51
|
Chaperone-Dependent Mechanisms as a Pharmacological Target for Neuroprotection. Int J Mol Sci 2023; 24:ijms24010823. [PMID: 36614266 PMCID: PMC9820882 DOI: 10.3390/ijms24010823] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
Modern pharmacotherapy of neurodegenerative diseases is predominantly symptomatic and does not allow vicious circles causing disease development to break. Protein misfolding is considered the most important pathogenetic factor of neurodegenerative diseases. Physiological mechanisms related to the function of chaperones, which contribute to the restoration of native conformation of functionally important proteins, evolved evolutionarily. These mechanisms can be considered promising for pharmacological regulation. Therefore, the aim of this review was to analyze the mechanisms of endoplasmic reticulum stress (ER stress) and unfolded protein response (UPR) in the pathogenesis of neurodegenerative diseases. Data on BiP and Sigma1R chaperones in clinical and experimental studies of Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease are presented. The possibility of neuroprotective effect dependent on Sigma1R ligand activation in these diseases is also demonstrated. The interaction between Sigma1R and BiP-associated signaling in the neuroprotection is discussed. The performed analysis suggests the feasibility of pharmacological regulation of chaperone function, possibility of ligand activation of Sigma1R in order to achieve a neuroprotective effect, and the need for further studies of the conjugation of cellular mechanisms controlled by Sigma1R and BiP chaperones.
Collapse
|
52
|
Ajoolabady A, Tang D, Kroemer G, Ren J. Ferroptosis in hepatocellular carcinoma: mechanisms and targeted therapy. Br J Cancer 2023; 128:190-205. [PMID: 36229582 PMCID: PMC9902568 DOI: 10.1038/s41416-022-01998-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/25/2022] [Accepted: 09/22/2022] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma is the most prevalent form of primary liver cancer with a multifactorial aetiology comprising genetic, environmental, and behavioural factors. Evading cell death is a defining hallmark of hepatocellular carcinoma, underpinning tumour growth, progression, and therapy resistance. Ferroptosis is a form of nonapoptotic cell death driven by an array of cellular events, including intracellular iron overload, free radical production, lipid peroxidation and activation of various cell death effectors, ultimately leading to rupture of the plasma membrane. Although induction of ferroptosis is an emerging strategy to suppress hepatocellular carcinoma, malignant cells manage to develop adaptive mechanisms, conferring resistance to ferroptosis and ferroptosis-inducing drugs. Herein, we aim at elucidating molecular mechanisms and signalling pathways involved in ferroptosis and offer our opinions on druggable targets and new therapeutic strategy in an attempt to restrain the growth and progression of hepatocellular carcinoma through induction of ferroptotic cell death.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, China.
| |
Collapse
|
53
|
Mysona BA, Zhao J, De Greef O, Beisel A, Patel PA, Berman L, Smith SB, Bollinger K. Sigma-1 receptor agonist, (+)-pentazocine, is neuroprotective in a Brown Norway rat microbead model of glaucoma. Exp Eye Res 2023; 226:109308. [PMID: 36400283 PMCID: PMC9839578 DOI: 10.1016/j.exer.2022.109308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/23/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022]
Abstract
PURPOSE Glaucoma is a worldwide leading cause of irreversible blindness. Standard treatments lower intraocular pressure (IOP). Novel treatments to prevent optic nerve (ON) degeneration are needed. Here, we investigate the hypothesis that sigma-1 receptor (S1R) agonist (+)-pentazocine (PTZ) is neuroprotective in a Brown Norway (BN) rat, microbead model of glaucoma. METHODS BN rats (9-11 weeks, male and female) were treated by intraperitoneal injection, 3 times per week with (+)-PTZ (2 mg/kg) or vehicle (VEH) alone. Treatment started 1 week prior to intraocular injection of polystyrene microbeads to elevate IOP. IOP was measured 2-3 times per week. Five weeks post microbead injection, rats were euthanized. ONs were removed, then fixed and processed for 63x oil, light microscope imaging of toluidine blue stained ON cross sections. To facilitate comparison of ON morphology from VEH and (+)-PTZ treated rats with similar ocular hypertensive insults, rats were assigned to low (IOP ≤15.8 mmHg), moderate (15.8 < IOP <28.0 mmHg), and high (IOP ≥28.0 mmHg) groups based on average IOP in the microbead injected eye. Axon numbers, axon density, axonal and glial areas, axon loss, and axon size distributions of naïve, bead, and contralateral ONs were assessed using QuPath program for automated image analysis. RESULTS (+)-PTZ treatment of BN rats protected ONs from damage caused by moderate IOP elevation. Treatment with (+)-PTZ significantly reduced axon loss and glial areas, and increased axon density and axonal areas compared to ONs from VEH treated rats with moderate IOP. (+)-PTZ-mediated neuroprotection was independent of IOP lowering effects. At average IOP ≥28.0 mmHg, (+)-PTZ treatment did not provide measurable neuroprotection. ONs from contralateral eyes exhibited subtle, complex changes in response to conditions in the bead eyes. CONCLUSIONS S1R agonist (+)-PTZ shows promise as a neuroprotective treatment for glaucoma. Future studies to understand the complex molecular mechanisms by which (+)-PTZ provides this neuroprotection are needed.
Collapse
Affiliation(s)
- Barbara A Mysona
- Department of Cellular Biology and Anatomy CB-2304, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, USA; James and Jean Culver Vision Discovery Institute, Department of Ophthalmology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| | - Jing Zhao
- James and Jean Culver Vision Discovery Institute, Department of Ophthalmology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, USA; Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| | - Oceane De Greef
- Student Training and Research Program, Graduate School, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| | - August Beisel
- Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, USA; Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| | - Parth A Patel
- Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| | - Lindsay Berman
- Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy CB-2304, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| | - Kathryn Bollinger
- James and Jean Culver Vision Discovery Institute, Department of Ophthalmology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, USA; Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
54
|
Alan E, Kerry Z, Sevin G. Molecular mechanisms of Alzheimer's disease: From therapeutic targets to promising drugs. Fundam Clin Pharmacol 2022; 37:397-427. [PMID: 36576325 DOI: 10.1111/fcp.12861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 12/06/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by cognitive impairment so widespread that it interferes with a person's ability to complete daily activities. AD is becoming increasingly common, and it is estimated that the number of patients will reach 152 million by 2050. Current treatment options for AD are symptomatic and have modest benefits. Therefore, considering the human, social, and economic burden of the disease, the development of drugs with the potential to alter disease progression has become a global priority. In this review, the molecular mechanisms involved in the pathology of AD were evaluated as therapeutic targets. The main aim of the review is to focus on new knowledge about mitochondrial dysfunction, oxidative stress, and neuronal transmission in AD, as well as a range of cellular signaling mechanisms and associated treatments. Important molecular interactions leading to AD were described in amyloid cascade and in tau protein function, oxidative stress, mitochondrial dysfunction, cholinergic and glutamatergic neurotransmission, cAMP-regulatory element-binding protein (CREB), the silent mating type information regulation 2 homolog 1 (SIRT-1), neuroinflammation (glial cells), and synaptic alterations. This review summarizes recent experimental and clinical research in AD pathology and analyzes the potential of therapeutic applications based on molecular disease mechanisms.
Collapse
Affiliation(s)
- Elif Alan
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Zeliha Kerry
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Gulnur Sevin
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| |
Collapse
|
55
|
Ahamad S, Bhat SA. The Emerging Landscape of Small-Molecule Therapeutics for the Treatment of Huntington's Disease. J Med Chem 2022; 65:15993-16032. [PMID: 36490325 DOI: 10.1021/acs.jmedchem.2c00799] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene (HTT). The new insights into HD's cellular and molecular pathways have led to the identification of numerous potent small-molecule therapeutics for HD therapy. The field of HD-targeting small-molecule therapeutics is accelerating, and the approval of these therapeutics to combat HD may be expected in the near future. For instance, preclinical candidates such as naphthyridine-azaquinolone, AN1, AN2, CHDI-00484077, PRE084, EVP4593, and LOC14 have shown promise for further optimization to enter into HD clinical trials. This perspective aims to summarize the advent of small-molecule therapeutics at various stages of clinical development for HD therapy, emphasizing their structure and design, therapeutic effects, and specific mechanisms of action. Further, we have highlighted the key drivers involved in HD pathogenesis to provide insights into the basic principle for designing promising anti-HD therapeutic leads.
Collapse
Affiliation(s)
- Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh, Uttar Pradesh202002, India
| | - Shahnawaz A Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh202002, India
| |
Collapse
|
56
|
Bezprozvanny I. Alzheimer's disease - Where do we go from here? Biochem Biophys Res Commun 2022; 633:72-76. [PMID: 36344168 DOI: 10.1016/j.bbrc.2022.08.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA; Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg State Polytechnical University, St. Petersburg, 195251, Russia.
| |
Collapse
|
57
|
Pooladgar P, Sakhabakhsh M, Taghva A, Soleiman-Meigooni S. Donepezil Beyond Alzheimer's Disease? A Narrative Review of Therapeutic Potentials of Donepezil in Different Diseases. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e128408. [PMID: 36942075 PMCID: PMC10024338 DOI: 10.5812/ijpr-128408] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022]
Abstract
Donepezil hydrochloride is an acetylcholine esterase inhibitor studied and approved to treat Alzheimer's disease (AD). However, this drug can have positive therapeutic potential in treating different conditions, including various neurodegenerative disorders such as other types of dementia, multiple sclerosis, Parkinson's disease, psychiatric and mood disorders, and even infectious diseases. Hence, this study reviewed the therapeutic potential of this drug in treating Alzheimer's and other diseases by reviewing the articles from databases including Web of Science, Scopus, PubMed, Cochrane, and Science Direct. It was shown that donepezil could affect the pathophysiology of these diseases via mechanisms such as increasing the concentration of acetylcholine, modulating local and systemic inflammatory processes, affecting acetylcholine receptors like nicotinic and muscarinic receptors, and activating various cellular signaling via receptors like sigma-1 receptors. Despite many therapeutic potentials, this drug has not yet been approved for treating non-Alzheimer's diseases, and more comprehensive studies are needed.
Collapse
Affiliation(s)
- Parham Pooladgar
- Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Sakhabakhsh
- Head of Department of Neurology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Arsia Taghva
- Department of Psychiatry, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
58
|
Ahmed H, Zheng MQ, Smart K, Fang H, Zhang L, Emery PR, Gao H, Ropchan J, Haider A, Tamagnan G, Carson RE, Ametamey SM, Huang Y. Evaluation of ( rac)-, ( R)-, and ( S)- 18F-OF-NB1 for Imaging GluN2B Subunit-Containing N-Methyl-d-Aspartate Receptors in Nonhuman Primates. J Nucl Med 2022; 63:1912-1918. [PMID: 35710735 PMCID: PMC9730915 DOI: 10.2967/jnumed.122.263977] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/07/2022] [Indexed: 01/11/2023] Open
Abstract
Despite 2 decades of research, no N-methyl-d-aspartate (NMDA) glutamate receptor (GluN) subtype 2B (GluN1/2B) radioligand is yet clinically validated. Previously, we reported on (rac)-18F-OF-NB1 as a promising GluN1/2B PET probe in rodents and its successful application for the visualization of GluN2B-containing NMDA receptors in postmortem brain tissues of patients with amyotrophic lateral sclerosis. In the current work, we report on the in vivo characterization of (rac)-, (R)-, and (S)-18F-OF-NB1 in nonhuman primates. Methods: PET scans were performed on rhesus monkeys. Plasma profiling was used to obtain the arterial input function. Regional brain time-activity curves were generated and fitted with the 1- and 2-tissue-compartment models and the multilinear analysis 1 method, and the corresponding regional volumes of distribution were calculated. Blocking studies with the GluN1/2B ligand Co 101244 (0.25 mg/kg) were performed for the enantiopure radiotracers. Receptor occupancy, nonspecific volume of distribution, and regional binding potential (BP ND) were obtained. Potential off-target binding toward σ1 receptors was assessed for (S)-18F-OF-NB1 using the σ1 receptor ligand FTC-146. Results: Free plasma fraction was moderate, ranging from 12% to 16%. All radiotracers showed high and heterogeneous brain uptake, with the highest levels in the cortex. (R)-18F-OF-NB1 showed the highest uptake and slowest washout kinetics of all tracers. The 1-tissue-compartment model and multilinear analysis 1 method fitted the regional time-activity curves well for all tracers and produced reliable regional volumes of distribution, which were higher for (R)- than (S)-18F-OF-NB1. Receptor occupancy by Co 101244 was 85% and 96% for (S)-18F-OF-NB1 and (R)-18F-OF-NB1, respectively. Pretreatment with FTC-146 at both a low (0.027 mg/kg) and high (0.125 mg/kg) dose led to a similar reduction (48% and 49%, respectively) in specific binding of (S)-18F-OF-NB1. Further, pretreatment with both Co 101244 and FTC-146 did not result in a further reduction in specific binding compared with Co 101244 alone in the same monkey (82% vs. 81%, respectively). Regional BP ND values ranged from 1.3 in the semiovale to 3.4 in the cingulate cortex for (S)-18F-OF-NB1. Conclusion: Both (R)- and (S)-18F-OF-NB1 exhibited high binding specificity to GluN2B subunit-containing NMDA receptors. The fast washout kinetics, good regional BP ND values, and high plasma free fraction render (S)-18F-OF-NB1 an attractive radiotracer for clinical translation.
Collapse
Affiliation(s)
- Hazem Ahmed
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
- PET Center, Yale University, New Haven, Connecticut; and
| | | | - Kelly Smart
- PET Center, Yale University, New Haven, Connecticut; and
| | - Hanyi Fang
- PET Center, Yale University, New Haven, Connecticut; and
- Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zhang
- PET Center, Yale University, New Haven, Connecticut; and
| | - Paul R Emery
- PET Center, Yale University, New Haven, Connecticut; and
| | - Hong Gao
- PET Center, Yale University, New Haven, Connecticut; and
| | - Jim Ropchan
- PET Center, Yale University, New Haven, Connecticut; and
| | - Achi Haider
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | | | | | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland;
| | - Yiyun Huang
- PET Center, Yale University, New Haven, Connecticut; and
| |
Collapse
|
59
|
From dopamine 4 to sigma 1: Synthesis, SAR and biological characterization of a piperidine scaffold of σ1 modulators. Eur J Med Chem 2022; 244:114840. [DOI: 10.1016/j.ejmech.2022.114840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/07/2022] [Accepted: 10/08/2022] [Indexed: 11/20/2022]
|
60
|
Ramesh M, Govindaraju T. Multipronged diagnostic and therapeutic strategies for Alzheimer's disease. Chem Sci 2022; 13:13657-13689. [PMID: 36544728 PMCID: PMC9710308 DOI: 10.1039/d2sc03932j] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/13/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and a major contributor to dementia cases worldwide. AD is clinically characterized by learning, memory, and cognitive deficits. The accumulation of extracellular amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs) of tau are the pathological hallmarks of AD and are explored as targets for clinical diagnosis and therapy. AD pathology is poorly understood and there are no fully approved diagnosis and treatments. Notwithstanding the gap, decades of research in understanding disease mechanisms have revealed the multifactorial nature of AD. As a result, multipronged and holistic approaches are pertinent to targeting multiple biomarkers and targets for developing effective diagnosis and therapeutics. In this perspective, recent developments in Aβ and tau targeted diagnostic and therapeutic tools are discussed. Novel indirect, combination, and circulating biomarkers as potential diagnostic targets are highlighted. We underline the importance of multiplexing and multimodal detection of multiple biomarkers to generate biomarker fingerprints as a reliable diagnostic strategy. The classical therapeutics targeting Aβ and tau aggregation pathways are described with bottlenecks in the strategy. Drug discovery efforts targeting multifaceted toxicity involving protein aggregation, metal toxicity, oxidative stress, mitochondrial damage, and neuroinflammation are highlighted. Recent efforts focused on multipronged strategies to rationally design multifunctional modulators targeting multiple pathological factors are presented as future drug development strategies to discover potential therapeutics for AD.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| |
Collapse
|
61
|
Colwell MJ, Tagomori H, Chapman S, Gillespie AL, Cowen PJ, Harmer CJ, Murphy SE. Pharmacological targeting of cognitive impairment in depression: recent developments and challenges in human clinical research. Transl Psychiatry 2022; 12:484. [PMID: 36396622 PMCID: PMC9671959 DOI: 10.1038/s41398-022-02249-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
Impaired cognition is often overlooked in the clinical management of depression, despite its association with poor psychosocial functioning and reduced clinical engagement. There is an outstanding need for new treatments to address this unmet clinical need, highlighted by our consultations with individuals with lived experience of depression. Here we consider the evidence to support different pharmacological approaches for the treatment of impaired cognition in individuals with depression, including treatments that influence primary neurotransmission directly as well as novel targets such as neurosteroid modulation. We also consider potential methodological challenges in establishing a strong evidence base in this area, including the need to disentangle direct effects of treatment on cognition from more generalised symptomatic improvement and the identification of sensitive, reliable and objective measures of cognition.
Collapse
Affiliation(s)
- Michael J Colwell
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Hosana Tagomori
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Sarah Chapman
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Amy L Gillespie
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Philip J Cowen
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Catherine J Harmer
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Susannah E Murphy
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK.
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK.
| |
Collapse
|
62
|
Sun H, Wu M, Wang M, Zhang X, Zhu J. The regulatory role of endoplasmic reticulum chaperone proteins in neurodevelopment. Front Neurosci 2022; 16:1032607. [DOI: 10.3389/fnins.2022.1032607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
The endoplasmic reticulum (ER) is the largest tubular reticular organelle spanning the cell. As the main site of protein synthesis, Ca2+ homeostasis maintenance and lipid metabolism, the ER plays a variety of essential roles in eukaryotic cells, with ER molecular chaperones participate in all these processes. In recent years, it has been reported that the abnormal expression of ER chaperones often leads to a variety of neurodevelopmental disorders (NDDs), including abnormal neuronal migration, neuronal morphogenesis, and synaptic function. Neuronal development is a complex and precisely regulated process. Currently, the mechanism by which neural development is regulated at the ER level remains under investigation. Therefore, in this work, we reviewed the recent advances in the roles of ER chaperones in neural development and developmental disorders caused by the deficiency of these molecular chaperones.
Collapse
|
63
|
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
|
64
|
Resende R, Fernandes T, Pereira AC, Marques AP, Pereira CF. Endoplasmic Reticulum-Mitochondria Contacts Modulate Reactive Oxygen Species-Mediated Signaling and Oxidative Stress in Brain Disorders: The Key Role of Sigma-1 Receptor. Antioxid Redox Signal 2022; 37:758-780. [PMID: 35369731 DOI: 10.1089/ars.2020.8231] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Mitochondria-Associated Membranes (MAMs) are highly dynamic endoplasmic reticulum (ER)-mitochondria contact sites that, due to the transfer of lipids and Ca2+ between these organelles, modulate several physiologic processes, such as ER stress response, mitochondrial bioenergetics and fission/fusion events, autophagy, and inflammation. In addition, these contacts are implicated in the modulation of the cellular redox status since several MAMs-resident proteins are involved in the generation of reactive oxygen species (ROS), which can act as both signaling mediators and deleterious molecules, depending on their intracellular levels. Recent Advances: In the past few years, structural and functional alterations of MAMs have been associated with the pathophysiology of several neurodegenerative diseases that are closely associated with the impairment of several MAMs-associated events, including perturbation of the redox state on the accumulation of high ROS levels. Critical Issues: Inter-organelle contacts must be tightly regulated to preserve cellular functioning by maintaining Ca2+ and protein homeostasis, lipid metabolism, mitochondrial dynamics and energy production, as well as ROS signaling. Simultaneously, these contacts should avoid mitochondrial Ca2+ overload, which might lead to energetic deficits and deleterious ROS accumulation, culminating in oxidative stress-induced activation of apoptotic cell death pathways, which are common features of many neurodegenerative diseases. Future Directions: Given that Sig-1R is an ER resident chaperone that is highly enriched at the MAMs and that controls ER to mitochondria Ca2+ flux, as well as oxidative and ER stress responses, its potential as a therapeutic target for neurodegenerative diseases such as Amyotrophic Lateral Sclerosis, Alzheimer, Parkinson, and Huntington diseases should be further explored. Antioxid. Redox Signal. 37, 758-780.
Collapse
Affiliation(s)
- Rosa Resende
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Tânia Fernandes
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Catarina Pereira
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Patrícia Marques
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Cláudia Fragão Pereira
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
65
|
Martin SF. Bridging Known and Unknown Unknowns: From Natural Products and Their Mimics to Unmet Needs in Neuroscience. Acc Chem Res 2022; 55:2397-2408. [PMID: 35960884 DOI: 10.1021/acs.accounts.1c00773] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Scientific excursions into the unknown are often characterized by unanticipated twists and turns that may lead in directions that never could have been predicted. Decisions made during the course of these explorations determine what we discover. This Account chronicles one such journey that began with a challenge encountered during the synthesis of a natural product and then unfolded over more than 30 years to focus on unmet needs in neuroscience. Specifically, while developing a concise approach to tetrahydroalstonine, a heteroyohimboid alkaloid having α-adrenergic activity, we faced the predicament of assembling a key intermediate. Solving this problem resulted in the serendipitous discovery of the vinylogous Mannich reaction and a productive program wherein we used this powerful construction as a key step in the syntheses of numerous alkaloids. However, we also realized that lessons learned from the synthesis of tetrahydroalstonine could be generalized to invent a new strategy for preparing diverse collections of substituted nitrogen heterocycles that could be screened against biological targets. The approach featured the combination of several reactants in a multicomponent assembly process to give a functionalized intermediate that could be elaborated by various ring-forming reactions to give heterocyclic scaffolds that could be further diversified. Screening these compound sets against a broad range of biological targets revealed some intriguing hits, but none of them led to a productive collaboration in translational research. Notwithstanding this setback, we screened curated members of our collections against proteins in the central nervous system and discovered some substituted B-norbenzomorphans that were selective for the enigmatic sigma-2 receptor (σ2R), an understudied protein that had been primarily associated with cancer. With scant knowledge of its role in neuroscience, we posited that small molecules that bind to σ2R might be neuroprotective, thus launching a new venture. In parallel investigations we prepared analogues of the initial hits, explored their effects in animal models of neurodegenerative and neurological conditions, and identified σ2R as transmembrane protein 97 (TMEM97). After first establishing the neuroprotective effects of several σ2R/TMEM97 ligands in a transgenic Caenorhabditis elegans model of neurodegeneration, we showed that one of these has procognitive effects and reduces levels of proinflammatory cytokines in a transgenic mouse model of Alzheimer's disease. We then identified a closely related σ2R/TMEM97 ligand that mitigates hippocampal-dependent memory deficits, prevents axon degeneration, and protects neurons and oligodendrocytes after traumatic brain injury. In a recent study, this compound was shown to protect retinal ganglion cells from retinal ischemia/reperfusion injury. In other collaborative investigations, we have shown that related, but structurally distinct, σ2R/TMEM97 ligands alleviate neuropathic pain, while a σ2R/TMEM97 ligand representing yet another chemotype reduces impairments associated with alcohol withdrawal. More recently, we have shown that σ2R/TMEM97 ligands enhance survival of cortical neurons in a neuronal model of Huntington's disease. Translational and mechanistic studies in these and other areas are in progress. Solving a problem we faced in natural product synthesis thus served as an unexpected gateway to discoveries that could lead to entirely new approaches to treat neurodegenerative and neurological conditions by targeting σ2R/TMEM97, a protein that has never been associated with these afflictions.
Collapse
Affiliation(s)
- Stephen F Martin
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
66
|
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
|
67
|
Dentoni G, Castro-Aldrete L, Naia L, Ankarcrona M. The Potential of Small Molecules to Modulate the Mitochondria-Endoplasmic Reticulum Interplay in Alzheimer's Disease. Front Cell Dev Biol 2022; 10:920228. [PMID: 36092728 PMCID: PMC9459385 DOI: 10.3389/fcell.2022.920228] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease affecting a growing number of elderly individuals. No disease-modifying drugs have yet been identified despite over 30 years of research on the topic, showing the need for further research on this multifactorial disease. In addition to the accumulation of amyloid β-peptide (Aβ) and hyperphosphorylated tau (p-tau), several other alterations have been associated with AD such as calcium (Ca2+) signaling, glucose-, fatty acid-, cholesterol-, and phospholipid metabolism, inflammation, and mitochondrial dysfunction. Interestingly, all these processes have been associated with the mitochondria-endoplasmic reticulum (ER) contact site (MERCS) signaling hub. We and others have hypothesized that the dysregulated MERCS function may be one of the main pathogenic pathways driving AD pathology. Due to the variety of biological processes overseen at the MERCS, we believe that they constitute unique therapeutic targets to boost the neuronal function and recover neuronal homeostasis. Thus, developing molecules with the capacity to correct and/or modulate the MERCS interplay can unleash unique therapeutic opportunities for AD. The potential pharmacological intervention using MERCS modulators in different models of AD is currently under investigation. Here, we survey small molecules with the potential to modulate MERCS structures and functions and restore neuronal homeostasis in AD. We will focus on recently reported examples and provide an overview of the current challenges and future perspectives to develop MERCS modulators in the context of translational research.
Collapse
Affiliation(s)
| | | | | | - Maria Ankarcrona
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Science and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
68
|
Fusco A, Olowofela B, Dagra A, Hatem R, Pierre K, Siyanaki MRH, Lucke–Wold B. Management of Neuropsychiatric Symptoms for Chronic Traumatic Encephalopathy. MEDPRESS PSYCHIATRY AND BEHAVIORAL SCIENCES 2022; 1:202209003. [PMID: 36745148 PMCID: PMC9893853 DOI: 10.33582/mppbs.2022.202209003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative disease associated with repeated head injury. The common presenting neuropsychiatric manifestations and diagnostic strategies for early diagnosis and subsequent treatment will be reviewed. This article discusses methods for injury prevention, risk assessment, and methods for supportive symptom management including lifestyle modifications, physical, occupational, and neurorehabilitation, and pharmaceutical management. Lastly, we propose the use of assessment tools validated for other neurodegenerative disorders in CTE to establish a baseline, track outcomes, and measure improvement in this population.
Collapse
Affiliation(s)
- Anna Fusco
- University of Florida College of Medicine, Gainesville, FL, USA
| | | | - Abeer Dagra
- University of Florida College of Medicine, Gainesville, FL, USA
| | - Rami Hatem
- University of Florida College of Medicine, Gainesville, FL, USA
| | - Kevin Pierre
- University of Florida Department of Radiology, Gainesville, FL, USA
| | | | | |
Collapse
|
69
|
Abstract
N,N-dimethyltryptamine (DMT) is a potent psychedelic naturally produced by many plants and animals, including humans. Whether or not DMT is significant to mammalian physiology, especially within the central nervous system, is a debate that started in the early 1960s and continues to this day. This review integrates historical and recent literature to clarify this issue, giving special attention to the most controversial subjects of DMT's biosynthesis, its storage in synaptic vesicles and the activation receptors like sigma-1. Less discussed topics, like DMT's metabolic regulation or the biased activation of serotonin receptors, are highlighted. We conclude that most of the arguments dismissing endogenous DMT's relevance are based on obsolete data or misleading assumptions. Data strongly suggest that DMT can be relevant as a neurotransmitter, neuromodulator, hormone and immunomodulator, as well as being important to pregnancy and development. Key experiments are addressed to definitely prove what specific roles DMT plays in mammalian physiology.
Collapse
Affiliation(s)
- Javier Hidalgo Jiménez
- ICEERS Foundation (International Center for Ethnobotanical Education, Research and Services), Barcelona, Spain
| | - José Carlos Bouso
- ICEERS Foundation (International Center for Ethnobotanical Education, Research and Services), Barcelona, Spain
| |
Collapse
|
70
|
Oumata N, Lu K, Teng Y, Cavé C, Peng Y, Galons H, Roques BP. Molecular mechanisms in Alzheimer's disease and related potential treatments such as structural target convergence of antibodies and simple organic molecules. Eur J Med Chem 2022; 240:114578. [PMID: 35841881 DOI: 10.1016/j.ejmech.2022.114578] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 12/12/2022]
Abstract
The amyloid cascade is the most frequently accepted hypothesis of Alzheimer's Disease (AD). According to this hypothesis, the formation of plaques precedes the appearance of fibrillary tangles. Therapeutic agents able to inhibit the formation of plaques are therefore considered as potential disease-modifying treatments (DMT) that could prevent or limit the progression of AD. Plaques are deposits formed by aggregates of amyloid-β (Aβ)-peptides. These peptides are metabolites of amyloid precursor protein (APP) first mediated by two enzymes: β-secretase 1 (BACE1) and γ-secretase. Molecular identification of these two enzymes has stimulated the development of their inhibitors. The clinical testing of these two classes of molecules has not been successful to date. The oligomerization of Aβ-peptides into plaques is now targeted by immunological approaches such as antibodies and vaccines. Structural consideration of the Aβ-peptide sequence led to the launch of the antibody Aducanumab. Several other antibodies are in late clinical phases. Progress in the understanding of the effects of N-truncated Aβ-peptides such as pE3-42, formed by the action of recently well characterized enzymes (aminopeptidase A, dipeptidylpeptidase-4 and glutaminyl cyclase) suggests that oligomerization can be limited either by enzyme inhibitors or antibody approaches. This strategy associating two structurally interconnected mechanisms is focused in this review.
Collapse
Affiliation(s)
- Nassima Oumata
- Unité de Technologies Chimiques et Biologiques pour la Santé, Université Paris Cité INSERM U1267, CNRS UMR 8258, 4 Avenue de l'Observatoire, Paris, 75006, France
| | - Kui Lu
- Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yuou Teng
- Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Christian Cavé
- UMR CNRS 8076 BioCIS, Faculty of Pharmacy, University Paris-Saclay, France
| | - Yu Peng
- Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Hervé Galons
- Unité de Technologies Chimiques et Biologiques pour la Santé, Université Paris Cité INSERM U1267, CNRS UMR 8258, 4 Avenue de l'Observatoire, Paris, 75006, France; Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Bernard P Roques
- Unité de Technologies Chimiques et Biologiques pour la Santé, Université Paris Cité INSERM U1267, CNRS UMR 8258, 4 Avenue de l'Observatoire, Paris, 75006, France.
| |
Collapse
|
71
|
Xu Z, Lei Y, Qin H, Zhang S, Li P, Yao K. Sigma-1 Receptor in Retina: Neuroprotective Effects and Potential Mechanisms. Int J Mol Sci 2022; 23:ijms23147572. [PMID: 35886921 PMCID: PMC9321618 DOI: 10.3390/ijms23147572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Retinal degenerative diseases are the major factors leading to severe visual impairment and even irreversible blindness worldwide. The therapeutic approach for retinal degenerative diseases is one extremely urgent and hot spot in science research. The sigma-1 receptor is a novel, multifunctional ligand-mediated molecular chaperone residing in endoplasmic reticulum (ER) membranes and the ER-associated mitochondrial membrane (ER-MAM); it is widely distributed in numerous organs and tissues of various species, providing protective effects on a variety of degenerative diseases. Over three decades, considerable research has manifested the neuroprotective function of sigma-1 receptor in the retina and has attempted to explore the molecular mechanism of action. In the present review, we will discuss neuroprotective effects of the sigma-1 receptor in retinal degenerative diseases, mainly in aspects of the following: the localization in different types of retinal neurons, the interactions of sigma-1 receptors with other molecules, the correlated signaling pathways, the influence of sigma-1 receptors to cellular functions, and the potential therapeutic effects on retinal degenerative diseases.
Collapse
|
72
|
Levoin N, Murthy AVR, Narendar V, Kumar NS, Aparna P, Bhavani AKD, Reddy CR, Mosset P, Grée R. Discovery of potent dual ligands for dopamine D4 and σ1 receptors. Bioorg Med Chem 2022; 69:116851. [PMID: 35753263 DOI: 10.1016/j.bmc.2022.116851] [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: 02/25/2022] [Revised: 05/04/2022] [Accepted: 05/20/2022] [Indexed: 11/27/2022]
Abstract
During our work on exploration of molecules with some piperidine-triazole scaffolds, we realized that our compounds display chemical similarity with some σ, as well as dopaminergic receptor ligands. Here we show that this series of molecules has indeed strong affinity both for σ1 and dopamine D4 receptors. Moreover, they appear selective towards σ2, dopamine paralogues D1, D2, D3 and D5 receptors and hERG channel. Extensive molecular dynamics with our lead compound AVRM-13 were carried out on σ1, supporting agonist activity of the ligand. Unexpectedly, several observations suggested the existence of a cation binding domain, a probable regulatory site for calcium.
Collapse
Affiliation(s)
- Nicolas Levoin
- Bioprojet-Biotech, 4 rue du Chesnay Beauregard, BP 96205, 35762 Saint Grégoire, France.
| | - Appala Venkata Ramana Murthy
- Chemveda Life Sciences India Pvt. Ltd., #B-11/1, IDA Uppal, Hyderabad 500039, Telangana, India; Jawaharlal Nehru Technological University Hyderabad, Kukatpally, Hyderabad 500 085, Telangana, India
| | - Vennu Narendar
- Chemveda Life Sciences India Pvt. Ltd., #B-11/1, IDA Uppal, Hyderabad 500039, Telangana, India
| | | | - Pasula Aparna
- Jawaharlal Nehru Technological University Hyderabad, Kukatpally, Hyderabad 500 085, Telangana, India
| | | | - Chada Raji Reddy
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Paul Mosset
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000 Rennes, France
| | - René Grée
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000 Rennes, France.
| |
Collapse
|
73
|
Ren P, Wang J, Li N, Li G, Ma H, Zhao Y, Li Y. Sigma-1 Receptors in Depression: Mechanism and Therapeutic Development. Front Pharmacol 2022; 13:925879. [PMID: 35784746 PMCID: PMC9243434 DOI: 10.3389/fphar.2022.925879] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/26/2022] [Indexed: 12/26/2022] Open
Abstract
Depression is the most common type of neuropsychiatric illness and has increasingly become a major cause of disability. Unfortunately, the recent global pandemic of COVID-19 has dramatically increased the incidence of depression and has significantly increased the burden of mental health care worldwide. Since full remission of the clinical symptoms of depression has not been achieved with current treatments, there is a constant need to discover new compounds that meet the major clinical needs. Recently, the roles of sigma receptors, especially the sigma-1 receptor subtype, have attracted increasing attention as potential new targets and target-specific drugs due to their translocation property that produces a broad spectrum of biological functions. Even clinical first-line antidepressants with or without affinity for sigma-1 receptors have different pharmacological profiles. Thus, the regulatory role of sigma-1 receptors might be useful in treating these central nervous system (CNS) diseases. In addition, long-term mental stress disrupts the homeostasis in the CNS. In this review, we discuss the topical literature concerning sigma-1 receptor antidepressant mechanism of action in the regulation of intracellular proteostasis, calcium homeostasis and especially the dynamic Excitatory/Inhibitory (E/I) balance in the brain. Furthermore, based on these discoveries, we discuss sigma-1 receptor ligands with respect to their promise as targets for fast-onset action drugs in treating depression.
Collapse
Affiliation(s)
- Peng Ren
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jingya Wang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Nanxi Li
- Department of Pharmaceutical Sciences, Beijng Institute of Radiation Medicine, Beijing, China
| | - Guangxiang Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hui Ma
- Beijing Institute of Basic Medical Sciences, Beijing, China
- *Correspondence: Hui Ma, ; Yongqi Zhao, ; Yunfeng Li,
| | - Yongqi Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, China
- *Correspondence: Hui Ma, ; Yongqi Zhao, ; Yunfeng Li,
| | - Yunfeng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- *Correspondence: Hui Ma, ; Yongqi Zhao, ; Yunfeng Li,
| |
Collapse
|
74
|
Milenina LS, Krutetskaya ZI, Antonov VG, Krutetskaya NI. Sigma-1 Receptor Ligands Chlorpromazine and Trifluoperazine Attenuate Ca 2+ Responses in Rat Peritoneal Macrophages. CELL AND TISSUE BIOLOGY 2022; 16:233-244. [PMID: 35668825 PMCID: PMC9136207 DOI: 10.1134/s1990519x22030075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/23/2022]
Abstract
Sigma-1 receptors are ubiquitous multifunctional ligand-regulated molecular chaperones in the endoplasmic reticulum membrane with a unique history, structure, and pharmacological profile. Sigma-1 receptors bind ligands of different chemical structure and pharmacological action and modulate a wide range of cellular processes in health and disease, including Ca2+ signaling. To elucidate the involvement of sigma-1 receptors in the processes of Ca2+ signaling in macrophages we studied the effect of sigma-1 receptor ligands, phenothiazine neuroleptics chlorpromazine and trifluoperazine, on Ca2+ responses induced by inhibitors of endoplasmic Ca2+-ATPases thapsigargin and cyclopiazonic acid, as well as by disulfide-containing immunomodulators Glutoxim and Molixan in rat peritoneal macrophages. Using Fura-2AM microfluorimetry we showed for the first time that chlorpromazine and trifluoperazine inhibit both phases of Ca2+ responses induced by Glutoxim, Molixan, thapsigargin, and cyclopiazonic acid in rat peritoneal macrophages. The data obtained indicate the participation of sigma-1 receptors in a complex signaling cascade caused by Glutoxim or Molixan and leading to an increase in intracellular Ca2+ concentration in macrophages. The results also indicate the involvement of sigma-1 receptors in the regulation of store-dependent Ca2+entry in macrophages.
Collapse
Affiliation(s)
- L. S. Milenina
- Department of Biophysics, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Z. I. Krutetskaya
- Department of Biophysics, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - V. G. Antonov
- Department of Clinical Biochemistry and Laboratory Diagnostics, Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - N. I. Krutetskaya
- Department of Biophysics, St. Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
75
|
Couly S, Goguadze N, Yasui Y, Kimura Y, Wang SM, Sharikadze N, Wu HE, Su TP. Knocking Out Sigma-1 Receptors Reveals Diverse Health Problems. Cell Mol Neurobiol 2022; 42:597-620. [PMID: 33095392 PMCID: PMC8062587 DOI: 10.1007/s10571-020-00983-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
Sigma-1 receptor (Sig-1R) is a protein present in several organs such as brain, lung, and heart. In a cell, Sig-1R is mainly located across the membranes of the endoplasmic reticulum and more specifically at the mitochondria-associated membranes. Despite numerous studies showing that Sig-1R could be targeted to rescue several cellular mechanisms in different pathological conditions, less is known about its fundamental relevance. In this review, we report results from various studies and focus on the importance of Sig-1R in physiological conditions by comparing Sig-1R KO mice to wild-type mice in order to investigate the fundamental functions of Sig-1R. We note that the Sig-1R deletion induces cognitive, psychiatric, and motor dysfunctions, but also alters metabolism of heart. Finally, taken together, observations from different experiments demonstrate that those dysfunctions are correlated to poor regulation of ER and mitochondria metabolism altered by stress, which could occur with aging.
Collapse
Affiliation(s)
- Simon Couly
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA.
| | - Nino Goguadze
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Yuriko Kimura
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Shao-Ming Wang
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Nino Sharikadze
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Hsiang-En Wu
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| |
Collapse
|
76
|
Todd TW, Petrucelli L. Modelling amyotrophic lateral sclerosis in rodents. Nat Rev Neurosci 2022; 23:231-251. [PMID: 35260846 DOI: 10.1038/s41583-022-00564-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2022] [Indexed: 12/11/2022]
Abstract
The efficient study of human disease requires the proper tools, one of the most crucial of which is an accurate animal model that faithfully recapitulates the human condition. The study of amyotrophic lateral sclerosis (ALS) is no exception. Although the majority of ALS cases are considered sporadic, most animal models of this disease rely on genetic mutations identified in familial cases. Over the past decade, the number of genes associated with ALS has risen dramatically and, with each new genetic variant, there is a drive to develop associated animal models. Rodent models are of particular importance as they allow for the study of ALS in the context of a living mammal with a comparable CNS. Such models not only help to verify the pathogenicity of novel mutations but also provide critical insight into disease mechanisms and are crucial for the testing of new therapeutics. In this Review, we aim to summarize the full spectrum of ALS rodent models developed to date.
Collapse
Affiliation(s)
- Tiffany W Todd
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA.
| |
Collapse
|
77
|
Bogár F, Fülöp L, Penke B. Novel Therapeutic Target for Prevention of Neurodegenerative Diseases: Modulation of Neuroinflammation with Sig-1R Ligands. Biomolecules 2022; 12:363. [PMID: 35327555 PMCID: PMC8945408 DOI: 10.3390/biom12030363] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases (NDDs) are characterized by progressive deterioration of the structure and function of cells and their networks in the nervous system. There are currently no drugs or other treatments that can stop the progression of NDDs. NDDs have many similarities and common pathways, e.g., formation of misfolded amyloid proteins, intra- and extracellular amyloid deposits, and chronic inflammation. Initially, the inflammation process has a cytoprotective function; however, an elevated and prolonged immune response has damaging effects and causes cell death. Neuroinflammation has been a target of drug development for treating and curing NDDs. Treatment of different NDDs with non-steroid anti-inflammatory drugs (NSAIDs) has failed or has given inconsistent results. The use of NSAIDs in diagnosed Alzheimer's disease is currently not recommended. Sigma-1 receptor (Sig-1R) is a novel target for NDD drug development. Sig-1R plays a key role in cellular stress signaling, and it regulates endoplasmic reticulum stress and unfolded protein response. Activation of Sig-1R provides neuroprotection in cell cultures and animal studies. Clinical trials demonstrated that several Sig-1R agonists (pridopidine, ANAVEX3-71, fluvoxamine, dextrometorphan) and their combinations have a neuroprotective effect and slow down the progression of distinct NDDs.
Collapse
Affiliation(s)
- Ferenc Bogár
- MTA-SZTE Biomimetic Systems Research Group, Eötvös Loránd Research Network (ELKH), Dóm Square 8, H-6720 Szeged, Hungary;
- Department of Medical Chemistry, University of Szeged, Dóm Square 8, H-6720 Szeged, Hungary;
| | - Lívia Fülöp
- Department of Medical Chemistry, University of Szeged, Dóm Square 8, H-6720 Szeged, Hungary;
| | - Botond Penke
- Department of Medical Chemistry, University of Szeged, Dóm Square 8, H-6720 Szeged, Hungary;
| |
Collapse
|
78
|
Borbély E, Varga V, Szögi T, Schuster I, Bozsó Z, Penke B, Fülöp L. Impact of Two Neuronal Sigma-1 Receptor Modulators, PRE084 and DMT, on Neurogenesis and Neuroinflammation in an Aβ 1-42-Injected, Wild-Type Mouse Model of AD. Int J Mol Sci 2022; 23:2514. [PMID: 35269657 PMCID: PMC8910266 DOI: 10.3390/ijms23052514] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/14/2022] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia characterized by cognitive dysfunctions. Pharmacological interventions to slow the progression of AD are intensively studied. A potential direction targets neuronal sigma-1 receptors (S1Rs). S1R ligands are recognized as promising therapeutic agents that may alleviate symptom severity of AD, possibly via preventing amyloid-β-(Aβ-) induced neurotoxicity on the endoplasmic reticulum stress-associated pathways. Furthermore, S1Rs may also modulate adult neurogenesis, and the impairment of this process is reported to be associated with AD. We aimed to investigate the effects of two S1R agonists, dimethyltryptamine (DMT) and PRE084, in an Aβ-induced in vivo mouse model characterizing neurogenic and anti-neuroinflammatory symptoms of AD, and the modulatory effects of S1R agonists were analyzed by immunohistochemical methods and western blotting. DMT, binding moderately to S1R but with high affinity to 5-HT receptors, negatively influenced neurogenesis, possibly as a result of activating both receptors differently. In contrast, the highly selective S1R agonist PRE084 stimulated hippocampal cell proliferation and differentiation. Regarding neuroinflammation, DMT and PRE084 significantly reduced Aβ1-42-induced astrogliosis, but neither had remarkable effects on microglial activation. In summary, the highly selective S1R agonist PRE084 may be a promising therapeutic agent for AD. Further studies are required to clarify the multifaceted neurogenic and anti-neuroinflammatory roles of these agonists.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lívia Fülöp
- Department of Medical Chemistry, University of Szeged, Dóm Tér 8, H-6720 Szeged, Hungary; (E.B.); (V.V.); (T.S.); (I.S.); (Z.B.); (B.P.)
| |
Collapse
|
79
|
Sukhorukov VS, Voronkova AS, Baranich TI, Gofman AA, Brydun AV, Knyazeva LA, Glinkina VV. Molecular Mechanisms of Interactions between Mitochondria and the Endoplasmic Reticulum: A New Look at How Important Cell Functions are Supported. Mol Biol 2022. [DOI: 10.1134/s0026893322010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
80
|
Raux PL, Drutel G, Revest JM, Vallée M. New perspectives on the role of the neurosteroid pregnenolone as an endogenous regulator of type-1 cannabinoid receptor (CB1R) activity and function. J Neuroendocrinol 2022; 34:e13034. [PMID: 34486765 DOI: 10.1111/jne.13034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 12/14/2022]
Abstract
Pregnenolone is a steroid with specific characteristics, being the first steroid to be synthesised from cholesterol at all sites of steroidogenesis, including the brain. For many years, pregnenolone was defined as an inactive precursor of all steroids because no specific target had been discovered. However, over the last decade, it has become a steroid of interest because it has been recognised as being a biomarker for brain-related disorders through the development of metabolomic approaches and advanced analytical methods. In addition, physiological roles for pregnenolone emerged when specific targets were discovered. In this review, we highlight the discovery of the selective interaction of pregnenolone with the type-1 cannabinoid receptor (CB1R). After describing the specific characteristic of CB1Rs, we discuss the newly discovered mechanisms of their regulation by pregnenolone. In particular, we describe the action of pregnenolone as a negative allosteric modulator and a specific signalling inhibitor of the CB1R. These particular characteristics of pregnenolone provide a great strategic opportunity for therapeutic development in CB1-related disorders. Finally, we outline new perspectives using innovative genetic tools for the discovery of original regulatory mechanisms of pregnenolone on CB1-related functions.
Collapse
Affiliation(s)
- Pierre-Louis Raux
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Guillaume Drutel
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Jean-Michel Revest
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Monique Vallée
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", Bordeaux, France
- University of Bordeaux, Bordeaux, France
| |
Collapse
|
81
|
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
|
82
|
Estévez-Silva HM, Mediavilla T, Giacobbo BL, Liu X, Sultan FR, Marcellino DJ. Pridopidine modifies disease phenotype in a SOD1 mouse model of Amyotrophic Lateral Sclerosis. Eur J Neurosci 2022; 55:1356-1372. [PMID: 35080077 PMCID: PMC9305776 DOI: 10.1111/ejn.15608] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/30/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a lethal and incurable neurodegenerative disease due to the loss of upper and lower motor neurons, which leads to muscle weakness, atrophy, and paralysis. Sigma‐1 receptor (σ‐1R) is a ligand‐operated protein that exhibits pro‐survival and anti‐apoptotic properties. In addition, mutations in its codifying gene are linked to development of juvenile ALS pointing to an important role in ALS. Here, we investigated the disease‐modifying effects of pridopidine, a σ‐1R agonist, using a delayed onset SOD1 G93A mouse model of ALS. Mice were administered a continuous release of pridopidine (3.0 mg/kg/day) for 4 weeks starting before the appearance of any sign of muscle weakness. Mice were monitored weekly and several behavioural tests were used to evaluate muscle strength, motor coordination and gait patterns. Pridopidine‐treated SOD1 G93A mice showed genotype‐specific effects with the prevention of cachexia. In addition, these effects exhibited significant improvement of motor behaviour 5 weeks after treatment ended. However, the survival of the animals was not extended. In summary, these results show that pridopidine can modify the disease phenotype of ALS‐associated cachexia and motor deficits in a SOD1 G93A mouse model.
Collapse
Affiliation(s)
- Héctor M Estévez-Silva
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden.,Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Tomás Mediavilla
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | | - Xijia Liu
- Umeå School of Business, Economics and Statistics, Umeå University, Umeå, Sweden
| | - Fahad R Sultan
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | |
Collapse
|
83
|
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
|
84
|
Maity S, Komal P, Kumar V, Saxena A, Tungekar A, Chandrasekar V. Impact of ER Stress and ER-Mitochondrial Crosstalk in Huntington's Disease. Int J Mol Sci 2022; 23:780. [PMID: 35054963 PMCID: PMC8775980 DOI: 10.3390/ijms23020780] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 02/07/2023] Open
Abstract
Accumulation of misfolded proteins is a common phenomenon of several neurodegenerative diseases. The misfolding of proteins due to abnormal polyglutamine (PolyQ) expansions are linked to the development of PolyQ diseases including Huntington's disease (HD). Though the genetic basis of PolyQ repeats in HD remains prominent, the primary molecular basis mediated by PolyQ toxicity remains elusive. Accumulation of misfolded proteins in the ER or disruption of ER homeostasis causes ER stress and activates an evolutionarily conserved pathway called Unfolded protein response (UPR). Protein homeostasis disruption at organelle level involving UPR or ER stress response pathways are found to be linked to HD. Due to dynamic intricate connections between ER and mitochondria, proteins at ER-mitochondria contact sites (mitochondria associated ER membranes or MAMs) play a significant role in HD development. The current review aims at highlighting the most updated information about different UPR pathways and their involvement in HD disease progression. Moreover, the role of MAMs in HD progression has also been discussed. In the end, the review has focused on the therapeutic interventions responsible for ameliorating diseased states via modulating either ER stress response proteins or modulating the expression of ER-mitochondrial contact proteins.
Collapse
Affiliation(s)
- Shuvadeep Maity
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS)-Pilani (Hyderabad Campus), Shameerpet-Mandal, Hyderabad 500078, Telangana, India; (P.K.); (V.K.); (A.S.); (A.T.); (V.C.)
| | | | | | | | | | | |
Collapse
|
85
|
Sodhi DK, Hagerman R. Fragile X Premutation: Medications, Therapy and Lifestyle Advice. Pharmgenomics Pers Med 2022; 14:1689-1699. [PMID: 35002287 PMCID: PMC8721286 DOI: 10.2147/pgpm.s338846] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
The fragile X premutation is characterized by 55–200 CGG repeats in the 5ʹ untranslated region of FMR1, whereas full fragile X mutation has greater than 200 repeats and full methylation, which manifests as fragile X syndrome (FXS). The premutation spectrum of clinical involvement includes fragile X-associated tremor/ataxia syndrome (FXTAS), fragile X-associated primary ovarian insufficiency (FXPOI), and fragile X-associated neuropsychiatric disorders (FXAND). In addition, premutation carriers also suffer from various other health problems such as endocrine abnormalities and autoimmune problems. In this paper, we have discussed different health issues faced by the carriers and interventions including medications, therapy and lifestyle changes that could improve their health.
Collapse
Affiliation(s)
- Deepika Kour Sodhi
- The MIND Institute, University of California Davis Health, Sacramento, CA, USA
| | - Randi Hagerman
- The MIND Institute, University of California Davis Health, Sacramento, CA, USA.,Department of Pediatrics, University of California Davis Health, Sacramento, CA, USA
| |
Collapse
|
86
|
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: 22] [Impact Index Per Article: 7.3] [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
|
87
|
Abdullah CS, Aishwarya R, Alam S, Remex NS, Morshed M, Nitu S, Miriyala S, Panchatcharam M, Hartman B, King J, Alfrad Nobel Bhuiyan M, Traylor J, Kevil CG, Orr AW, Bhuiyan MS. The molecular role of Sigmar1 in regulating mitochondrial function through mitochondrial localization in cardiomyocytes. Mitochondrion 2022; 62:159-175. [PMID: 34902622 PMCID: PMC8790786 DOI: 10.1016/j.mito.2021.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 01/03/2023]
Abstract
Sigmar1 is a widely expressed molecular chaperone protein in mammalian cell systems. Accumulating research demonstrated the cardioprotective roles of pharmacologic Sigmar1 activation by ligands in preclinical rodent models of cardiac injury. Extensive biochemical and immuno-electron microscopic research demonstrated Sigmar1's sub-cellular localization largely depends on cell and organ types. Despite comprehensive studies, Sigmar1's direct molecular role in cardiomyocytes remains elusive. In the present study, we determined Sigmar1's subcellular localization, transmembrane topology, and function using complementary microscopy, biochemical, and functional assays in cardiomyocytes. Quantum dots in transmission electron microscopy showed Sigmar1 labeled quantum dots on the mitochondrial membranes, lysosomes, and sarcoplasmic reticulum-mitochondrial interface. Subcellular fractionation of heart cell lysates confirmed Sigmar1's localization in purified mitochondria fraction and lysosome fraction. Immunocytochemistry confirmed Sigmar1 colocalization with mitochondrial proteins in isolated adult mouse cardiomyocytes. Sigmar1's mitochondrial localization was further confirmed by Sigmar1 colocalization with Mito-Tracker in isolated mouse heart mitochondria. A series of biochemical experiments, including alkaline extraction and proteinase K treatment of purified heart mitochondria, demonstrated Sigmar1 as an integral mitochondrial membrane protein. Sigmar1's structural requirement for mitochondrial localization was determined by expressing FLAG-tagged Sigmar1 fragments in cells. Full-length Sigmar1 and Sigmar1's C terminal-deletion fragments were able to localize to the mitochondrial membrane, whereas N-terminal deletion fragment was unable to incorporate into the mitochondria. Finally, functional assays using extracellular flux analyzer and high-resolution respirometry showed Sigmar1 siRNA knockdown significantly altered mitochondrial respiration in cardiomyocytes. Overall, we found that Sigmar1 localizes to mitochondrial membranes and is indispensable for maintaining mitochondrial respiratory homeostasis in cardiomyocytes.
Collapse
Affiliation(s)
- Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Richa Aishwarya
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Shafiul Alam
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Sadia Nitu
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Sumitra Miriyala
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Manikandan Panchatcharam
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Brandon Hartman
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Judy King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | | | - James Traylor
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA.
| |
Collapse
|
88
|
Erustes AG, Guarache GC, Guedes EDC, Leão AHFF, Pereira GJDS, Smaili SS. α-Synuclein Interactions in Mitochondria-ER Contacts: A Possible Role in Parkinson's Disease. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2022; 5:25152564221119347. [PMID: 37366506 PMCID: PMC10243560 DOI: 10.1177/25152564221119347] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Endoplasmic reticulum-mitochondria contact sites regulate various biological processes, such as mitochondrial dynamics, calcium homeostasis, autophagy and lipid metabolism. Notably, dysfunctions in these contact sites are closely related to neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease and amyotrophic lateral sclerosis. However, details about the role of endoplasmic reticulum-mitochondria contact sites in neurodegenerative diseases remain unknown. In Parkinson's disease, interactions between α-synuclein in the contact sites and components of tether complexes that connect organelles can lead to various dysfunctions, especially with regards to calcium homeostasis. This review will summarize the main tether complexes present in endoplasmic reticulum-mitochondria contact sites, and their roles in calcium homeostasis and trafficking. We will discuss the impact of α-synuclein accumulation, its interaction with tethering complex components and the implications in Parkinson's disease pathology.
Collapse
Affiliation(s)
- Adolfo Garcia Erustes
- Department of Pharmacology, Escola Paulista
de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Gabriel Cicolin Guarache
- Department of Pharmacology, Escola Paulista
de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Erika da Cruz Guedes
- Department of Pharmacology, Escola Paulista
de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Soraya Soubhi Smaili
- Department of Pharmacology, Escola Paulista
de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
89
|
Martín-Guerrero SM, Markovinovic A, Mórotz GM, Salam S, Noble W, Miller CCJ. Targeting ER-Mitochondria Signaling as a Therapeutic Target for Frontotemporal Dementia and Related Amyotrophic Lateral Sclerosis. Front Cell Dev Biol 2022; 10:915931. [PMID: 35693938 PMCID: PMC9184680 DOI: 10.3389/fcell.2022.915931] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/05/2022] [Indexed: 11/18/2022] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are two major neurodegenerative diseases. FTD is the second most common cause of dementia and ALS is the most common form of motor neuron disease. These diseases are now known to be linked. There are no cures or effective treatments for FTD or ALS and so new targets for therapeutic intervention are required but this is hampered by the large number of physiological processes that are damaged in FTD/ALS. Many of these damaged functions are now known to be regulated by signaling between the endoplasmic reticulum (ER) and mitochondria. This signaling is mediated by "tethering" proteins that serve to recruit ER to mitochondria. One tether strongly associated with FTD/ALS involves an interaction between the ER protein VAPB and the mitochondrial protein PTPIP51. Recent studies have shown that ER-mitochondria signaling is damaged in FTD/ALS and that this involves breaking of the VAPB-PTPIP51 tethers. Correcting disrupted tethering may therefore correct many other downstream damaged features of FTD/ALS. Here, we review progress on this topic with particular emphasis on targeting of the VAPB-PTPIP51 tethers as a new drug target.
Collapse
Affiliation(s)
- Sandra M Martín-Guerrero
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Andrea Markovinovic
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Gábor M Mórotz
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Shaakir Salam
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Christopher C J Miller
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
90
|
Dalwadi DA, Kim S, Schetz J, Schreihofer DA, Kim S. Brain-derived neurotrophic factor for high-throughput evaluation of selective Sigma-1 receptor ligands. J Pharmacol Toxicol Methods 2022; 113:107129. [PMID: 34678430 PMCID: PMC9358981 DOI: 10.1016/j.vascn.2021.107129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/13/2021] [Accepted: 10/13/2021] [Indexed: 01/03/2023]
Abstract
The Sigma-1 receptor (S1R) is an endoplasmic reticulum (ER) chaperone protein that has been implicated in attenuating inflammatory stress-mediated brain injuries. Selective S1R agonists represent a new class of therapeutic agent for treating neuropsychiatric and neurodegenerative disorders, however, to date, no S1R ligand has been approved for therapeutic purposes. We used three potential methods on known and potential S1R ligands to develop an unambiguous high-throughput cell screen for S1R activity. We screened known and potential S1R ligands using radioligand binding and previously reported markers of S1R activity including BDNF release, modulation of IP3 mediated calcium release, and modulation of NGF-induced neurite sprouting. Here, we present results several prototypical S1R compounds and some compounds with the potential for drug repurposing. Using an in-situ ELISA approach we demonstrated that these compounds could stimulate S1R-mediated BDNF release, which is a valuable therapeutic property since BDNF plays a critical role in neuronal support. These compounds were classified as S1R agonists because the BDNF response was comparable to the prototypical agonist 4-PPBP and because it could be reversed by a S1R selective concentration of the antagonist BD1063. When modulation of IP3 mediated calcium response and NGF-induced neurite sprouting were used as a measure of S1R activation, we were unable to reproduce the published results and determined that they are not reliable measures for evaluating functional properties of S1R ligands.
Collapse
Affiliation(s)
- Dhwanil A Dalwadi
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Department of Medicine, Division of Gastroenterology, Oregon Health Sciences University, Portland, OR 97239, USA
| | - Stephanie Kim
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; University of Texas Medical Branch at Galveston, School of Medicine, Galveston, TX 77555, USA
| | - John Schetz
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Derek A Schreihofer
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Seongcheol Kim
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Department of Cellular and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA.
| |
Collapse
|
91
|
Biophysical Evaluation and In Vitro Controlled Release of Two Isomeric Adamantane Phenylalkylamines with Antiproliferative/Anticancer and Analgesic Activity. Molecules 2021; 27:molecules27010007. [PMID: 35011245 PMCID: PMC8746252 DOI: 10.3390/molecules27010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 11/26/2022] Open
Abstract
The aqueous dissolution profile of the isomeric synthetic adamantane phenylalkylamine hydrochlorides I and II was probed. These adducts have shown significant antiproliferative/anticancer activity associated with an analgesic profile against neuropathic pain. They are both devoid of toxic effects and show appreciable enzymatic human plasma stability. The structures of these two compounds have been elucidated using 2D NMR experiments, which were used to study their predominant conformations. Compound II’s scaffold appeared more flexible, as shown by the NOE spatial interactions between the alkyl bridge chain, the aromatic rings, and the adamantane nucleus. Conversely, compound I appeared very rigid, as it did not share significant NOEs between the aforementioned structural segments. MD simulations confirmed the NOE results. The aqueous dissolution profile of both molecules fits well with their minimum energy conformers’ features, which stem from the NOE data; this was nicely demonstrated, especially in the case of compound II.
Collapse
|
92
|
Ooi K, Hu L, Feng Y, Han C, Ren X, Qian X, Huang H, Chen S, Shi Q, Lin H, Wang J, Zhu D, Wang R, Xia C. Sigma-1 Receptor Activation Suppresses Microglia M1 Polarization via Regulating Endoplasmic Reticulum-Mitochondria Contact and Mitochondrial Functions in Stress-Induced Hypertension Rats. Mol Neurobiol 2021; 58:6625-6646. [PMID: 34601668 DOI: 10.1007/s12035-021-02488-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/08/2021] [Indexed: 12/31/2022]
Abstract
Exposure to stress plays a detrimental role in the pathogenesis of hypertension via neuroinflammation pathways. Microglial neuroinflammation in the rostral ventrolateral medulla (RVLM) exacerbates stress-induced hypertension (SIH) by increasing sympathetic hyperactivity. Mitochondria of microglia are the regulators of innate immune response. Sigma-1R (σ-1R) localizes to the mitochondria-associated membranes (MAMs) and regulates endoplasmic reticulum (ER) and mitochondria communication, in part through its chaperone activity. The present study aims to investigate the protective role of σ-1R on microglial-mediated neuroinflammation. Stress-induced hypertension (SIH) was induced in rats using electric foot shocks and intermittent noise. Arterial blood pressure (ABP), heart rate (HR), and renal sympathetic nerve activity (RSNA) were measured to evaluate the sympathetic nervous system (SNS) activities. SKF10047 (100 µM), an agonist of σ-1R, was administrated to rats, then σ-1R localization and MAM alterations were detected by immuno-electron microscopy. Mitochondrial calcium homeostasis was examined in primary microglia and/or BV-2 microglia cells. The effect of SKF10047 treatment on the mitochondrial respiratory function of cultured microglia was measured using a Seahorse Extracellular Flux Analyzer. Confocal microscopic images were performed to indicate mitochondrial dynamics. Stress reduces σ-1R's localization at the MAMs, leading to decreased ER-mitochondria contact and IP3R-GRP75-VDAC calcium transport complexes expression in the RVLM of rats. SKF10047 promotes the length and coverage of MAMs in the prorenin-treated microglia. Prorenin treatment increases mitoROS levels, and inhibits Ca2+ signalling between the two organelles, therefore negatively affects ATP production in BV2 cells, and these effects are reversed by SKF10047 treatment. We found mitochondrial hyperfusion and microglial M1 polarization in prorenin-treated microglia. SKF10047 suppresses microglial M1 polarization and RVLM neuroinflammation, subsequently ameliorates sympathetic hyperactivity in stress-induced hypertensive rats. Sigma-1 receptor activation suppresses microglia M1 polarization and neuroinflammation via regulating endoplasmic reticulum-mitochondria contact and mitochondrial functions in stress-induced hypertension rats.
Collapse
Affiliation(s)
- Kokwin Ooi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Li Hu
- Department of Cardiovascular Diseases, Renji Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chenzhi Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Xiaorong Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Xinyi Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Haofeng Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Sijia Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Qi Shi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Hong Lin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Jijiang Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Danian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Rui Wang
- Department of Cardiovascular Diseases, Yangpu District Central Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200090, People's Republic of China.
| | - Chunmei Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
93
|
Mouffak S, Shubbar Q, Saleh E, El-Awady R. Recent advances in management of COVID-19: A review. Biomed Pharmacother 2021; 143:112107. [PMID: 34488083 PMCID: PMC8390390 DOI: 10.1016/j.biopha.2021.112107] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/09/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused and is still causing significant mortality and economic consequences all over the globe. As of today, there are three U.S Food and Drug administration (FDA) approved vaccines, Pfizer-BioNTech, Moderna and Janssen COVID-19 vaccine. Also, the antiviral drug remdesivir and two combinations of monoclonal antibodies are authorized for Emergency use (EUA) in certain patients. Furthermore, baricitinib was approved in Japan (April 23, 2021). Despite available vaccines and EUA, pharmacological therapy for the prevention and treatment of COVID-19 is still highly required. There are several ongoing clinical trials investigating the efficacy of clinically available drugs in treating COVID-19. In this study, selected novel pharmacological agents for the possible treatment of COVID-19 will be discussed. Point of discussion will cover mechanism of action, supporting evidence for safety and efficacy and reached stage in development. Drugs were classified into three classes according to the phase of viral life cycle they target. Phase I, the early infective phase, relies on supportive care and symptomatic treatment as needed. In phase II, the pulmonary phase, treatment aims at inhibiting viral entry or replication. Drugs used during this phase are famotidine, monoclonal antibodies, nanobodies, ivermectin, remdesivir, camostat mesylate and other antiviral agents. Finally, phase III, the hyper-inflammatory phase, tocilizumab, dexamethasone, selective serotonin reuptake inhibitors (SSRI), and melatonin are used. The aim of this study is to summarize current findings and suggest gaps in knowledge that can influence future COVID-19 treatment study design.
Collapse
Affiliation(s)
- Soraya Mouffak
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Qamar Shubbar
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ekram Saleh
- Clinical Biochemistry and Molecular Biology Unit, Cancer Biology department, National Cancer Institute, Cairo University, Egypt
| | - Raafat El-Awady
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates; Sharjah Institute of Medical Research, University of Sharjah, Sharjah, United Arab Emirates.
| |
Collapse
|
94
|
Wu NH, Ye Y, Wan BB, Yu YD, Liu C, Chen QJ. Emerging Benefits: Pathophysiological Functions and Target Drugs of the Sigma-1 Receptor in Neurodegenerative Diseases. Mol Neurobiol 2021; 58:5649-5666. [PMID: 34383254 DOI: 10.1007/s12035-021-02524-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023]
Abstract
The sigma-1 receptor (Sig-1R) is encoded by the SIGMAR1 gene and is a nonopioid transmembrane receptor located in the mitochondrial-associated endoplasmic reticulum membrane (MAM). It helps to locate endoplasmic reticulum calcium channels, regulates calcium homeostasis, and acts as a molecular chaperone to control cell fate and participate in signal transduction. It plays an important role in protecting neurons through a variety of signaling pathways and participates in the regulation of cognition and motor behavior closely related to neurodegenerative diseases. Based on its neuroprotective effects, Sig-1R has now become a breakthrough target for alleviating Alzheimer's disease and other neurodegenerative diseases. This article reviews the most cutting-edge research on the function of Sig-1R under normal or pathologic conditions and target drugs of the sigma-1 receptor in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ning-Hua Wu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
- Basic Medical College, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Yu Ye
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Bin-Bin Wan
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Yuan-Dong Yu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China.
| | - Qing-Jie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China.
| |
Collapse
|
95
|
Connection Lost, MAM: Errors in ER-Mitochondria Connections in Neurodegenerative Diseases. Brain Sci 2021; 11:brainsci11111437. [PMID: 34827436 PMCID: PMC8615542 DOI: 10.3390/brainsci11111437] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 01/12/2023] Open
Abstract
Mitochondria associated membranes (MAMs), as the name suggests, are the membranes that physically and biochemically connect mitochondria with endoplasmic reticulum. MAMs not only structurally but also functionally connect these two important organelles within the cell which were previously thought to exist independently. There are multiple points of communication between ER-mitochondria and MAMs play an important role in both ER and mitochondria functions such as Ca2+ homeostasis, proteostasis, mitochondrial bioenergetics, movement, and mitophagy. The number of disease-related proteins and genes being associated with MAMs has been continually on the rise since its discovery. There is an overwhelming overlap between the biochemical functions of MAMs and processes affected in neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Thus, MAMs have received well-deserving and much delayed attention as modulators for ER-mitochondria communication and function. This review briefly discusses the recent progress made in this now fast developing field full of promise for very exciting future therapeutic discoveries.
Collapse
|
96
|
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
|
97
|
Shi M, Chen F, Chen Z, Yang W, Yue S, Zhang J, Chen X. Sigma-1 Receptor: A Potential Therapeutic Target for Traumatic Brain Injury. Front Cell Neurosci 2021; 15:685201. [PMID: 34658788 PMCID: PMC8515188 DOI: 10.3389/fncel.2021.685201] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 09/13/2021] [Indexed: 12/29/2022] Open
Abstract
The sigma-1 receptor (Sig-1R) is a chaperone receptor that primarily resides at the mitochondria-associated endoplasmic reticulum (ER) membrane (MAM) and acts as a dynamic pluripotent modulator regulating cellular pathophysiological processes. Multiple pharmacological studies have confirmed the beneficial effects of Sig-1R activation on cellular calcium homeostasis, excitotoxicity modulation, reactive oxygen species (ROS) clearance, and the structural and functional stability of the ER, mitochondria, and MAM. The Sig-1R is expressed broadly in cells of the central nervous system (CNS) and has been reported to be involved in various neurological disorders. Traumatic brain injury (TBI)-induced secondary injury involves complex and interrelated pathophysiological processes such as cellular apoptosis, glutamate excitotoxicity, inflammatory responses, endoplasmic reticulum stress, oxidative stress, and mitochondrial dysfunction. Thus, given the pluripotent modulation of the Sig-1R in diverse neurological disorders, we hypothesized that the Sig-1R may affect a series of pathophysiology after TBI. This review summarizes the current knowledge of the Sig-1R, its mechanistic role in various pathophysiological processes of multiple CNS diseases, and its potential therapeutic role in TBI.
Collapse
Affiliation(s)
- Mingming Shi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurosurgery, Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurosurgery, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Fanglian Chen
- Department of Neurosurgery, Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurosurgery, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Zhijuan Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Weidong Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuyuan Yue
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurosurgery, Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurosurgery, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurosurgery, Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurosurgery, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| |
Collapse
|
98
|
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
|
99
|
Reyes ST, Deacon RMJ, Guo SG, Altimiras FJ, Castillo JB, van der Wildt B, Morales AP, Park JH, Klamer D, Rosenberg J, Oberman LM, Rebowe N, Sprouse J, Missling CU, McCurdy CR, Cogram P, Kaufmann WE, Chin FT. Effects of the sigma-1 receptor agonist blarcamesine in a murine model of fragile X syndrome: neurobehavioral phenotypes and receptor occupancy. Sci Rep 2021; 11:17150. [PMID: 34433831 PMCID: PMC8387417 DOI: 10.1038/s41598-021-94079-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/21/2021] [Indexed: 11/08/2022] Open
Abstract
Fragile X syndrome (FXS), a disorder of synaptic development and function, is the most prevalent genetic form of intellectual disability and autism spectrum disorder. FXS mouse models display clinically-relevant phenotypes, such as increased anxiety and hyperactivity. Despite their availability, so far advances in drug development have not yielded new treatments. Therefore, testing novel drugs that can ameliorate FXS' cognitive and behavioral impairments is imperative. ANAVEX2-73 (blarcamesine) is a sigma-1 receptor (S1R) agonist with a strong safety record and preliminary efficacy evidence in patients with Alzheimer's disease and Rett syndrome, other synaptic neurodegenerative and neurodevelopmental disorders. S1R's role in calcium homeostasis and mitochondrial function, cellular functions related to synaptic function, makes blarcamesine a potential drug candidate for FXS. Administration of blarcamesine in 2-month-old FXS and wild type mice for 2 weeks led to normalization in two key neurobehavioral phenotypes: open field test (hyperactivity) and contextual fear conditioning (associative learning). Furthermore, there was improvement in marble-burying (anxiety, perseverative behavior). It also restored levels of BDNF, a converging point of many synaptic regulators, in the hippocampus. Positron emission tomography (PET) and ex vivo autoradiographic studies, using the highly selective S1R PET ligand [18F]FTC-146, demonstrated the drug's dose-dependent receptor occupancy. Subsequent analyses also showed a wide but variable brain regional distribution of S1Rs, which was preserved in FXS mice. Altogether, these neurobehavioral, biochemical, and imaging data demonstrates doses that yield measurable receptor occupancy are effective for improving the synaptic and behavioral phenotype in FXS mice. The present findings support the viability of S1R as a therapeutic target in FXS, and the clinical potential of blarcamesine in FXS and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Samantha T Reyes
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Robert M J Deacon
- FRAXA-DVI, FRAXA, Santiago, Chile
- IEB, Faculty of Science, University of Chile, Santiago, Chile
- Fraunhofer Chile Research, Center for Systems Biotechnology, Santiago, Chile
| | - Scarlett G Guo
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Francisco J Altimiras
- FRAXA-DVI, FRAXA, Santiago, Chile
- Faculty of Engineering and Business, Universidad de las Américas, Santiago, Chile
| | - Jessa B Castillo
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | | | - Aimara P Morales
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Jun Hyung Park
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Daniel Klamer
- Anavex Life Sciences Corp., New York, NY, 10019, USA
| | - Jarrett Rosenberg
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Lindsay M Oberman
- Center for Neuroscience & Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nell Rebowe
- Anavex Life Sciences Corp., New York, NY, 10019, USA
| | | | | | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Patricia Cogram
- FRAXA-DVI, FRAXA, Santiago, Chile
- IEB, Faculty of Science, University of Chile, Santiago, Chile
- Fraunhofer Chile Research, Center for Systems Biotechnology, Santiago, Chile
| | - Walter E Kaufmann
- Anavex Life Sciences Corp., New York, NY, 10019, USA.
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Frederick T Chin
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
100
|
Martin P, Reeder T, Sourbron J, de Witte PAM, Gammaitoni AR, Galer BS. An Emerging Role for Sigma-1 Receptors in the Treatment of Developmental and Epileptic Encephalopathies. Int J Mol Sci 2021; 22:8416. [PMID: 34445144 PMCID: PMC8395113 DOI: 10.3390/ijms22168416] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022] Open
Abstract
Developmental and epileptic encephalopathies (DEEs) are complex conditions characterized primarily by seizures associated with neurodevelopmental and motor deficits. Recent evidence supports sigma-1 receptor modulation in both neuroprotection and antiseizure activity, suggesting that sigma-1 receptors may play a role in the pathogenesis of DEEs, and that targeting this receptor has the potential to positively impact both seizures and non-seizure outcomes in these disorders. Recent studies have demonstrated that the antiseizure medication fenfluramine, a serotonin-releasing drug that also acts as a positive modulator of sigma-1 receptors, reduces seizures and improves everyday executive functions (behavior, emotions, cognition) in patients with Dravet syndrome and Lennox-Gastaut syndrome. Here, we review the evidence for sigma-1 activity in reducing seizure frequency and promoting neuroprotection in the context of DEE pathophysiology and clinical presentation, using fenfluramine as a case example. Challenges and opportunities for future research include developing appropriate models for evaluating sigma-1 receptors in these syndromic epileptic conditions with multisystem involvement and complex clinical presentation.
Collapse
Affiliation(s)
- Parthena Martin
- Zogenix, Inc., Emeryville, CA 94608, USA; (P.M.); (T.R.); (A.R.G.)
| | - Thadd Reeder
- Zogenix, Inc., Emeryville, CA 94608, USA; (P.M.); (T.R.); (A.R.G.)
| | - Jo Sourbron
- University Hospital KU Leuven, 3000 Leuven, Belgium;
| | - Peter A. M. de Witte
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences at KU Leuven, 3000 Leuven, Belgium;
| | | | - Bradley S. Galer
- Zogenix, Inc., Emeryville, CA 94608, USA; (P.M.); (T.R.); (A.R.G.)
| |
Collapse
|