhibits significant protective effects against TNBS-induced colitis, which appears to be at least partly mediated by the inhibition of inflammation and oxidative stress, and enhancement of antioxidant properties. Collectively, these results suggest that PRE084 might be an effective drug for the treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Hyun Il Seo
- Department of Internal Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea
| | - Seong Chun Kwon
- Department of Physiology, Catholic Kwandong University College of Medicine, Gangneung, Korea
| | - Jae Young Kwak
- Department of Surgery, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea
| |
Collapse
|
29
|
Estévez-Silva HM, Cuesto G, Romero N, Brito-Armas JM, Acevedo-Arozena A, Acebes Á, Marcellino DJ. Pridopidine Promotes Synaptogenesis and Reduces Spatial Memory Deficits in the Alzheimer's Disease APP/PS1 Mouse Model. Neurotherapeutics 2022; 19:1566-1587. [PMID: 35917088 PMCID: PMC9606189 DOI: 10.1007/s13311-022-01280-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2022] [Indexed: 10/16/2022] Open
Abstract
Sigma-1 receptor agonists have recently gained a great deal of interest due to their anti-amnesic, neuroprotective, and neurorestorative properties. Compounds such as PRE-084 or pridopidine (ACR16) are being studied as a potential treatment against cognitive decline associated with neurodegenerative disease, also to include Alzheimer's disease. Here, we performed in vitro experiments using primary neuronal cell cultures from rats to evaluate the abilities of ACR16 and PRE-084 to induce new synapses and spines formation, analyzing the expression of the possible genes and proteins involved. We additionally examined their neuroprotective properties against neuronal death mediated by oxidative stress and excitotoxicity. Both ACR16 and PRE-084 exhibited a concentration-dependent neuroprotective effect against NMDA- and H2O2-related toxicity, in addition to promoting the formation of new synapses and dendritic spines. However, only ACR16 generated dendritic spines involved in new synapse establishment, maintaining a more expanded activation of MAPK/ERK and PI3K/Akt signaling cascades. Consequently, ACR16 was also evaluated in vivo, and a dose of 1.5 mg/kg/day was administered intraperitoneally in APP/PS1 mice before performing the Morris water maze. ACR16 diminished the spatial learning and memory deficits observed in APP/PS1 transgenic mice via PI3K/Akt pathway activation. These data point to ACR16 as a pharmacological tool to prevent synapse loss and memory deficits associated with Alzheimer's disease, due to its neuroprotective properties against oxidative stress and excitotoxicity, as well as the promotion of new synapses and spines through a mechanism that involves AKT and ERK signaling pathways.
Collapse
Affiliation(s)
- Héctor M Estévez-Silva
- Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Germán Cuesto
- Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Ninovska Romero
- Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
| | - José Miguel Brito-Armas
- Unidad de Investigación, Hospital Universitario de Canarias, ITB-ULL/CIBERNED, Tenerife, Spain
| | - Abraham Acevedo-Arozena
- Unidad de Investigación, Hospital Universitario de Canarias, ITB-ULL/CIBERNED, Tenerife, Spain
| | - Ángel Acebes
- Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain.
| | | |
Collapse
|
30
|
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
|
31
|
Ivanov SV, Ostrovskaya RU. Neuroprotective substances: are they able to protect the pancreatic beta-cells too? Endocr Metab Immune Disord Drug Targets 2022; 22:834-841. [PMID: 35240968 DOI: 10.2174/1871530322666220303162844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/01/2021] [Accepted: 12/30/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Growing evidences demonstrate a close relationship between type 2 diabetes (T2D) and neurodegenerative disorders such as Alzheimer's disease. The similarity of physiological and pathological processes, occurring in pancreatic β-cells and neurons over the course of these pathologies, allows to raise the question of the practicability of studying neuroprotective substances for their potential antidiabetic activity. OBJECTIVE This review analyzes studies of antidiabetic and cytoprotective action on pancreatic β-cells of the neuroprotective compounds that can attenuate the oxidative stress and enhance the expression of neurotrophins: low-molecular-weight NGF mimetic compound GK-2, selective anxiolytic afobazole, antidepressants lithium chloride and lithium carbonate on the rat streptozotocin model of T2D. RESULTS It was found that all above-listed neuroprotective substances have a pronounced antidiabetic activity. The decrease in the β-cells number, the average area of the pancreatic islets, as well as the violation of their morphological structure caused by the streptozotocin was significantly weakened by the therapy with the investigated neuroprotective substances. The extent of these morphological changes clearly correlates with the antihyperglycemic effect of these compounds. CONCLUSION The presented data indicate that the neuroprotective substances attenuating the damaging effect of oxidative stress and neurotrophins deficit cannot only protect neurons but also exert their cytoprotective effect towards pancreatic β-cells. These data may provide a theoretical basis for the further study of neuroprotective drugs as potential therapeutic options for T2D prevention and treatment.
Collapse
Affiliation(s)
- Sergei V Ivanov
- Institute of Pharmacology Russian Academy of Medical Sciences Laboratory of Psychopharmacology Russian Federation
| | - Rita U Ostrovskaya
- Laboratory of PsychopharmacologyInstitute of Pharmacology Russian Academy of Medical SciencesRussian
| |
Collapse
|
32
|
Shin SM, Wang F, Qiu C, Itson-Zoske B, Hogan QH, Yu H. Sigma-1 receptor activity in primary sensory neurons is a critical driver of neuropathic pain. Gene Ther 2022; 29:1-15. [PMID: 32424233 PMCID: PMC7671947 DOI: 10.1038/s41434-020-0157-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/21/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022]
Abstract
The Sigma-1 receptor (σ1R) is highly expressed in the primary sensory neurons (PSNs) that are the critical site of initiation and maintenance of pain following peripheral nerve injury. By immunoblot and immunohistochemistry, we observed increased expression of both σ1R and σ1R-binding immunoglobulin protein (BiP) in the lumbar (L) dorsal root ganglia (DRG) ipsilateral to painful neuropathy induced by spared nerve injury (SNI). To evaluate the therapeutic potential of PSN-targeted σ1R inhibition at a selected segmental level, we designed a recombinant adeno-associated viral (AAV) vector expressing a small hairpin RNA (shRNA) against rat σ1R. Injection of this vector into the L4/L5 DRGs induced downregulation of σ1R in DRG neurons of all size groups, while expression of BiP was not affected. This was accompanied by attenuation of SNI-induced cutaneous mechanical and thermal hypersensitivity. Whole-cell current-clamp recordings of dissociated neurons showed that knockdown of σ1R suppressed neuronal excitability, suggesting that σ1R silencing attenuates pain by reversal of injury-induced neuronal hyperexcitability. These findings support a critical role of σ1R in modulating PSN nociceptive functions, and that the nerve injury-induced elevated σ1R activity in the PSNs can be a significant driver of neuropathic pain. Further understanding the role of PSN-σ1R in pain pathology may open routes to exploit this system for DRG-targeted pain therapy.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA
| | - Fei Wang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, PR China
| | - Chensheng Qiu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266000, PR China
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA.
| |
Collapse
|
33
|
Rodríguez LR, Lapeña-Luzón T, Benetó N, Beltran-Beltran V, Pallardó FV, Gonzalez-Cabo P, Navarro JA. Therapeutic Strategies Targeting Mitochondrial Calcium Signaling: A New Hope for Neurological Diseases? Antioxidants (Basel) 2022; 11:antiox11010165. [PMID: 35052668 PMCID: PMC8773297 DOI: 10.3390/antiox11010165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/13/2022] Open
Abstract
Calcium (Ca2+) is a versatile secondary messenger involved in the regulation of a plethora of different signaling pathways for cell maintenance. Specifically, intracellular Ca2+ homeostasis is mainly regulated by the endoplasmic reticulum and the mitochondria, whose Ca2+ exchange is mediated by appositions, termed endoplasmic reticulum-mitochondria-associated membranes (MAMs), formed by proteins resident in both compartments. These tethers are essential to manage the mitochondrial Ca2+ influx that regulates the mitochondrial function of bioenergetics, mitochondrial dynamics, cell death, and oxidative stress. However, alterations of these pathways lead to the development of multiple human diseases, including neurological disorders, such as amyotrophic lateral sclerosis, Friedreich's ataxia, and Charcot-Marie-Tooth. A common hallmark in these disorders is mitochondrial dysfunction, associated with abnormal mitochondrial Ca2+ handling that contributes to neurodegeneration. In this work, we highlight the importance of Ca2+ signaling in mitochondria and how the mechanism of communication in MAMs is pivotal for mitochondrial maintenance and cell homeostasis. Lately, we outstand potential targets located in MAMs by addressing different therapeutic strategies focused on restoring mitochondrial Ca2+ uptake as an emergent approach for neurological diseases.
Collapse
Affiliation(s)
- Laura R. Rodríguez
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain; (T.L.-L.); (N.B.); (V.B.-B.); (F.V.P.)
- Associated Unit for Rare Diseases INCLIVA-CIPF, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
- Correspondence: (L.R.R.); (P.G.-C.); (J.A.N.)
| | - Tamara Lapeña-Luzón
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain; (T.L.-L.); (N.B.); (V.B.-B.); (F.V.P.)
- Associated Unit for Rare Diseases INCLIVA-CIPF, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Noelia Benetó
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain; (T.L.-L.); (N.B.); (V.B.-B.); (F.V.P.)
- Associated Unit for Rare Diseases INCLIVA-CIPF, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Vicent Beltran-Beltran
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain; (T.L.-L.); (N.B.); (V.B.-B.); (F.V.P.)
| | - Federico V. Pallardó
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain; (T.L.-L.); (N.B.); (V.B.-B.); (F.V.P.)
- Associated Unit for Rare Diseases INCLIVA-CIPF, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Pilar Gonzalez-Cabo
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain; (T.L.-L.); (N.B.); (V.B.-B.); (F.V.P.)
- Associated Unit for Rare Diseases INCLIVA-CIPF, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
- Correspondence: (L.R.R.); (P.G.-C.); (J.A.N.)
| | - Juan Antonio Navarro
- Department of Genetics, Universitat de València-INCLIVA, 46100 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- Correspondence: (L.R.R.); (P.G.-C.); (J.A.N.)
| |
Collapse
|
34
|
Administration of N,N-dimethyltryptamine (DMT) in psychedelic therapeutics and research and the study of endogenous DMT. Psychopharmacology (Berl) 2022; 239:1749-1763. [PMID: 35064294 PMCID: PMC8782705 DOI: 10.1007/s00213-022-06065-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/11/2022] [Indexed: 12/20/2022]
Abstract
As with all drugs, the route, form, and/or dose of a substance administered or applied can play a defining role in its overall pharmacology and use as a therapeutic. This review will focus on these factors as they relate to the psychedelic N,N-dimethyltryptamine (DMT). It will examine the positive and negative aspects of different formulations and routes of administration of DMT and the observed effects from such administrations in the form of ayahuasca teas; oral "pharmahuasca"; injections by intravenous (IV) and intramuscular (IM) routes; inhalation, insufflation; and other routes; and high-dose, low-dose, and "micro-dose" effects. The review will consider possible oral route of administration alternatives that would not require concomitant use of a monoamine oxidase inhibitor. The review will then address the current research findings for DMT from in vivo and in vitro studies as well as the possibility that these findings may be revealing the role of endogenous DMT in normal brain function.
Collapse
|
35
|
Gaja-Capdevila N, Hernández N, Navarro X, Herrando-Grabulosa M. Sigma-1 Receptor is a Pharmacological Target to Promote Neuroprotection in the SOD1 G93A ALS Mice. Front Pharmacol 2021; 12:780588. [PMID: 34955848 PMCID: PMC8702863 DOI: 10.3389/fphar.2021.780588] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/08/2021] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disorder characterized by the death of motoneurons (MNs) with a poor prognosis. There is no available cure, thus, novel therapeutic targets are urgently needed. Sigma-1 receptor (Sig-1R) has been reported as a target to treat experimental models of degenerative diseases and, importantly, mutations in the Sig-1R gene cause several types of motoneuron disease (MND). In this study we compared the potential therapeutic effect of three Sig-1R ligands, the agonists PRE-084 and SA4503 and the antagonist BD1063, in the SOD1G93A mouse model of ALS. Pharmacological administration was from 8 to 16 weeks of age, and the neuromuscular function and disease progression were evaluated using nerve conduction and rotarod tests. At the end of follow up (16 weeks), samples were harvested for histological and molecular analyses. The results showed that PRE-084, as well as BD1063 treatment was able to preserve neuromuscular function of the hindlimbs and increased the number of surviving MNs in the treated female SOD1G93A mice. SA4503 tended to improve motor function and preserved neuromuscular junctions (NMJ), but did not improve MN survival. Western blot analyses revealed that the autophagic flux and the endoplasmic reticulum stress, two pathways implicated in the physiopathology of ALS, were not modified with Sig-1R treatments in SOD1G93A mice. In conclusion, Sig-1R ligands are promising tools for ALS treatment, although more research is needed to ascertain their mechanisms of action.
Collapse
Affiliation(s)
- Núria Gaja-Capdevila
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Neus Hernández
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Xavier Navarro
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Mireia Herrando-Grabulosa
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
36
|
Sharma N, Patel C, Shenkman M, Kessel A, Ben-Tal N, Lederkremer GZ. The Sigma-1 receptor is an ER-localized type II membrane protein. J Biol Chem 2021; 297:101299. [PMID: 34648767 PMCID: PMC8561001 DOI: 10.1016/j.jbc.2021.101299] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/06/2021] [Accepted: 10/06/2021] [Indexed: 11/10/2022] Open
Abstract
The Sigma-1 receptor (S1R) is a transmembrane protein with important roles in cellular homeostasis in normal physiology and in disease. Especially in neurodegenerative diseases, S1R activation has been shown to provide neuroprotection by modulating calcium signaling, mitochondrial function and reducing endoplasmic reticulum (ER) stress. S1R missense mutations are one of the causes of the neurodegenerative Amyotrophic Lateral Sclerosis and distal hereditary motor neuronopathies. Although the S1R has been studied intensively, basic aspects remain controversial, such as S1R topology and whether it reaches the plasma membrane. To address these questions, we have undertaken several approaches. C-terminal tagging with a small biotin-acceptor peptide and BirA biotinylation in cells suggested a type II membrane orientation (cytosolic N-terminus). However, N-terminal tagging gave an equal probability for both possible orientations. This might explain conflicting reports in the literature, as tags may affect the protein topology. Therefore, we studied untagged S1R using a protease protection assay and a glycosylation mapping approach, introducing N-glycosylation sites. Both methods provided unambiguous results showing that the S1R is a type II membrane protein with a short cytosolic N-terminal tail. Assessments of glycan processing, surface fluorescence-activated cell sorting, and cell surface biotinylation indicated ER retention, with insignificant exit to the plasma membrane, in the absence or presence of S1R agonists or of ER stress. These findings may have important implications for S1R-based therapeutic approaches.
Collapse
Affiliation(s)
- Neeraj Sharma
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Chaitanya Patel
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Marina Shenkman
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Amit Kessel
- School of Neurobiology, Biochemistry and Biophysics, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nir Ben-Tal
- School of Neurobiology, Biochemistry and Biophysics, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Gerardo Z Lederkremer
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
37
|
Stelfa G, Vavers E, Svalbe B, Serzants R, Miteniece A, Lauberte L, Grinberga S, Gukalova B, Dambrova M, Zvejniece L. Reduced GFAP Expression in Bergmann Glial Cells in the Cerebellum of Sigma-1 Receptor Knockout Mice Determines the Neurobehavioral Outcomes after Traumatic Brain Injury. Int J Mol Sci 2021; 22:11611. [PMID: 34769042 PMCID: PMC8584110 DOI: 10.3390/ijms222111611] [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: 08/30/2021] [Revised: 10/19/2021] [Accepted: 10/23/2021] [Indexed: 11/17/2022] Open
Abstract
Neuroprotective effects of Sigma-1 receptor (S1R) ligands have been observed in multiple animal models of neurodegenerative diseases. Traumatic brain injury (TBI)-related neurodegeneration can induce long-lasting physical, cognitive, and behavioral disabilities. The aim of our study was to evaluate the role of S1R in the development of neurological deficits after TBI. Adult male wild-type CD-1 (WT) and S1R knockout (S1R-/-) mice were subjected to lateral fluid percussion injury, and behavioral and histological outcomes were assessed for up to 12 months postinjury. Neurological deficits and motor coordination impairment were less pronounced in S1R-/- mice with TBI than in WT mice with TBI 24 h after injury. TBI-induced short-term memory impairments were present in WT but not S1R-/- mice 7 months after injury. Compared to WT animals, S1R-/- mice exhibited better motor coordination and less pronounced despair behavior for up to 12 months postinjury. TBI induced astrocyte activation in the cortex of WT but not S1R-/- mice. S1R-/- mice presented a significantly reduced GFAP expression in Bergmann glial cells in the molecular layer of the cerebellum compared to WT mice. Our findings suggest that S1R deficiency reduces TBI-induced motor coordination impairments by reducing GFAP expression in Bergmann glial cells in the cerebellum.
Collapse
Affiliation(s)
- Gundega Stelfa
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, LV-1006 Riga, Latvia; (E.V.); (B.S.); (R.S.); (A.M.); (L.L.); (M.D.); (L.Z.)
- Faculty of Veterinary Medicine, Latvia University of Life Sciences and Technologies, K Helmana Str. 8, LV-3001 Jelgava, Latvia
| | - Edijs Vavers
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, LV-1006 Riga, Latvia; (E.V.); (B.S.); (R.S.); (A.M.); (L.L.); (M.D.); (L.Z.)
| | - Baiba Svalbe
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, LV-1006 Riga, Latvia; (E.V.); (B.S.); (R.S.); (A.M.); (L.L.); (M.D.); (L.Z.)
| | - Rinalds Serzants
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, LV-1006 Riga, Latvia; (E.V.); (B.S.); (R.S.); (A.M.); (L.L.); (M.D.); (L.Z.)
- Department of Pharmaceutical Chemistry, Riga Stradins University, Dzirciema Str. 16, LV-1007 Riga, Latvia
| | - Anna Miteniece
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, LV-1006 Riga, Latvia; (E.V.); (B.S.); (R.S.); (A.M.); (L.L.); (M.D.); (L.Z.)
- Department of Life Sciences, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Lasma Lauberte
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, LV-1006 Riga, Latvia; (E.V.); (B.S.); (R.S.); (A.M.); (L.L.); (M.D.); (L.Z.)
| | - Solveiga Grinberga
- Laboratory of Physical Organic Chemistry, Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, LV-1006 Riga, Latvia; (S.G.); (B.G.)
| | - Baiba Gukalova
- Laboratory of Physical Organic Chemistry, Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, LV-1006 Riga, Latvia; (S.G.); (B.G.)
| | - Maija Dambrova
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, LV-1006 Riga, Latvia; (E.V.); (B.S.); (R.S.); (A.M.); (L.L.); (M.D.); (L.Z.)
- Department of Pharmaceutical Chemistry, Riga Stradins University, Dzirciema Str. 16, LV-1007 Riga, Latvia
| | - Liga Zvejniece
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, LV-1006 Riga, Latvia; (E.V.); (B.S.); (R.S.); (A.M.); (L.L.); (M.D.); (L.Z.)
| |
Collapse
|
38
|
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
|
39
|
Kozlowska U, Nichols C, Wiatr K, Figiel M. From psychiatry to neurology: Psychedelics as prospective therapeutics for neurodegenerative disorders. J Neurochem 2021; 162:89-108. [PMID: 34519052 DOI: 10.1111/jnc.15509] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/30/2021] [Accepted: 09/05/2021] [Indexed: 12/22/2022]
Abstract
The studies of psychedelics, especially psychedelic tryptamines like psilocybin, are rapidly gaining interest in neuroscience research. Much of this interest stems from recent clinical studies demonstrating that they have a unique ability to improve the debilitating symptoms of major depressive disorder (MDD) long-term after only a single treatment. Indeed, the Food and Drug Administration (FDA) has recently designated two Phase III clinical trials studying the ability of psilocybin to treat forms of MDD with "Breakthrough Therapy" status. If successful, the use of psychedelics to treat psychiatric diseases like depression would be revolutionary. As more evidence appears in the scientific literature to support their use in psychiatry to treat MDD on and substance use disorders (SUD), recent studies with rodents revealed that their therapeutic effects might extend beyond treating MDD and SUD. For example, psychedelics may have efficacy in the treatment and prevention of brain injury and neurodegenerative diseases such as Alzheimer's Disease. Preclinical work has highlighted psychedelics' ability to induce neuroplasticity and synaptogenesis, and neural progenitor cell proliferation. Psychedelics may also act as immunomodulators by reducing levels of proinflammatory biomarkers, including IL-1β, IL-6, and tumor necrosis factor-α (TNF-α). Their exact molecular mechanisms, and induction of cellular interactions, especially between neural and glial cells, leading to therapeutic efficacy, remain to be determined. In this review, we discuss recent findings and information on how psychedelics may act therapeutically on cells within the central nervous system (CNS) during brain injuries and neurodegenerative diseases.
Collapse
Affiliation(s)
- Urszula Kozlowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland.,Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Charles Nichols
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Kalina Wiatr
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Maciej Figiel
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
40
|
Abstract
Neuropsychiatric sequalae to coronavirus disease 2019 (COVID-19) infection are beginning to emerge, like previous Spanish influenza and severe acute respiratory syndrome episodes. Streptococcal infection in paediatric patients causing obsessive compulsive disorder (PANDAS) is another recent example of an infection-based psychiatric disorder. Inflammation associated with neuropsychiatric disorders has been previously reported but there is no standard clinical management approach established. Part of the reason is that it is unclear what factors determine the specific neuronal vulnerability and the efficacy of anti-inflammatory treatment in neuroinflammation. The emerging COVID-19 data suggested that in the acute stage, widespread neuronal damage appears to be the result of abnormal and overactive immune responses and cytokine storm is associated with poor prognosis. It is still too early to know if there are long-term-specific neuronal or brain regional damages associated with COVID-19, resulting in distinct neuropsychiatric disorders. In several major psychiatric disorders where neuroinflammation is present, patients with abnormal inflammatory markers may also experience less than favourable response or treatment resistance when standard treatment is used alone. Evidence regarding the benefits of co-administered anti-inflammatory agents such as COX-2 inhibitor is encouraging in selected patients though may not benefit others. Disease-modifying therapies are increasingly being applied to neuropsychiatric diseases characterised by abnormal or hyperreactive immune responses. Adjunct anti-inflammatory treatment may benefit selected patients and is definitely an important component of clinical management in the presence of neuroinflammation.
Collapse
|
41
|
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
|
42
|
Shenkman M, Geva M, Gershoni-Emek N, Hayden MR, Lederkremer GZ. Pridopidine reduces mutant huntingtin-induced endoplasmic reticulum stress by modulation of the Sigma-1 receptor. J Neurochem 2021; 158:467-481. [PMID: 33871049 DOI: 10.1111/jnc.15366] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/18/2020] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
The endoplasmic reticulum (ER)-localized Sigma-1 receptor (S1R) is neuroprotective in models of neurodegenerative diseases, among them Huntington disease (HD). Recent clinical trials in HD patients and preclinical studies in cellular and mouse HD models suggest a therapeutic potential for the high-affinity S1R agonist pridopidine. However, the molecular mechanisms of the cytoprotective effect are unclear. We have previously reported strong induction of ER stress by toxic mutant huntingtin (mHtt) oligomers, which is reduced upon sequestration of these mHtt oligomers into large aggregates. Here, we show that pridopidine significantly ameliorates mHtt-induced ER stress in cellular HD models, starting at low nanomolar concentrations. Pridopidine reduced the levels of markers of the three branches of the unfolded protein response (UPR), showing the strongest effects on the PKR-like endoplasmic reticulum kinase (PERK) branch. The effect is S1R-dependent, as it is abolished in cells expressing mHtt in which the S1R was deleted using CRISPR/Cas9 technology. mHtt increased the level of the detergent-insoluble fraction of S1R, suggesting a compensatory cellular mechanism that responds to increased ER stress. Pridopidine further enhanced the levels of insoluble S1R, suggesting the stabilization of activated S1R oligomers. These S1R oligomeric species appeared in ER-localized patches, and not in the mitochondria-associated membranes nor the ER-derived quality control compartment. The colocalization of S1R with the chaperone BiP was significantly reduced by mHtt, and pridopidine restored this colocalization to normal, unstressed levels. Pridopidine increased toxic oligomeric mHtt recruitment into less toxic large sodium dodecyl sulfate-insoluble aggregates, suggesting that this in turn reduces ER stress and cytotoxicity.
Collapse
Affiliation(s)
- Marina Shenkman
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Michal Geva
- Prilenia Therapeutics Development LTD, Herzliya, Israel
| | | | | | - Gerardo Z Lederkremer
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
43
|
Barwick SR, Siddiq MS, Wang J, Xiao H, Marshall B, Perry E, Smith SB. Sigma 1 Receptor Co-Localizes with NRF2 in Retinal Photoreceptor Cells. Antioxidants (Basel) 2021; 10:antiox10060981. [PMID: 34205384 PMCID: PMC8234060 DOI: 10.3390/antiox10060981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/25/2022] Open
Abstract
Sigma 1 receptor (Sig1R), a modulator of cell survival, has emerged as a novel target for retinal degenerative disease. Studies have shown that activation of Sig1R, using the high affinity ligand (+)-pentazocine ((+)-PTZ), improves cone function in a severe retinopathy model. The rescue is accompanied by normalization of levels of NRF2, a key transcription factor that regulates the antioxidant response. The interaction of Sig1R with a number of proteins has been investigated; whether it interacts with NRF2, however, is not known. We used co-immunoprecipitation (co-IP), proximity ligation assay (PLA), and electron microscopy (EM) immunodetection methods to investigate this question in the 661W cone photoreceptor cell line. For co-IP experiments, immune complexes were precipitated by protein A/G agarose beads and immunodetected using anti-NRF2 antibody. For PLA, cells were incubated with anti-Sig1R polyclonal and anti-NRF2 monoclonal antibodies, then subsequently with (−)-mouse and (+)-rabbit PLA probes. For EM analysis, immuno-EM gold labeling was performed using nanogold-enhanced labeling with anti-NRF2 and anti-Sig1R antibodies, and data were confirmed using colloidal gold labeling. The co-IP experiment suggested that NRF2 was bound in a complex with Sig1R. The PLA assays detected abundant orange fluorescence in cones, indicating that Sig1R and NRF2 were within 40 nm of each other. EM immunodetection confirmed co-localization of Sig1R with NRF2 in cells and in mouse retinal tissue. This study is the first to report co-localization of Sig1R-NRF2 and supports earlier studies implicating modulation of NRF2 as a mechanism by which Sig1R mediates retinal neuroprotection.
Collapse
Affiliation(s)
- Shannon R. Barwick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Mevish S. Siddiq
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
| | - Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Brendan Marshall
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
| | - Elizabeth Perry
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +1-706-721-7392; Fax: +1-706-721-6120
| |
Collapse
|
44
|
Zhemkov V, Ditlev JA, Lee WR, Wilson M, Liou J, Rosen MK, Bezprozvanny I. The role of sigma 1 receptor in organization of endoplasmic reticulum signaling microdomains. eLife 2021; 10:e65192. [PMID: 33973848 PMCID: PMC8112866 DOI: 10.7554/elife.65192] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Sigma 1 receptor (S1R) is a 223-amino-acid-long transmembrane endoplasmic reticulum (ER) protein. S1R modulates activity of multiple effector proteins and is a well-established drug target. However, signaling functions of S1R in cells are poorly understood. Here, we test the hypothesis that biological activity of S1R in cells can be explained by its ability to interact with cholesterol and to form cholesterol-enriched microdomains in the ER membrane. By performing experiments in reduced reconstitution systems, we demonstrate direct effects of cholesterol on S1R clustering. We identify a novel cholesterol-binding motif in the transmembrane region of human S1R. Mutations of this motif impair association of recombinant S1R with cholesterol beads, affect S1R clustering in vitro and disrupt S1R subcellular localization. We demonstrate that S1R-induced membrane microdomains have increased local membrane thickness and that increased local cholesterol concentration and/or membrane thickness in these microdomains can modulate signaling of inositol-requiring enzyme 1α in the ER. Further, S1R agonists cause disruption of S1R clusters, suggesting that biological activity of S1R agonists is linked to remodeling of ER membrane microdomains. Our results provide novel insights into S1R-mediated signaling mechanisms in cells.
Collapse
Affiliation(s)
- Vladimir Zhemkov
- Department of Physiology, UT Southwestern Medical Center at DallasDallasUnited States
| | - Jonathon A Ditlev
- Department of Biophysics, Howard Hughes Medical Institute, UT Southwestern Medical Center at DallasDallasUnited States
| | - Wan-Ru Lee
- Department of Physiology, UT Southwestern Medical Center at DallasDallasUnited States
| | - Mikaela Wilson
- Department of Physiology, UT Southwestern Medical Center at DallasDallasUnited States
| | - Jen Liou
- Department of Physiology, UT Southwestern Medical Center at DallasDallasUnited States
| | - Michael K Rosen
- Department of Biophysics, Howard Hughes Medical Institute, UT Southwestern Medical Center at DallasDallasUnited States
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center at DallasDallasUnited States
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic UniversitySt. PetersburgRussian Federation
| |
Collapse
|
45
|
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
|
46
|
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
|
47
|
Johannessen Landmark C, Potschka H, Auvin S, Wilmshurst JM, Johannessen SI, Kasteleijn-Nolst Trenité D, Wirrell EC. The role of new medical treatments for the management of developmental and epileptic encephalopathies: Novel concepts and results. Epilepsia 2021; 62:857-873. [PMID: 33638459 DOI: 10.1111/epi.16849] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 12/20/2022]
Abstract
Developmental and epileptic encephalopathies (DEEs) are among the most challenging of all epilepsies to manage, given the exceedingly frequent and often severe seizure types, pharmacoresistance to conventional antiseizure medications, and numerous comorbidities. During the past decade, efforts have focused on development of new treatment options for DEEs, with several recently approved in the United States or Europe, including cannabidiol as an orphan drug in Dravet and Lennox-Gastaut syndromes and everolimus as a possible antiepileptogenic and precision drug for tuberous sclerosis complex, with its impact on the mammalian target of rapamycin pathway. Furthermore, fenfluramine, an old drug, was repurposed as a novel therapy in the treatment of Dravet syndrome. The evolution of new insights into pathophysiological processes of various DEEs provides possibilities to investigate novel and repurposed drugs and to place them into the context of their role in future management of these patients. The purpose of this review is to provide an overview of these new medical treatment options for the DEEs and to discuss the clinical implications of these results for improved treatment.
Collapse
Affiliation(s)
- Cecilie Johannessen Landmark
- Program for Pharmacy, Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway.,National Center for Epilepsy, Oslo University Hospital, Oslo, Norway.,Section for Clinical Pharmacology, Department of Pharmacology, Oslo University Hospital, Oslo, Norway
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Stéphane Auvin
- Pediatric Neurology Department, Robert Debré Hospital, Public Hospital Network of Paris, Paris, France.,Mixed Unit of Research NeuroDiderot U1141, University of Paris, Paris, France
| | - Jo M Wilmshurst
- Paediatric Neurology Department, Red Cross War Memorial Children's Hospital, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Svein I Johannessen
- National Center for Epilepsy, Oslo University Hospital, Oslo, Norway.,Section for Clinical Pharmacology, Department of Pharmacology, Oslo University Hospital, Oslo, Norway
| | | | - Elaine C Wirrell
- Divisions of Child and Adolescent Neurology and Epilepsy, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
48
|
Pontisso I, Combettes L. Role of Sigma-1 Receptor in Calcium Modulation: Possible Involvement in Cancer. Genes (Basel) 2021; 12:139. [PMID: 33499031 PMCID: PMC7911422 DOI: 10.3390/genes12020139] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022] Open
Abstract
Ca2+ signaling plays a pivotal role in the control of cellular homeostasis and aberrant regulation of Ca2+ fluxes have a strong impact on cellular functioning. As a consequence of this ubiquitous role, Ca2+ signaling dysregulation is involved in the pathophysiology of multiple diseases including cancer. Indeed, multiple studies have highlighted the role of Ca2+ fluxes in all the steps of cancer progression. In particular, the transfer of Ca2+ at the ER-mitochondrial contact sites, also known as mitochondrial associated membranes (MAMs), has been shown to be crucial for cancer cell survival. One of the proteins enriched at this site is the sigma-1 receptor (S1R), a protein that has been described as a Ca2+-sensitive chaperone that exerts a protective function in cells in various ways, including the modulation of Ca2+ signaling. Interestingly, S1R is overexpressed in many types of cancer even though the exact mechanisms by which it promotes cell survival are not fully elucidated. This review summarizes the findings describing the roles of S1R in the control of Ca2+ signaling and its involvement in cancer progression.
Collapse
Affiliation(s)
- Ilaria Pontisso
- UMR 1282, INSERM, Laboratoire de Biologie et Pharmacologie Appliquée, Ecole Normale Supérieure Paris Saclay, 91190 Gif Sur Yvette, France;
- Faculté des Sciences, Université Paris-Saclay, 91405 Orsay, France
| | - Laurent Combettes
- UMR 1282, INSERM, Laboratoire de Biologie et Pharmacologie Appliquée, Ecole Normale Supérieure Paris Saclay, 91190 Gif Sur Yvette, France;
- Faculté des Sciences, Université Paris-Saclay, 91405 Orsay, France
| |
Collapse
|
49
|
Xie XY, Li YY, Ma WH, Chen AF, Sun YT, Lee JY, Riad A, Xu DH, Mach RH, Huang YS. Synthesis, binding, and functional properties of tetrahydroisoquinolino-2-alkyl phenones as selective σ 2R/TMEM97 ligands. Eur J Med Chem 2021; 209:112906. [PMID: 33049607 DOI: 10.1016/j.ejmech.2020.112906] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/01/2020] [Accepted: 10/03/2020] [Indexed: 02/07/2023]
Abstract
Sigma-2 receptor (σ2R/TMEM97) has been implicated to play important roles in multiple cellular dysfunctions, such as cell neoplastic proliferation, neuro-inflammation, neurodegeneration, etc. Selective σ2 ligands are believed to be promising pharmacological tools to regulate or diagnose various disorders. As an ongoing effort of discovery of new and selective σ2 ligands, we have synthesized a series of tetrahydroisoquinolino-2-alkyl phenone analogs and identified that 10 of them have moderate to potent affinity and selectivity for σ2R/TMEM97. Especially, 4 analogs showed Ki values ranging from 0.38 to 5.1 nM for σ2R/TMEM97 with no or low affinity for sigma-1 receptor (σ1R). Functional assays indicated that these 4 most potent analogs had no effects on intracellular calcium concentration and were classified as putative σ2R/TMEM97 antagonists according to current understanding. The σ2R/TMEM97 has been suggested to play important roles in the central nervous system. Based on published pharmacological and clinical results from several regarded σ2R/TMEM97 antagonists, these analogs may potentially be useful for the treatment of various neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiao-Yang Xie
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Yu-Yun Li
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Wen-Hui Ma
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Ai-Fang Chen
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Yu-Tong Sun
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Ji Youn Lee
- Department of Radiology, University of Pennsylvania, 231 S. 34th St, Philadelphia, PA, 19104, USA
| | - Aladdin Riad
- Department of Radiology, University of Pennsylvania, 231 S. 34th St, Philadelphia, PA, 19104, USA
| | - Dao-Hua Xu
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Robert H Mach
- Department of Radiology, University of Pennsylvania, 231 S. 34th St, Philadelphia, PA, 19104, USA.
| | - Yun-Sheng Huang
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China.
| |
Collapse
|
50
|
Ye N, Qin W, Tian S, Xu Q, Wold EA, Zhou J, Zhen XC. Small Molecules Selectively Targeting Sigma-1 Receptor for the Treatment of Neurological Diseases. J Med Chem 2020; 63:15187-15217. [PMID: 33111525 DOI: 10.1021/acs.jmedchem.0c01192] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The sigma-1 (σ1) receptor, an enigmatic protein originally classified as an opioid receptor subtype, is now understood to possess unique structural and functional features of its own and play critical roles to widely impact signaling transduction by interacting with receptors, ion channels, lipids, and kinases. The σ1 receptor is implicated in modulating learning, memory, emotion, sensory systems, neuronal development, and cognition and accordingly is now an actively pursued drug target for various neurological and neuropsychiatric disorders. Evaluation of the five selective σ1 receptor drug candidates (pridopidine, ANAVEX2-73, SA4503, S1RA, and T-817MA) that have entered clinical trials has shown that reaching clinical approval remains an evasive and important goal. This review provides up-to-date information on the selective targeting of σ1 receptors, including their history, function, reported crystal structures, and roles in neurological diseases, as well as a useful collation of new chemical entities as σ1 selective orthosteric ligands or allosteric modulators.
Collapse
Affiliation(s)
- Na Ye
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wangzhi Qin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Sheng Tian
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Qingfeng Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Eric A Wold
- Chemical Biology Program, Department of Pharmacology and Toxicology, and Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, and Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Xue-Chu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|