1
|
Wan J, van Ouwerkerk A, Mouren JC, Heredia C, Pradel L, Ballester B, Andrau JC, Spicuglia S. Comprehensive mapping of genetic variation at Epromoters reveals pleiotropic association with multiple disease traits. Nucleic Acids Res 2024:gkae1270. [PMID: 39727170 DOI: 10.1093/nar/gkae1270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/28/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
There is growing evidence that a wide range of human diseases and physiological traits are influenced by genetic variation of cis-regulatory elements. We and others have shown that a subset of promoter elements, termed Epromoters, also function as enhancer regulators of distal genes. This opens a paradigm in the study of regulatory variants, as single nucleotide polymorphisms (SNPs) within Epromoters might influence the expression of several (distal) genes at the same time, which could disentangle the identification of disease-associated genes. Here, we built a comprehensive resource of human Epromoters using newly generated and publicly available high-throughput reporter assays. We showed that Epromoters display intrinsic and epigenetic features that distinguish them from typical promoters. By integrating Genome-Wide Association Studies (GWAS), expression Quantitative Trait Loci (eQTLs) and 3D chromatin interactions, we found that regulatory variants at Epromoters are concurrently associated with more disease and physiological traits, as compared with typical promoters. To dissect the regulatory impact of Epromoter variants, we evaluated their impact on regulatory activity by analyzing allelic-specific high-throughput reporter assays and provided reliable examples of pleiotropic Epromoters. In summary, our study represents a comprehensive resource of regulatory variants supporting the pleiotropic role of Epromoters.
Collapse
Affiliation(s)
- Jing Wan
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| | - Antoinette van Ouwerkerk
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| | | | - Carla Heredia
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, UMR 5535, Montpellier, France
| | - Lydie Pradel
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| | - Benoit Ballester
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
| | - Jean-Christophe Andrau
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, UMR 5535, Montpellier, France
| | - Salvatore Spicuglia
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| |
Collapse
|
2
|
Barghash RF, Gemmati D, Awad AM, Elbakry MMM, Tisato V, Awad K, Singh AV. Navigating the COVID-19 Therapeutic Landscape: Unveiling Novel Perspectives on FDA-Approved Medications, Vaccination Targets, and Emerging Novel Strategies. Molecules 2024; 29:5564. [PMID: 39683724 PMCID: PMC11643501 DOI: 10.3390/molecules29235564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Amidst the ongoing global challenge of the SARS-CoV-2 pandemic, the quest for effective antiviral medications remains paramount. This comprehensive review delves into the dynamic landscape of FDA-approved medications repurposed for COVID-19, categorized as antiviral and non-antiviral agents. Our focus extends beyond conventional narratives, encompassing vaccination targets, repurposing efficacy, clinical studies, innovative treatment modalities, and future outlooks. Unveiling the genomic intricacies of SARS-CoV-2 variants, including the WHO-designated Omicron variant, we explore diverse antiviral categories such as fusion inhibitors, protease inhibitors, transcription inhibitors, neuraminidase inhibitors, nucleoside reverse transcriptase, and non-antiviral interventions like importin α/β1-mediated nuclear import inhibitors, neutralizing antibodies, and convalescent plasma. Notably, Molnupiravir emerges as a pivotal player, now licensed in the UK. This review offers a fresh perspective on the historical evolution of COVID-19 therapeutics, from repurposing endeavors to the latest developments in oral anti-SARS-CoV-2 treatments, ushering in a new era of hope in the battle against the pandemic.
Collapse
Affiliation(s)
- Reham F. Barghash
- Institute of Chemical Industries Research, National Research Centre, Dokki, Cairo 12622, Egypt
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Cairo 12451, Egypt
| | - Donato Gemmati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Ahmed M. Awad
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Mustafa M. M. Elbakry
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Cairo 12451, Egypt
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo 11566, Egypt
| | - Veronica Tisato
- Centre Hemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| | - Kareem Awad
- Institute of Pharmaceutical and Drug Industries Research, National Research Center, Dokki, Cairo 12622, Egypt;
| | - Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| |
Collapse
|
3
|
Gnacek P, Piergies N, Niemiec P, Kowalska O, Oćwieja M. Spectroscopic studies under properties of chlorpromazine conjugated to gold nanoparticles. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 320:124588. [PMID: 38870699 DOI: 10.1016/j.saa.2024.124588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/01/2024] [Accepted: 06/02/2024] [Indexed: 06/15/2024]
Abstract
Scientific studies have demonstrated that conjugates of anticancer drugs with metal nanoparticles (MeNPs) lead to a more effective deactivation of tumor cells compared to free drugs. Similarly, it has been established that conjugates of antibiotics with MeNPs exhibit higher biocidal activity against bacteria than their unbound counterparts. However, limited information is available regarding conjugates formed from drugs other than anticancer and antibiotics. Therefore, our research aims to develop synthesis methods for conjugates of chlorpromazine (CPZ), a neuroleptic, with gold nanoparticles (AuNPs). CPZ-AuNP conjugates were prepared through a ligand exchange reaction conducted on the surface of quasi-spherical, negatively charged citrate-stabilized TC-AuNPs with an average size of 55 ± 5 nm. UV-vis spectroscopy was employed to determine the stability range of the conjugates under controlled conditions of pH and ionic strength. Based on electrokinetic measurements, it was observed that the zeta potential of CPZ-AuNP conjugates strongly depends on the amount of CPZ adsorbed on the TC-AuNP surface. Additionally, the conjugates exhibited an isoelectric point at pH 8.8. Surface-enhanced Raman spectroscopy (SERS) and surface-enhanced infrared absorption spectroscopy (SEIRA) were employed to elucidate the adsorption structure of CPZ on TC-AuNPs. The interpretation of the spectra was conducted based on the Raman and FTIR spectra of CPZ, along with calculations performed using Density Functional Theory (DFT). The results indicated that CPZ primarily interacts with the TC-AuNP surface through the angularly oriented phenothiazine ring and the propylene bridge. Furthermore, it was demonstrated that the C-N-C fragment is perpendicular to the surface of the TC-AuNP with which it interacts. The findings from this analysis suggest the potential for further research on the use of these conjugates in biomedical applications.
Collapse
Affiliation(s)
- Patrycja Gnacek
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, PL-30239 Krakow, Poland.
| | - Natalia Piergies
- Institute of Nuclear Physics Polish Academy of Sciences, PL-31342 Krakow, Poland.
| | - Piotr Niemiec
- Faculty of Mathematics and Natural Sciences, Department of Chemistry, University of Applied Sciences in Tarnow, Mickiewicza 8, 33-100 Tarnow, Poland.
| | - Oliwia Kowalska
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, PL-30239 Krakow, Poland.
| | - Magdalena Oćwieja
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, PL-30239 Krakow, Poland.
| |
Collapse
|
4
|
Nazir F, John Kombe Kombe A, Khalid Z, Bibi S, Zhang H, Wu S, Jin T. SARS-CoV-2 replication and drug discovery. Mol Cell Probes 2024; 77:101973. [PMID: 39025272 DOI: 10.1016/j.mcp.2024.101973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
The coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has killed millions of people and continues to wreak havoc across the globe. This sudden and deadly pandemic emphasizes the necessity for anti-viral drug development that can be rapidly administered to reduce morbidity, mortality, and virus propagation. Thus, lacking efficient anti-COVID-19 treatment, and especially given the lengthy drug development process as well as the critical death tool that has been associated with SARS-CoV-2 since its outbreak, drug repurposing (or repositioning) constitutes so far, the ideal and ready-to-go best approach in mitigating viral spread, containing the infection, and reducing the COVID-19-associated death rate. Indeed, based on the molecular similarity approach of SARS-CoV-2 with previous coronaviruses (CoVs), repurposed drugs have been reported to hamper SARS-CoV-2 replication. Therefore, understanding the inhibition mechanisms of viral replication by repurposed anti-viral drugs and chemicals known to block CoV and SARS-CoV-2 multiplication is crucial, and it opens the way for particular treatment options and COVID-19 therapeutics. In this review, we highlighted molecular basics underlying drug-repurposing strategies against SARS-CoV-2. Notably, we discussed inhibition mechanisms of viral replication, involving and including inhibition of SARS-CoV-2 proteases (3C-like protease, 3CLpro or Papain-like protease, PLpro) by protease inhibitors such as Carmofur, Ebselen, and GRL017, polymerases (RNA-dependent RNA-polymerase, RdRp) by drugs like Suramin, Remdesivir, or Favipiravir, and proteins/peptides inhibiting virus-cell fusion and host cell replication pathways, such as Disulfiram, GC376, and Molnupiravir. When applicable, comparisons with SARS-CoV inhibitors approved for clinical use were made to provide further insights to understand molecular basics in inhibiting SARS-CoV-2 replication and draw conclusions for future drug discovery research.
Collapse
Affiliation(s)
- Farah Nazir
- Center of Disease Immunity and Investigation, College of Medicine, Lishui University, Lishui, 323000, China
| | - Arnaud John Kombe Kombe
- Laboratory of Structural Immunology, Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zunera Khalid
- Laboratory of Structural Immunology, Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Shaheen Bibi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Anhui, China
| | - Hongliang Zhang
- Center of Disease Immunity and Investigation, College of Medicine, Lishui University, Lishui, 323000, China
| | - Songquan Wu
- Center of Disease Immunity and Investigation, College of Medicine, Lishui University, Lishui, 323000, China.
| | - Tengchuan Jin
- Center of Disease Immunity and Investigation, College of Medicine, Lishui University, Lishui, 323000, China; Laboratory of Structural Immunology, Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Anhui, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei, 230027, China; Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
5
|
Oćwieja M, Barbasz A, Kowalska O, Maciejewska-Prończuk J, Lada A. The Adsorption of Chlorpromazine on the Surface of Gold Nanoparticles and Its Effect on the Toxicity to Selected Mammalian Cells. MATERIALS (BASEL, SWITZERLAND) 2024; 17:4774. [PMID: 39410344 PMCID: PMC11477946 DOI: 10.3390/ma17194774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024]
Abstract
Chlorpromazine (CPZ) is a first-generation neuroleptic with well-established antitumor and antiviral properties. Currently, numerous studies are focused on developing new methods for CPZ delivery; however, the knowledge regarding its conjugates with metal nanoparticles remains limited. The aim of this study was to prepare CPZ conjugates with gold nanoparticles (AuNPs) and evaluate their biological activity on human lymphocytes (HUT-78 and COLO 720L), as well as human (COLO 679) and murine (B16-F0) melanoma cells, in comparison to the effects induced by unconjugated CPZ molecules and AuNPs with well-defined properties. During the treatment of cells with CPZ, AuNPs, and CPZ-AuNP conjugates, changes in mitochondrial activity, membrane integrity, and the secretion of lipid peroxidation mediators were studied using standard biological assays such as MTT, LDH, and MDA assays. It was found that positively charged CPZ-AuNP conjugates more effectively reduced cell viability compared to AuNPs alone. The dose-dependent membrane damage was correlated with oxidative stress resulting from exposure to CPZ-AuNP conjugates. The activity of the conjugates depended on their composition and the size of the AuNPs. It was concluded that conjugating CPZ to AuNPs reduced its biological activity, while the cellular response to the treatment varied depending on the specific cell type.
Collapse
Affiliation(s)
- Magdalena Oćwieja
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, PL-30239 Krakow, Poland;
| | - Anna Barbasz
- Department of Biochemistry and Biophysics, Institute of Biology and Earth Sciences, University of the National Education Commission, Podchorazych 2, PL-30084 Krakow, Poland;
| | - Oliwia Kowalska
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, PL-30239 Krakow, Poland;
| | - Julia Maciejewska-Prończuk
- Department of Chemical and Process Engineering, Cracow University of Technology, Warszawska 24, PL-31155 Krakow, Poland;
| | - Agata Lada
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, University of Applied Sciences in Tarnow, Mickiewicza 8, PL-33100 Tarnow, Poland;
| |
Collapse
|
6
|
Máthé D, Szalay G, Cseri L, Kis Z, Pályi B, Földes G, Kovács N, Fülöp A, Szepesi Á, Hajdrik P, Csomos A, Zsembery Á, Kádár K, Katona G, Mucsi Z, Rózsa BJ, Kovács E. Monitoring correlates of SARS-CoV-2 infection in cell culture using a two-photon-active calcium-sensitive dye. Cell Mol Biol Lett 2024; 29:105. [PMID: 39030477 PMCID: PMC11264913 DOI: 10.1186/s11658-024-00619-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/26/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND The organism-wide effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral infection are well studied, but little is known about the dynamics of how the infection spreads in time among or within cells due to the scarcity of suitable high-resolution experimental systems. It has been reported that SARS-CoV-2 infection pathways converge at calcium influx and subcellular calcium distribution changes. Imaging combined with a proper staining technique is an effective tool for studying subcellular calcium-related infection and replication mechanisms at such resolutions. METHODS Using two-photon (2P) fluorescence imaging with our novel Ca-selective dye, automated image analysis and clustering analysis were applied to reveal titer and variant effects on SARS-CoV-2-infected Vero E6 cells. RESULTS The application of a new calcium sensor molecule is shown, combined with a high-end 2P technique for imaging and identifying the patterns associated with cellular infection damage within cells. Vero E6 cells infected with SARS-CoV-2 variants, D614G or B.1.1.7, exhibit elevated cytosolic calcium levels, allowing infection monitoring by tracking the cellular changes in calcium level by the internalized calcium sensor. The imaging provides valuable information on how the level and intracellular distribution of calcium are perturbed during the infection. Moreover, two-photon calcium sensing allowed the distinction of infections by two studied viral variants via cluster analysis of the image parameters. This approach will facilitate the study of cellular correlates of infection and their quantification depending on viral variants and viral load. CONCLUSIONS We propose a new two-photon microscopy-based method combined with a cell-internalized sensor to quantify the level of SARS-CoV-2 infection. We optimized the applied dye concentrations to not interfere with viral fusion and viral replication events. The presented method ensured the proper monitoring of viral infection, replication, and cell fate. It also enabled distinguishing intracellular details of cell damage, such as vacuole and apoptotic body formation. Using clustering analysis, 2P microscopy calcium fluorescence images were suitable to distinguish two different viral variants in cell cultures. Cellular harm levels read out by calcium imaging were quantitatively related to the initial viral multiplicity of infection numbers. Thus, 2P quantitative calcium imaging might be used as a correlate of infection or a correlate of activity in cellular antiviral studies.
Collapse
Affiliation(s)
- Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, Tűzoltó utca 37-47, 1094, Budapest, Hungary.
- In Vivo Imaging Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine, Tűzoltó utca 37-47, 1094, Budapest, Hungary.
- HUN-REN Physical Virology Research Group, Semmelweis University, Tűzoltó utca 37-47, 1094, Budapest, Hungary.
| | - Gergely Szalay
- Laboratory of 3D Functional Network and Dendritic Imaging, HUN-REN Institute of Experimental Medicine, Szigony utca 43, 1083, Budapest, Hungary
- BrainVisionCenter, Liliom utca 43-45, 1094, Budapest, Hungary
| | - Levente Cseri
- BrainVisionCenter, Liliom utca 43-45, 1094, Budapest, Hungary
- Femtonics Ltd., Tűzoltó utca 59, 1094, Budapest, Hungary
| | - Zoltán Kis
- National Center for Public Health, Albert Flórián út 2-6, 1097, Budapest, Hungary
| | - Bernadett Pályi
- National Center for Public Health, Albert Flórián út 2-6, 1097, Budapest, Hungary
| | - Gábor Földes
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Heart and Vascular Center, Semmelweis University, Városmajor utca. 68, 1122, Budapest, Hungary
| | - Noémi Kovács
- In Vivo Imaging Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine, Tűzoltó utca 37-47, 1094, Budapest, Hungary
| | - Anna Fülöp
- Femtonics Ltd., Tűzoltó utca 59, 1094, Budapest, Hungary
| | - Áron Szepesi
- Laboratory of 3D Functional Network and Dendritic Imaging, HUN-REN Institute of Experimental Medicine, Szigony utca 43, 1083, Budapest, Hungary
- BrainVisionCenter, Liliom utca 43-45, 1094, Budapest, Hungary
| | - Polett Hajdrik
- Department of Biophysics and Radiation Biology, Semmelweis University, Tűzoltó utca 37-47, 1094, Budapest, Hungary
| | - Attila Csomos
- Femtonics Ltd., Tűzoltó utca 59, 1094, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, 1117, Budapest, Hungary
| | - Ákos Zsembery
- Department of Oral Biology, Faculty of Dentistry, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Kristóf Kádár
- Department of Oral Biology, Faculty of Dentistry, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Gergely Katona
- Two-Photon Measurement Technology Group, The Faculty of Information Technology, Pázmány Péter Catholic University, Szigony utca 50/A, 1083, Budapest, Hungary
| | - Zoltán Mucsi
- BrainVisionCenter, Liliom utca 43-45, 1094, Budapest, Hungary.
- Femtonics Ltd., Tűzoltó utca 59, 1094, Budapest, Hungary.
- Institute of Chemistry, Faculty of Materials Science and Engineering, University of Miskolc, Egyetem tér 1, 3515, Miskolc, Hungary.
| | - Balázs József Rózsa
- Laboratory of 3D Functional Network and Dendritic Imaging, HUN-REN Institute of Experimental Medicine, Szigony utca 43, 1083, Budapest, Hungary.
- BrainVisionCenter, Liliom utca 43-45, 1094, Budapest, Hungary.
- Two-Photon Measurement Technology Group, The Faculty of Information Technology, Pázmány Péter Catholic University, Szigony utca 50/A, 1083, Budapest, Hungary.
| | - Ervin Kovács
- Two-Photon Measurement Technology Group, The Faculty of Information Technology, Pázmány Péter Catholic University, Szigony utca 50/A, 1083, Budapest, Hungary.
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok körútja 2, 1117, Budapest, Hungary.
| |
Collapse
|
7
|
Yuan Z, Ye J, Liu B, Zhang L. Unraveling the role of autophagy regulation in Crohn's disease: from genetic mechanisms to potential therapeutics. ADVANCED BIOTECHNOLOGY 2024; 2:14. [PMID: 39883213 PMCID: PMC11740883 DOI: 10.1007/s44307-024-00021-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 01/31/2025]
Abstract
Autophagy serves as the primary intracellular degradation mechanism in which damaged organelles and self-cytoplasmic proteins are transported to the lysosome for degradation. Crohn's disease, an idiopathic chronic inflammatory disorder of the gastrointestinal tract, manifests in diverse regions of the digestive system. Recent research suggests that autophagy modulation may be a new avenue for treating Crohn's disease, and several promising small-molecule modulators of autophagy have been reported as therapeutic options. In this review, we discuss in detail how mutations in autophagy-related genes function in Crohn's disease and summarize the modulatory effects on autophagy of small-molecule drugs currently used for Crohn's disease treatment. Furthermore, we delve into the therapeutic potential of small-molecule autophagy inducers on Crohn's disease, emphasizing the prospects for development in this field. We aim to highlight the significance of autophagy modulation in Crohn's disease, with the aspiration of contributing to the development of more efficacious treatments that can alleviate their suffering, and improve their quality of life.
Collapse
Affiliation(s)
- Ziyue Yuan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jing Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
8
|
Alkafaas SS, Abdallah AM, Hassan MH, Hussien AM, Elkafas SS, Loutfy SA, Mikhail A, Murad OG, Elsalahaty MI, Hessien M, Elshazli RM, Alsaeed FA, Ahmed AE, Kamal HK, Hafez W, El-Saadony MT, El-Tarabily KA, Ghosh S. Molecular docking as a tool for the discovery of novel insight about the role of acid sphingomyelinase inhibitors in SARS- CoV-2 infectivity. BMC Public Health 2024; 24:395. [PMID: 38321448 PMCID: PMC10848368 DOI: 10.1186/s12889-024-17747-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Recently, COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants, caused > 6 million deaths. Symptoms included respiratory strain and complications, leading to severe pneumonia. SARS-CoV-2 attaches to the ACE-2 receptor of the host cell membrane to enter. Targeting the SARS-CoV-2 entry may effectively inhibit infection. Acid sphingomyelinase (ASMase) is a lysosomal protein that catalyzes the conversion of sphingolipid (sphingomyelin) to ceramide. Ceramide molecules aggregate/assemble on the plasma membrane to form "platforms" that facilitate the viral intake into the cell. Impairing the ASMase activity will eventually disrupt viral entry into the cell. In this review, we identified the metabolism of sphingolipids, sphingolipids' role in cell signal transduction cascades, and viral infection mechanisms. Also, we outlined ASMase structure and underlying mechanisms inhibiting viral entry 40 with the aid of inhibitors of acid sphingomyelinase (FIASMAs). In silico molecular docking analyses of FIASMAs with inhibitors revealed that dilazep (S = - 12.58 kcal/mol), emetine (S = - 11.65 kcal/mol), pimozide (S = - 11.29 kcal/mol), carvedilol (S = - 11.28 kcal/mol), mebeverine (S = - 11.14 kcal/mol), cepharanthine (S = - 11.06 kcal/mol), hydroxyzin (S = - 10.96 kcal/mol), astemizole (S = - 10.81 kcal/mol), sertindole (S = - 10.55 kcal/mol), and bepridil (S = - 10.47 kcal/mol) have higher inhibition activity than the candidate drug amiodarone (S = - 10.43 kcal/mol), making them better options for inhibition.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Abanoub Mosaad Abdallah
- Narcotic Research Department, National Center for Social and Criminological Research (NCSCR), Giza, 11561, Egypt
| | - Mai H Hassan
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Aya Misbah Hussien
- Biotechnology department at Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt
- Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Nanotechnology Research Center, British University, Cairo, Egypt
| | - Abanoub Mikhail
- Department of Physics, Faculty of Science, Minia University, Minia, Egypt
- Faculty of Physics, ITMO University, Saint Petersburg, Russia
| | - Omnia G Murad
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mohamed I Elsalahaty
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Rami M Elshazli
- Biochemistry and Molecular Genetics Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University - Egypt, New Damietta, 34517, Egypt
| | - Fatimah A Alsaeed
- Department of Biology, College of Science, King Khalid University, Muhayl, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | - Hani K Kamal
- Anatomy and Histology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Wael Hafez
- NMC Royal Hospital, 16Th Street, 35233, Khalifa City, Abu Dhabi, United Arab Emirates
- Medical Research Division, Department of Internal Medicine, The National Research Centre, 12622, 33 El Buhouth St, Ad Doqi, Dokki, Cairo Governorate, Egypt
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, 15551, United Arab Emirates
| | - Soumya Ghosh
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, 9301, South Africa
- Natural & Medical Science Research Center, University of Nizwa, Nizwa, Oman
| |
Collapse
|
9
|
Xi Z, Jiang Y, Ma Z, Li Q, Xi X, Fan C, Zhu S, Zhang J, Xu L. Using Mesoporous Silica-Based Dual Biomimetic Nano-Erythrocytes for an Improved Antitumor Effect. Pharmaceutics 2023; 15:2785. [PMID: 38140125 PMCID: PMC10747987 DOI: 10.3390/pharmaceutics15122785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/27/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
The nano-delivery system with a dual biomimetic effect can penetrate deeper in tumor microenvironments (TMEs) and release sufficient antitumor drugs, which has attracted much attention. In this study, we synthesized erythrocyte-like mesoporous silica nanoparticles (EMSNs) as the core loaded with doxorubicin (DOX) and coated them with calcium phosphate (CaP) and erythrocyte membrane (EM) to obtain DOX/EsPMs. The transmission electron microscopy (TEM), fluorescent co-localization and protein bands of SDS-PAGE were used to confirm the complete fabrication of EsPMs. The EsPMs with erythrocyte-like shape exhibited superior penetration ability in in vitro diffusion and tumor-sphere penetration experiments. Intracellular Ca2+ and ROS detection experiments showed that the CaP membranes of EsPMs with pH-sensitivity could provide Ca2+ continuously to induce reactive oxide species' (ROS) generation in the TME. The EM as a perfect "camouflaged clothing" which could confuse macrophagocytes into prolonging blood circulation. Hemolysis and non-specific protein adsorption tests proved the desirable biocompatibility of EsPMs. An in vivo pharmacodynamics evaluation showed that the DOX/EsPMs group had a satisfactory tumor-inhibition effect. These advantages of the nano-erythrocytes suggest that by modifying the existing materials to construct a nano-delivery system, nanoparticles will achieve a biomimetic effect from both their structure and function with a facilitated and sufficient drug release profile, which is of great significance for antitumor therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lu Xu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China; (Z.X.); (Y.J.); (Z.M.); (Q.L.); (X.X.); (C.F.); (S.Z.); (J.Z.)
| |
Collapse
|
10
|
Song A, Phandthong R, Talbot P. Endocytosis inhibitors block SARS-CoV-2 pseudoparticle infection of mink lung epithelium. Front Microbiol 2023; 14:1258975. [PMID: 38033586 PMCID: PMC10682793 DOI: 10.3389/fmicb.2023.1258975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Both spill over and spill back of SARS-CoV-2 virus have been reported on mink farms in Europe and the United States. Zoonosis is a public health concern as dangerous mutated forms of the virus could be introduced into the human population through spillback. Methods The purpose of our study was to determine the SARS-CoV-2 entry mechanism using the mink lung epithelial cell line (Mv1Lu) and to block entry with drug inhibitors. Results Mv1Lu cells were susceptible to SARS-CoV-2 viral pseudoparticle infection, validating them as a suitable disease model for COVID-19. Inhibitors of TMPRSS2 and of endocytosis, two pathways of viral entry, were tested to identify those that blocked infection. TMPRSS2 inhibitors had minimal impact, which can be explained by the apparent lack of activity of this enzyme in the mink and its localization within the cell, not on the cell surface. Discussion Dyngo4a, a small molecule endocytosis inhibitor, significantly reduced infection, supporting the conclusion that the entry of the SARS-CoV-2 virus into Mv1Lu cells occurs primarily through endocytosis. The small molecule inhibitors that were effective in this study could potentially be used therapeutically to prevent SARS-CoV-2 infection in mink populations. This study will facilitate the development of therapeutics to prevent zoonotic transmission of SARS-CoV-2 variants to other animals, including humans.
Collapse
Affiliation(s)
- Ann Song
- Cell, Molecular, and Developmental Biology Graduate Program, University of California, Riverside, Riverside, CA, United States
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Rattapol Phandthong
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Prue Talbot
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
11
|
Awad AM, Hansen K, Del Rio D, Flores D, Barghash RF, Kakkola L, Julkunen I, Awad K. Insights into COVID-19: Perspectives on Drug Remedies and Host Cell Responses. Biomolecules 2023; 13:1452. [PMID: 37892134 PMCID: PMC10604481 DOI: 10.3390/biom13101452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
In light of the COVID-19 global pandemic caused by SARS-CoV-2, ongoing research has centered on minimizing viral spread either by stopping viral entry or inhibiting viral replication. Repurposing antiviral drugs, typically nucleoside analogs, has proven successful at inhibiting virus replication. This review summarizes current information regarding coronavirus classification and characterization and presents the broad clinical consequences of SARS-CoV-2 activation of the angiotensin-converting enzyme 2 (ACE2) receptor expressed in different human cell types. It provides publicly available knowledge on the chemical nature of proposed therapeutics and their target biomolecules to assist in the identification of potentially new drugs for the treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ahmed M. Awad
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Kamryn Hansen
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Diana Del Rio
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Derek Flores
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Reham F. Barghash
- Institute of Chemical Industries Research, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Laura Kakkola
- Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
- Clinical Microbiology, Turku University Hospital, 20521 Turku, Finland
| | - Kareem Awad
- Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
- Department of Therapeutic Chemistry, Institute of Pharmaceutical and Drug Industries Research, National Research Center, Dokki, Cairo 12622, Egypt
| |
Collapse
|
12
|
Niort K, Dancourt J, Boedec E, Al Amir Dache Z, Lavieu G, Tareste D. Cholesterol and Ceramide Facilitate Membrane Fusion Mediated by the Fusion Peptide of the SARS-CoV-2 Spike Protein. ACS OMEGA 2023; 8:32729-32739. [PMID: 37720777 PMCID: PMC10500581 DOI: 10.1021/acsomega.3c03610] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/17/2023] [Indexed: 09/19/2023]
Abstract
SARS-CoV-2 entry into host cells is mediated by the Spike (S) protein of the viral envelope. The S protein is composed of two subunits: S1 that induces binding to the host cell via its interaction with the ACE2 receptor of the cell surface and S2 that triggers fusion between viral and cellular membranes. Fusion by S2 depends on its heptad repeat domains that bring membranes close together and its fusion peptide (FP) that interacts with and perturbs the membrane structure to trigger fusion. Recent studies have suggested that cholesterol and ceramide lipids from the cell surface may facilitate SARS-CoV-2 entry into host cells, but their exact mode of action remains unknown. We have used a combination of in vitro liposome-liposome and in situ cell-cell fusion assays to study the lipid determinants of S-mediated membrane fusion. Our findings reveal that both cholesterol and ceramide lipids facilitate fusion, suggesting that targeting these lipids could be effective against SARS-CoV-2. As a proof of concept, we examined the effect of chlorpromazine (CPZ), an antipsychotic drug known to perturb membrane structure. Our results show that CPZ effectively inhibits S-mediated membrane fusion, thereby potentially impeding SARS-CoV-2 entry into the host cell.
Collapse
Affiliation(s)
- Kristina Niort
- Université
Paris Cité, Inserm UMR-S 1266, Institute of Psychiatry and
Neuroscience of Paris (IPNP), Paris 75014, France
| | - Julia Dancourt
- Université
Paris Cité, Inserm U 1316, CNRS UMR 7057, Laboratoire Matières
et Systèmes Complexes (MSC), Paris 75006, France
| | - Erwan Boedec
- Université
Paris Cité, Inserm UMR-S 1266, Institute of Psychiatry and
Neuroscience of Paris (IPNP), Paris 75014, France
| | - Zahra Al Amir Dache
- Université
Paris Cité, Inserm U 1316, CNRS UMR 7057, Laboratoire Matières
et Systèmes Complexes (MSC), Paris 75006, France
| | - Grégory Lavieu
- Université
Paris Cité, Inserm U 1316, CNRS UMR 7057, Laboratoire Matières
et Systèmes Complexes (MSC), Paris 75006, France
| | - David Tareste
- Université
Paris Cité, Inserm UMR-S 1266, Institute of Psychiatry and
Neuroscience of Paris (IPNP), Paris 75014, France
| |
Collapse
|
13
|
Mir IH, Thirunavukkarasu C. The relevance of acid sphingomyelinase as a potential target for therapeutic intervention in hepatic disorders: current scenario and anticipated trends. Arch Toxicol 2023; 97:2069-2087. [PMID: 37248308 PMCID: PMC10226719 DOI: 10.1007/s00204-023-03529-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023]
Abstract
Acid sphingomyelinase (ASMase) serves as one of the most remarkable enzymes in sphingolipid biology. ASMase facilitates the hydrolysis of sphingomyelin, yielding ceramide and phosphorylcholine via the phospholipase C signal transduction pathway. Owing to its prominent intervention in apoptosis, ASMase, and its product ceramide is now at the bleeding edge of lipid research due to the coalesced efforts of several research institutions over the past 40 years. ASMase-catalyzed ceramide synthesis profoundly alters the physiological properties of membrane structure in response to a broad range of stimulations, orchestrating signaling cascades for endoplasmic reticulum stress, autophagy, and lysosomal membrane permeabilization, which influences the development of hepatic disorders, such as steatohepatitis, hepatic fibrosis, drug-induced liver injury, and hepatocellular carcinoma. As a result, the potential to modulate the ASMase action with appropriate pharmaceutical antagonists has sparked a lot of curiosity. This article emphasizes the fundamental mechanisms of the systems that govern ASMase aberrations in various hepatic pathologies. Furthermore, we present an insight into the potential therapeutic agents used to mitigate ASMase irregularities and the paramountcy of such inhibitors in drug repurposing.
Collapse
Affiliation(s)
- Ishfaq Hassan Mir
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, 605 014, India
| | | |
Collapse
|
14
|
Peng Y, Chen SY, Wang ZN, Zhou ZQ, Sun J, Zhang GA, Li J, Wang L, Zhao JC, Tang XX, Wang DY, Zhong NS. Dicoumarol is an effective post-exposure prophylactic for SARS-CoV-2 Omicron infection in human airway epithelium. Signal Transduct Target Ther 2023; 8:242. [PMID: 37301869 PMCID: PMC10256976 DOI: 10.1038/s41392-023-01511-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Repurposing existing drugs to inhibit SARS-CoV-2 infection in airway epithelial cells (AECs) is a quick way to find novel treatments for COVID-19. Computational screening has found dicoumarol (DCM), a natural anticoagulant, to be a potential SARS-CoV-2 inhibitor, but its inhibitory effects and possible working mechanisms remain unknown. Using air-liquid interface culture of primary human AECs, we demonstrated that DCM has potent antiviral activity against the infection of multiple Omicron variants (including BA.1, BQ.1 and XBB.1). Time-of-addition and drug withdrawal assays revealed that early treatment (continuously incubated after viral absorption) of DCM could markedly inhibit Omicron replication in AECs, but DCM did not affect the absorption, exocytosis and spread of viruses or directly eliminate viruses. Mechanistically, we performed single-cell sequencing analysis (a database of 77,969 cells from different airway locations from 10 healthy volunteers) and immunofluorescence staining, and showed that the expression of NAD(P)H quinone oxidoreductase 1 (NQO1), one of the known DCM targets, was predominantly localised in ciliated AECs. We further found that the NQO1 expression level was positively correlated with both the disease severity of COVID-19 patients and virus copy levels in cultured AECs. In addition, DCM treatment downregulated NQO1 expression and disrupted signalling pathways associated with SARS-CoV-2 disease outcomes (e.g., Endocytosis and COVID-19 signalling pathways) in cultured AECs. Collectively, we demonstrated that DCM is an effective post-exposure prophylactic for SARS-CoV-2 infection in the human AECs, and these findings could help physicians formulate novel treatment strategies for COVID-19.
Collapse
Affiliation(s)
- Yang Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Otolaryngology, Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shi-Ying Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhao-Ni Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zi-Qing Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gui-An Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jia Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Laboratory, Guangzhou, China
| | - Lei Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Laboratory, Guangzhou, China
| | - Jin-Cun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiao Xiao Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou Laboratory, Guangzhou, China.
| | - De-Yun Wang
- Department of Otolaryngology, Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou Laboratory, Guangzhou, China.
| |
Collapse
|
15
|
Hessien M, Donia T, Tabll AA, Adly E, Abdelhafez TH, Attia A, Alkafaas SS, Kuna L, Glasnovic M, Cosic V, Smolic R, Smolic M. Mechanistic-Based Classification of Endocytosis-Related Inhibitors: Does It Aid in Assigning Drugs against SARS-CoV-2? Viruses 2023; 15:v15051040. [PMID: 37243127 DOI: 10.3390/v15051040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) canonically utilizes clathrin-mediated endocytosis (CME) and several other endocytic mechanisms to invade airway epithelial cells. Endocytic inhibitors, particularly those targeting CME-related proteins, have been identified as promising antiviral drugs. Currently, these inhibitors are ambiguously classified as chemical, pharmaceutical, or natural inhibitors. However, their varying mechanisms may suggest a more realistic classification system. Herein, we present a new mechanistic-based classification of endocytosis inhibitors, in which they are segregated among four distinct classes including: (i) inhibitors that disrupt endocytosis-related protein-protein interactions, and assembly or dissociation of complexes; (ii) inhibitors of large dynamin GTPase and/or kinase/phosphatase activities associated with endocytosis; (iii) inhibitors that modulate the structure of subcellular components, especially the plasma membrane, and actin; and (iv) inhibitors that cause physiological or metabolic alterations in the endocytosis niche. Excluding antiviral drugs designed to halt SARS-CoV-2 replication, other drugs, either FDA-approved or suggested through basic research, could be systematically assigned to one of these classes. We observed that many anti-SARS-CoV-2 drugs could be included either in class III or IV as they interfere with the structural or physiological integrity of subcellular components, respectively. This perspective may contribute to our understanding of the relative efficacy of endocytosis-related inhibitors and support the optimization of their individual or combined antiviral potential against SARS-CoV-2. However, their selectivity, combined effects, and possible interactions with non-endocytic cellular targets need more clarification.
Collapse
Affiliation(s)
- Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Thoria Donia
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Ashraf A Tabll
- National Research Centre, Microbial Biotechnology Department, Biotechnology Research Institute, Giza 12622, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo 11517, Egypt
| | - Eiman Adly
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Tawfeek H Abdelhafez
- National Research Centre, Microbial Biotechnology Department, Biotechnology Research Institute, Giza 12622, Egypt
| | - Amany Attia
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Lucija Kuna
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Marija Glasnovic
- Department of Medicine, Family Medicine and History of Medicine, Faculty of Medicine Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Vesna Cosic
- Department of Paediatrics and Gynaecology with Obstetrics, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Robert Smolic
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Martina Smolic
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
- Department of Pharmacology, Faculty of Medicine Osijek, University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
16
|
Batool R, Soler M, Colavita F, Fabeni L, Matusali G, Lechuga LM. Biomimetic nanoplasmonic sensor for rapid evaluation of neutralizing SARS-CoV-2 monoclonal antibodies as antiviral therapy. Biosens Bioelectron 2023; 226:115137. [PMID: 36796306 PMCID: PMC9904857 DOI: 10.1016/j.bios.2023.115137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/16/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
Monoclonal antibody (mAb) therapy is one of the most promising immunotherapies that have shown the potential to prevent or neutralize the effects of COVID-19 in patients at very early stages, with a few formulations recently approved by the European and American medicine agencies. However, a main bottleneck for their general implementation resides in the time-consuming, laborious, and highly-specialized techniques employed for the manufacturing and assessing of these therapies, excessively increasing their prices and delaying their administration to the patients. We propose a biomimetic nanoplasmonic biosensor as a novel analytical technique for the screening and evaluation of COVID-19 mAb therapies in a simpler, faster, and reliable manner. By creating an artificial cell membrane on the plasmonic sensor surface, our label-free sensing approach enables real-time monitoring of virus-cell interactions as well as direct analysis of antibody blocking effects in only 15 min assay time. We have achieved detection limits in the 102 TCID50/mL range for the study of SARS-CoV-2 viruses, which allows to perform neutralization assays by only employing a low-volume sample with common viral loads. We have demonstrated the accuracy of the biosensor for the evaluation of two different neutralizing antibodies targeting both Delta and Omicron variants of SARS-CoV-2, with half maximal inhibitory concentrations (IC50) determined in the ng/mL range. Our user-friendly and reliable technology could be employed in biomedical and pharmaceutical laboratories to accelerate, cheapen, and simplify the development of effective immunotherapies for COVID-19 and other serious infectious diseases or cancer.
Collapse
Affiliation(s)
- Razia Batool
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, BIST, CIBER-BBN, Spain
| | - Maria Soler
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, BIST, CIBER-BBN, Spain.
| | - Francesca Colavita
- National Institute for Infectious Disease "L. Spallanzani", IRCCS, Rome, Italy
| | - Lavinia Fabeni
- National Institute for Infectious Disease "L. Spallanzani", IRCCS, Rome, Italy
| | - Giulia Matusali
- National Institute for Infectious Disease "L. Spallanzani", IRCCS, Rome, Italy
| | - Laura M Lechuga
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, BIST, CIBER-BBN, Spain.
| |
Collapse
|
17
|
Kandeel M. An overview of the recent progress in Middle East Respiratory Syndrome Coronavirus (MERS-CoV) drug discovery. Expert Opin Drug Discov 2023; 18:385-400. [PMID: 36971501 DOI: 10.1080/17460441.2023.2192921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
INTRODUCTION The Middle East respiratory syndrome coronavirus (MERS-CoV) has remained a public health concern since it first emerged in 2012. Although many potential treatments for MERS-CoV have been developed and tested, none have had complete success in stopping the spread of this deadly disease. MERS-CoV replication comprises attachment, entry, fusion and replication steps. Targeting these events may lead to the creation of medications that effectively treat MERS-CoV infection. AREAS COVERED This review updates the research on the development of inhibitors of MERS-CoV. The main topics are MERS-CoV‒related proteins and host cell proteins that are involved in viral protein activation and infection. EXPERT OPINION Research on discovering drugs that can inhibit MERS-CoV started at a slow pace, and although efforts have steadily increased, clinical trials for new drugs specifically targeting MERS-CoV have not been extensive enough. The explosion in efforts to find new medications for the SARS-CoV-2 virus indirectly enhanced the volume of data on MERS-CoV inhibition by including MERS-CoV in drug assays. The appearance of COVID-19 completely transformed the data available on MERS-CoV inhibition. Despite the fact that new infected cases are constantly being diagnosed, there are currently no approved vaccines for or inhibitors of MERS-CoV.
Collapse
|
18
|
Roy SS, Sharma S, Rizvi ZA, Sinha D, Gupta D, Rophina M, Sehgal P, Sadhu S, Tripathy MR, Samal S, Maiti S, Scaria V, Sivasubbu S, Awasthi A, Harshan KH, Jain S, Chowdhury S. G4-binding drugs, chlorpromazine and prochlorperazine, repurposed against COVID-19 infection in hamsters. Front Mol Biosci 2023; 10:1133123. [PMID: 37006620 PMCID: PMC10061221 DOI: 10.3389/fmolb.2023.1133123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has caused millions of infections and deaths worldwide. Limited treatment options and the threat from emerging variants underline the need for novel and widely accessible therapeutics. G-quadruplexes (G4s) are nucleic acid secondary structures known to affect many cellular processes including viral replication and transcription. We identified heretofore not reported G4s with remarkably low mutation frequency across >5 million SARS-CoV-2 genomes. The G4 structure was targeted using FDA-approved drugs that can bind G4s - Chlorpromazine (CPZ) and Prochlorperazine (PCZ). We found significant inhibition in lung pathology and lung viral load of SARS-CoV-2 challenged hamsters when treated with CPZ or PCZ that was comparable to the widely used antiviral drug Remdesivir. In support, in vitro G4 binding, inhibition of reverse transcription from RNA isolated from COVID-infected humans, and attenuated viral replication and infectivity in Vero cell cultures were clear in case of both CPZ and PCZ. Apart from the wide accessibility of CPZ/PCZ, targeting relatively invariant nucleic acid structures poses an attractive strategy against viruses like SARS-CoV-2, which spread fast and accumulate mutations quickly.
Collapse
Affiliation(s)
- Shuvra Shekhar Roy
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shalu Sharma
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Zaigham Abbas Rizvi
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Dipanjali Sinha
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Divya Gupta
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | - Mercy Rophina
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Paras Sehgal
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Srikanth Sadhu
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Manas Ranjan Tripathy
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Sweety Samal
- Translational Health Science and Technology Institute, Faridabad, 411008, India
| | - Souvik Maiti
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
- CSIR-National Chemical Laboratory, Pune, 121001, India
| | - Vinod Scaria
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Amit Awasthi
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Krishnan H. Harshan
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | - Sanjeev Jain
- Molecular Genetics Laboratory, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Shantanu Chowdhury
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
- *Correspondence: Shantanu Chowdhury,
| |
Collapse
|
19
|
Gaseous nitric oxide failed to inhibit the replication cycle of SARS-CoV-2 in vitro. Nitric Oxide 2023; 132:27-33. [PMID: 36706864 PMCID: PMC9873364 DOI: 10.1016/j.niox.2023.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
Nitric oxide (NO) has been shown to have antimicrobial activity in vitro and in some in vivo models, while the virucidal activity of NO remains elusive. Some studies using NO donors have suggested that NO could be a potential candidate to treat SARS-CoV infection. The Covid-19 pandemic raised the hypothesis that NO gas might have an impact on Sars-CoV-2 replication cycle and might be considered as a candidate therapy to treat COVID-19. To our knowledge, there are no in vitro preclinical studies demonstrating a virucidal effect of gaseous NO on SARS-CoV-2. This study aims to determine whether gaseous NO has an impact on the replication cycle of SARS-CoV-2 in vitro. To that end, SARS-CoV-2 infected epithelial (VeroE6) and pulmonary (A549-hACE2) cells were treated with repeated doses of gaseous NO at different concentrations known to be efficient against bacteria. Our results show that exposing SARS-CoV-2 infected-cells to NO gas even at high doses (160 ppm, 6 h) does not influence the replication cycle of the virus in vitro. We report here that NO gas has no antiviral properties in vitro on SARS-COV-2. Therefore, there is no rationale for its usage in clinical settings to treat COVID-19 patients for direct antiviral purposes, which does not exclude other potential physiological benefits of this gas.
Collapse
|
20
|
Desloratadine, an FDA-approved cationic amphiphilic drug, inhibits SARS-CoV-2 infection in cell culture and primary human nasal epithelial cells by blocking viral entry. Sci Rep 2022; 12:21053. [PMID: 36473907 PMCID: PMC9726831 DOI: 10.1038/s41598-022-25399-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
The 2019 global coronavirus (COVID-19) pandemic has brought the world to a grinding halt, highlighting the urgent need for therapeutic and preventive solutions to slow the spread of emerging viruses. The objective of this study was to assess the anti-SARS-CoV-2 effectiveness of 8 FDA-approved cationic amphiphilic drugs (CADs). SARS-CoV-2-infected Vero cells, Calu-3 cells and primary Human Nasal Epithelial Cells (HNEC) were used to investigate the effects of CADs and revealed their antiviral mode of action. Among the CADs tested, desloratadine, a commonly used antiallergic, well-tolerated with no major side effects, potently reduced the production of SARS-CoV-2 RNA in Vero-E6 cells. Interestingly, desloratadine was also effective against HCoV-229E and HCoV-OC43 showing that it possessed broad-spectrum anti-coronavirus activity. Investigation of its mode of action revealed that it targeted an early step of virus lifecycle and blocked SARS-CoV-2 entry through the endosomal pathway. Finally, the ex vivo kinetic of the antiviral effect of desloratadine was evaluated on primary Human Nasal Epithelial Cells (HNEC), showing a significant delay of viral RNA production with a maximal reduction reached after 72 h of treatment. Thus, this treatment could provide a substantial contribution to prophylaxis and systemic therapy of COVID-19 or other coronaviruses infections and requires further studies.
Collapse
|
21
|
Feng Y, Grotegut S, Jovanovic P, Gandin V, Olson SH, Murad R, Beall A, Colayco S, De-Jesus P, Chanda S, English BP, Singer RH, Jackson M, Topisirovic I, Ronai ZA. Inhibition of coronavirus HCoV-OC43 by targeting the eIF4F complex. Front Pharmacol 2022; 13:1029093. [PMID: 36532738 PMCID: PMC9751428 DOI: 10.3389/fphar.2022.1029093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
The translation initiation complex 4F (eIF4F) is a rate-limiting factor in protein synthesis. Alterations in eIF4F activity are linked to several diseases, including cancer and infectious diseases. To this end, coronaviruses require eIF4F complex activity to produce proteins essential for their life cycle. Efforts to target coronaviruses by abrogating translation have been largely limited to repurposing existing eIF4F complex inhibitors. Here, we report the results of a high throughput screen to identify small molecules that disrupt eIF4F complex formation and inhibit coronavirus RNA and protein levels. Of 338,000 small molecules screened for inhibition of the eIF4F-driven, CAP-dependent translation, we identified SBI-1232 and two structurally related analogs, SBI-5844 and SBI-0498, that inhibit human coronavirus OC43 (HCoV-OC43; OC43) with minimal cell toxicity. Notably, gene expression changes after OC43 infection of Vero E6 or A549 cells were effectively reverted upon treatment with SBI-5844 or SBI-0498. Moreover, SBI-5844 or SBI-0498 treatment effectively impeded the eIF4F complex assembly, with concomitant inhibition of newly synthesized OC43 nucleocapsid protein and OC43 RNA and protein levels. Overall, we identify SBI-5844 and SBI-0498 as small molecules targeting the eIF4F complex that may limit coronavirus transcripts and proteins, thereby representing a basis for developing novel therapeutic modalities against coronaviruses.
Collapse
Affiliation(s)
- Yongmei Feng
- Cancer Center at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Stefan Grotegut
- Conrad Prebys Center for Chemical Genomics at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Predrag Jovanovic
- Lady Davis Institute, SMBD Jewish General Hospital, Gerald Bronfman Department of Oncology and Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Valentina Gandin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - Steven H. Olson
- Conrad Prebys Center for Chemical Genomics at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Rabi Murad
- Cancer Center at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Anne Beall
- Immunology and Infectious Disease Center at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Sharon Colayco
- Immunology and Infectious Disease Center at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Paul De-Jesus
- Immunology and Infectious Disease Center at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Sumit Chanda
- Immunology and Infectious Disease Center at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Brian P. English
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - Robert H. Singer
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - Michael Jackson
- Conrad Prebys Center for Chemical Genomics at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Ivan Topisirovic
- Lady Davis Institute, SMBD Jewish General Hospital, Gerald Bronfman Department of Oncology and Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Ze’ev A. Ronai
- Cancer Center at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| |
Collapse
|
22
|
Li J, Xue Y, Wang X, Smith LS, He B, Liu S, Zhu H. Tissue- and cell-expression of druggable host proteins provide insights into repurposing drugs for COVID-19. Clin Transl Sci 2022; 15:2796-2811. [PMID: 36259251 PMCID: PMC9747131 DOI: 10.1111/cts.13400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 01/26/2023] Open
Abstract
Several human host proteins play important roles in the lifecycle of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Many drugs targeting these host proteins have been investigated as potential therapeutics for coronavirus disease 2019 (COVID-19). The tissue-specific expressions of selected host proteins were summarized using proteomics data retrieved from the Human Protein Atlas, ProteomicsDB, Human Proteome Map databases, and a clinical COVID-19 study. Protein expression features in different cell lines were summarized based on recent proteomics studies. The half-maximal effective concentration or half-maximal inhibitory concentration values were collected from in vitro studies. The pharmacokinetic data were mainly from studies in healthy subjects or non-COVID-19 patients. Considerable tissue-specific expression patterns were observed for several host proteins. ACE2 expression in the lungs was significantly lower than in many other tissues (e.g., the kidneys and intestines); TMPRSS2 expression in the lungs was significantly lower than in other tissues (e.g., the prostate and intestines). The expression levels of endocytosis-associated proteins CTSL, CLTC, NPC1, and PIKfyve in the lungs were comparable to or higher than most other tissues. TMPRSS2 expression was markedly different between cell lines, which could be associated with the cell-dependent antiviral activities of several drugs. Drug delivery receptor ICAM1 and CTSB were expressed at a higher level in the lungs than in other tissues. In conclusion, the cell- and tissue-specific proteomics data could help interpret the in vitro antiviral activities of host-directed drugs in various cells and aid the transition of the in vitro findings to clinical research to develop safe and effective therapeutics for COVID-19.
Collapse
Affiliation(s)
- Jiapeng Li
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| | - Yanling Xue
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| | - Xinwen Wang
- Department of Pharmaceutical SciencesNortheast Ohio Medical University College of PharmacyRootstownOhioUSA
| | - Logan S. Smith
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| | - Bing He
- Department of Computational Medicine and BioinformaticsUniversity of MichiganAnn ArborMichiganUSA
| | - Shuhan Liu
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| | - Hao‐Jie Zhu
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| |
Collapse
|
23
|
Pires De Souza GA, Le Bideau M, Boschi C, Wurtz N, Colson P, Aherfi S, Devaux C, La Scola B. Choosing a cellular model to study SARS-CoV-2. Front Cell Infect Microbiol 2022; 12:1003608. [PMID: 36339347 PMCID: PMC9634005 DOI: 10.3389/fcimb.2022.1003608] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/03/2022] [Indexed: 08/04/2023] Open
Abstract
As new pathogens emerge, new challenges must be faced. This is no different in infectious disease research, where identifying the best tools available in laboratories to conduct an investigation can, at least initially, be particularly complicated. However, in the context of an emerging virus, such as SARS-CoV-2, which was recently detected in China and has become a global threat to healthcare systems, developing models of infection and pathogenesis is urgently required. Cell-based approaches are crucial to understanding coronavirus infection biology, growth kinetics, and tropism. Usually, laboratory cell lines are the first line in experimental models to study viral pathogenicity and perform assays aimed at screening antiviral compounds which are efficient at blocking the replication of emerging viruses, saving time and resources, reducing the use of experimental animals. However, determining the ideal cell type can be challenging, especially when several researchers have to adapt their studies to specific requirements. This review strives to guide scientists who are venturing into studying SARS-CoV-2 and help them choose the right cellular models. It revisits basic concepts of virology and presents the currently available in vitro models, their advantages and disadvantages, and the known consequences of each choice.
Collapse
Affiliation(s)
- Gabriel Augusto Pires De Souza
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Marion Le Bideau
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Céline Boschi
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Nathalie Wurtz
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Philippe Colson
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Sarah Aherfi
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Christian Devaux
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
- Department of Biological Sciences (INSB), Centre National de la Recherche Scientifique, Marseille, France
| | - Bernard La Scola
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| |
Collapse
|
24
|
Zhang Y, Chen Y, Wu B, Liu D, Fu L, Huang F. Synthesis of tetrahedron DNA nanostructures for detecting the activation of cell signal transduction via their specific binding to transcriptional factors. NANOSCALE 2022; 14:15101-15110. [PMID: 36205195 DOI: 10.1039/d2nr01954j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
A technique for detecting the activation of cell signal transduction is particularly important for disease diagnosis and therapy. Transcriptional factor (TF) activities that could indicate the status of cell signal transduction are a favorable target for cell signal detection. Tetrahedron DNA nanostructures (TDNs) which contain specific binding sequences of TFs were designed and synthesized in this research, and their effects on detecting cell signal transduction were evaluated. We found that FAM-labeled TDNs with the indicated TF binding sequences could specifically bind to activated TFs of hypoxia signaling or TGF-β signaling. Signaling pathway activities detected via TDNs could be exhibited by various methods including fluorescence imaging, flow cytometry and fluorescence spectrometer analysis. The reliability of this new technique is in line with the classical dual luciferase reporter assay system. This work develops a novel and effective tool to examine the activation of intracellular signaling pathways via nanotechnology. In addition, good stability and programmability of TDNs ensure their widespread application in various signaling pathways.
Collapse
Affiliation(s)
- Ying Zhang
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| | - Yue Chen
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| | - Bing Wu
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| | - Danqing Liu
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| | - Lengxi Fu
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| | - Fei Huang
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| |
Collapse
|
25
|
Mousavi S, Zare S, Mirzaei M, Feizi A. Novel Drug Design for Treatment of COVID-19: A Systematic Review of Preclinical Studies. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2022; 2022:2044282. [PMID: 36199815 PMCID: PMC9527439 DOI: 10.1155/2022/2044282] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/23/2022] [Accepted: 08/03/2022] [Indexed: 11/27/2022]
Abstract
Background Since the beginning of the novel coronavirus (SARS-CoV-2) disease outbreak, there has been an increasing interest in discovering potential therapeutic agents for this disease. In this regard, we conducted a systematic review through an overview of drug development (in silico, in vitro, and in vivo) for treating COVID-19. Methods A systematic search was carried out in major databases including PubMed, Web of Science, Scopus, EMBASE, and Google Scholar from December 2019 to March 2021. A combination of the following terms was used: coronavirus, COVID-19, SARS-CoV-2, drug design, drug development, In silico, In vitro, and In vivo. A narrative synthesis was performed as a qualitative method for the data synthesis of each outcome measure. Results A total of 2168 articles were identified through searching databases. Finally, 315 studies (266 in silico, 34 in vitro, and 15 in vivo) were included. In studies with in silico approach, 98 article study repurposed drug and 91 studies evaluated herbal medicine on COVID-19. Among 260 drugs repurposed by the computational method, the best results were observed with saquinavir (n = 9), ritonavir (n = 8), and lopinavir (n = 6). Main protease (n = 154) following spike glycoprotein (n = 62) and other nonstructural protein of virus (n = 45) was among the most studied targets. Doxycycline, chlorpromazine, azithromycin, heparin, bepridil, and glycyrrhizic acid showed both in silico and in vitro inhibitory effects against SARS-CoV-2. Conclusion The preclinical studies of novel drug design for COVID-19 focused on main protease and spike glycoprotein as targets for antiviral development. From evaluated structures, saquinavir, ritonavir, eucalyptus, Tinospora cordifolia, aloe, green tea, curcumin, pyrazole, and triazole derivatives in in silico studies and doxycycline, chlorpromazine, and heparin from in vitro and human monoclonal antibodies from in vivo studies showed promised results regarding efficacy. It seems that due to the nature of COVID-19 disease, finding some drugs with multitarget antiviral actions and anti-inflammatory potential is valuable and some herbal medicines have this potential.
Collapse
Affiliation(s)
- Sarah Mousavi
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shima Zare
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahmoud Mirzaei
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Awat Feizi
- Department of Epidemiology and Biostatistics, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
26
|
Chlorpromazine, a Clinically Approved Drug, Inhibits SARS-CoV-2 Nucleocapsid-Mediated Induction of IL-6 in Human Monocytes. Molecules 2022; 27:molecules27123651. [PMID: 35744777 PMCID: PMC9228867 DOI: 10.3390/molecules27123651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic, caused by the rapidly spreading SARS-CoV-2 virus, led to the unprecedented mobilization of scientists, resulting in the rapid development of vaccines and potential pharmaceuticals. Although COVID-19 symptoms are moderately severe in most people, in some cases the disease can result in pneumonia and acute respiratory failure as well as can be fatal. The severe course of COVID-19 is associated with a hyperinflammatory state called a cytokine storm. One of the key cytokines creating a proinflammatory environment is IL-6, which is secreted mainly by monocytes and macrophages. Therefore, this cytokine has become a target for some therapies that inhibit its biological action; however, these therapies are expensive, and their availability is limited in poorer countries. Thus, new cheaper drugs that can overcome the severe infections of COVID-19 are needed. Here, we show that chlorpromazine inhibits the expression and secretion of IL-6 by monocytes activated by SARS-CoV-2 virus nucleocapsid protein and affects the activity of NF-κB and MEK/ERK signaling. Our results, including others, indicate that chlorpromazine, which has been used for several decades as a neuroleptic, exerts antiviral and immunomodulatory activity, is safe and inexpensive, and might be a desirable drug to support the therapy of patients with COVID-19.
Collapse
|
27
|
Abstract
Long COVID refers to the lingering symptoms which persist or appear after the acute illness. The dominant long COVID symptoms in the two years since the pandemic began (2020-2021) have been depression, anxiety, fatigue, concentration and cognitive impairments with few reports of psychosis. Whether other symptoms will appear later on is not yet known. For example, dopamine-dependent movement disorders generally take many years before first symptoms are seen. Post-stroke depression and anxiety may explain many of the early long COVID cases. Hemorrhagic, hypoxic and inflammatory damages of the central nervous system, unresolved systematic inflammation, metabolic impairment, cerebral vascular accidents such as stroke, hypoxia from pulmonary damages and fibrotic changes are among the major causes of long COVID. Glucose metabolic and hypoxic brain issues likely predispose subjects with pre-existing diabetes, cardiovascular or lung problems to long COVID as well. Preliminary data suggest that psychotropic medications may not be a danger but could instead be beneficial in combating COVID-19 infection. The same is true for diabetes medications such as metformin. Thus, a focus on sigma-1 receptor ligands and glucose metabolism is expected to be useful for new drug development as well as the repurposing of current drugs. The reported protective effects of psychotropics and antihistamines against COVID-19, the earlier reports of reduced number of sigma-1 receptors in post-mortem schizophrenic brains, with many antidepressant and antipsychotic drugs being antihistamines with significant affinity for the sigma-1 receptor, support the role of sigma and histamine receptors in neuroinflammation and viral infections. Literature and data in all these areas are accumulating at a fast rate. We reviewed and discussed the relevant and important literature.
Collapse
|
28
|
Tang H, Liu Y, Ren R, Liu Y, He Y, Qi Z, Peng H, Zhao P. Identification of clinical candidates against West Nile Virus by activity screening
in vitro
and effect evaluation
in vivo. J Med Virol 2022; 94:4918-4925. [DOI: 10.1002/jmv.27891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Hailin Tang
- Department of Microbiology, Faculty of Naval MedicineNavy Medical UniversityShanghai200433People's Republic of China
| | - Yang Liu
- Department of Microbiology, Faculty of Naval MedicineNavy Medical UniversityShanghai200433People's Republic of China
| | - Ruiwen Ren
- Center for Disease Control and Prevention of Southern Theater CommandGuangdong Guangzhou510507People's Republic of China
| | - Yan Liu
- Department of Microbiology, Faculty of Naval MedicineNavy Medical UniversityShanghai200433People's Republic of China
| | - Yanhua He
- Department of Microbiology, Faculty of Naval MedicineNavy Medical UniversityShanghai200433People's Republic of China
| | - Zhongtian Qi
- Department of Microbiology, Faculty of Naval MedicineNavy Medical UniversityShanghai200433People's Republic of China
| | - Haoran Peng
- Department of Microbiology, Faculty of Naval MedicineNavy Medical UniversityShanghai200433People's Republic of China
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval MedicineNavy Medical UniversityShanghai200433People's Republic of China
| |
Collapse
|
29
|
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
|
30
|
Gurin L, Evangelist M, Laverty P, Hanley K, Corcoran J, Herbsman J, Im B, Frontera J, Flanagan S, Galetta S, Lewis A. Early Neurorehabilitation and Recovery from Disorders of Consciousness After Severe COVID-19. Neurocrit Care 2022; 36:357-371. [PMID: 34611810 PMCID: PMC8491764 DOI: 10.1007/s12028-021-01359-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Early neurorehabilitation improves outcomes in patients with disorders of consciousness (DoC) after brain injury, but its applicability in COVID-19 is unknown. We describe our experience implementing an early neurorehabilitation protocol for patients with COVID-19-associated DoC in the intensive care unit (ICU) and evaluate factors associated with recovery. METHODS During the initial COVID-19 surge in New York City between March 10 and May 20, 2020, faced with a disproportionately high number of ICU patients with prolonged unresponsiveness, we developed and implemented an early neurorehabilitation protocol, applying standard practices from brain injury rehabilitation care to the ICU setting. Twenty-one patients with delayed recovery of consciousness after severe COVID-19 participated in a pilot early neurorehabilitation program that included serial Coma Recovery Scale-Revised (CRS-R) assessments, multimodal treatment, and access to clinicians specializing in brain injury medicine. We retrospectively compared clinical features of patients who did and did not recover to the minimally conscious state (MCS) or better, defined as a CRS-R total score (TS) ≥ 8, before discharge. We additionally examined factors associated with best CRS-R TS, last CRS-R TS, hospital length of stay, and time on mechanical ventilation. RESULTS Patients underwent CRS-R assessments a median of six (interquartile range [IQR] 3-10) times before discharge, beginning a median of 48 days (IQR 40-55) from admission. Twelve (57%) patients recovered to MCS after a median of 8 days (IQR 2-14) off continuous sedation; they had lower body mass index (p = 0.009), lower peak serum C-reactive protein levels (p = 0.023), higher minimum arterial partial pressure of oxygen (p = 0.028), and earlier fentanyl discontinuation (p = 0.018). CRS-R scores fluctuated over time, and the best CRS-R TS was significantly higher than the last CRS-R TS (median 8 [IQR 5-23] vs. 5 [IQR 3-18], p = 0.002). Earlier fentanyl (p = 0.001) and neuromuscular blockade (p = 0.015) discontinuation correlated with a higher last CRS-R TS. CONCLUSIONS More than half of our cohort of patients with prolonged unresponsiveness following severe COVID-19 recovered to MCS or better before hospital discharge, achieving a clinical benchmark known to have relatively favorable long-term prognostic implications in DoC of other etiologies. Hypoxia, systemic inflammation, sedation, and neuromuscular blockade may impact diagnostic assessment and prognosis, and fluctuations in level of consciousness make serial assessments essential. Early neurorehabilitation of these patients in the ICU can be accomplished but is associated with unique challenges. Further research should evaluate factors associated with longer-term neurologic recovery and benefits of early rehabilitation in patients with severe COVID-19.
Collapse
Affiliation(s)
- Lindsey Gurin
- Department of Neurology, New York University Langone Medical Center, New York, NY, USA.
- Department of Rehabilitation Medicine, New York University Langone Medical Center, New York, NY, USA.
- Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA.
| | - Megan Evangelist
- Department of Rehabilitation Medicine, New York University Langone Medical Center, New York, NY, USA
| | - Patricia Laverty
- Department of Rehabilitation Medicine, New York University Langone Medical Center, New York, NY, USA
| | - Kaitlin Hanley
- Department of Rehabilitation Medicine, New York University Langone Medical Center, New York, NY, USA
| | - John Corcoran
- Department of Rehabilitation Medicine, New York University Langone Medical Center, New York, NY, USA
| | - Jodi Herbsman
- Department of Rehabilitation Medicine, New York University Langone Medical Center, New York, NY, USA
| | - Brian Im
- Department of Rehabilitation Medicine, New York University Langone Medical Center, New York, NY, USA
| | - Jennifer Frontera
- Department of Neurology, New York University Langone Medical Center, New York, NY, USA
| | - Steven Flanagan
- Department of Rehabilitation Medicine, New York University Langone Medical Center, New York, NY, USA
| | - Steven Galetta
- Department of Neurology, New York University Langone Medical Center, New York, NY, USA
| | - Ariane Lewis
- Department of Neurology, New York University Langone Medical Center, New York, NY, USA
- Department of Neurosurgery, New York University Langone Medical Center, New York, NY, USA
| |
Collapse
|
31
|
Hashimoto Y, Suzuki T, Hashimoto K. Mechanisms of action of fluvoxamine for COVID-19: a historical review. Mol Psychiatry 2022; 27:1898-1907. [PMID: 34997196 PMCID: PMC8739627 DOI: 10.1038/s41380-021-01432-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/13/2021] [Accepted: 12/23/2021] [Indexed: 12/18/2022]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) accelerates the discovery of prophylactic and therapeutic drugs for persons infected with the virus. Drug repurposing for the COVID-19 pandemic has received particular attention. Increasing clinical data suggest that antidepressant use in early-stage subjects with COVID-19 might be associated with a reduced risk of intubation or death. Among the antidepressants, fluvoxamine is the most attractive drug for mild to moderate subjects with COVID-19. In this article, we review the mechanisms of action (i.e., serotonin transporter, sigma-1 receptor, and acid sphingomyelinase) of fluvoxamine for COVID-19. Furthermore, we discuss a possible link between maternal COVID-19 infection and a risk for neuropsychiatric disorders (i.e., autism spectrum disorder and schizophrenia) in offspring.
Collapse
Affiliation(s)
- Yaeko Hashimoto
- Department of Respirology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Takuji Suzuki
- Department of Respirology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
32
|
Zhao L, Li S, Zhong W. Mechanism of Action of Small-Molecule Agents in Ongoing Clinical Trials for SARS-CoV-2: A Review. Front Pharmacol 2022; 13:840639. [PMID: 35281901 PMCID: PMC8916227 DOI: 10.3389/fphar.2022.840639] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/28/2022] [Indexed: 01/18/2023] Open
Abstract
Since the first reports from December 2019, COVID-19 caused an overwhelming global pandemic that has affected 223 countries, seriously endangering public health and creating an urgent need for effective drugs to treat SARS-CoV-2 infection. Currently, there is a lack of safe, effective, and specific therapeutic drugs for COVID-19, with mainly supportive and symptomatic treatments being administered to patients. The preferred option for responding to an outbreak of acute infectious disease is through drug repurposing, saving valuable time that would otherwise be lost in preclinical and clinical research, hastening clinical introduction, and lowering treatment costs. Alternatively, researchers seek to design and discover novel small-molecule candidate drugs targeting the key proteins in the life cycle of SARS-CoV-2 through an in-depth study of the infection mechanism, thus obtaining a number of candidate compounds with favorable antiviral effects in preclinical and clinical settings. There is an urgent need to further elucidate the efficacy and mechanism of action of potential anti-SARS-CoV-2 small-molecule drugs. Herein, we review the candidate small-molecule anti-SARS-CoV-2 drugs in ongoing clinical trials, with a major focus on their mechanisms of action in an attempt to provide useful insight for further research and development of small-molecule compounds against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lei Zhao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- Beijing Sunho Pharmaceutical Co., Ltd., Beijing, China
| | - Song Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
33
|
Mueller JK, Riederer P, Müller WE. Neuropsychiatric Drugs Against COVID-19: What is the Clinical
Evidence? PHARMACOPSYCHIATRY 2022; 55:7-15. [DOI: 10.1055/a-1717-2381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AbstractSince the beginning of the coronavirus disease (COVID)-19 pandemic, the need for
effective treatments for COVID-19 led to the idea of
“repurposing” drugs for antiviral treatment. Several
antipsychotics and antidepressants have been tested for in vitro activity
against the severe acute respiratory syndrome coronavirus 2. Chlorpromazine,
other phenothiazine antipsychotics, and the antidepressant fluoxetine were found
to be rather potent in these studies. However, whether effective plasma
concentrations can be obtained with clinically accepted doses of these drugs is
not clear. Data of COVID-19 patients are not yet available but several clinical
studies are currently underway.The specific serotonin reuptake inhibitor fluvoxamine is a potent Sigma-1
receptor agonist and reduces inflammation in animal models of cytokine-stress.
Accordingly, fluvoxamine treatment was superior to placebo in reducing impaired
respiratory function and other symptoms of inflammation in COVID-19 patients in
a placebo-controlled clinical study and another open clinical trial. The
beneficial effects of fluvoxamine on the course of COVID-19 were recently
confirmed in a large placebo-controlled double-blind trial with several hundred
patients.Inflammation represents a major risk factor for many psychiatric disorders which
explains the high susceptibilitiy of COVID-19 patients for psychiatric diseases.
Many antidepressants and antipsychotics possess anti-inflammatory properties
independent of sigma-1 activity which might be important to reduce psychiatric
symptoms of COVID-19 patients and to improve respiratory dysfunction and other
consequences of inflammation. This might explain the rather unspecific benefit
which has been reported for several cohorts of COVID-19 patients treated with
different psychotropic drugs.
Collapse
Affiliation(s)
- Juliane K. Mueller
- Department of Psychiatry, Psychosomatic Medicine, and Psychotherapy,
University Hospital Frankfurt, Frankfurt/M, Germany
| | - Peter Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy,
University Hospital Würzburg, Würzburg, Germany
- University of Southern Denmark Odense, J.B. Winslows Vey Odense,
Denmark
| | - Walter E. Müller
- Department of Pharmacology und Clinical Pharmacy, University Frankfurt,
Frankfurt/M, Germany
| |
Collapse
|
34
|
Fred SM, Kuivanen S, Ugurlu H, Casarotto PC, Levanov L, Saksela K, Vapalahti O, Castrén E. Antidepressant and Antipsychotic Drugs Reduce Viral Infection by SARS-CoV-2 and Fluoxetine Shows Antiviral Activity Against the Novel Variants in vitro. Front Pharmacol 2022; 12:755600. [PMID: 35126106 PMCID: PMC8809408 DOI: 10.3389/fphar.2021.755600] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/09/2021] [Indexed: 12/29/2022] Open
Abstract
Repurposing of currently available drugs is a valuable strategy to tackle the consequences of COVID-19. Recently, several studies have investigated the effect of psychoactive drugs on SARS-CoV-2 in cell culture models as well as in clinical practice. Our aim was to expand these studies and test some of these compounds against newly emerged variants. Several antidepressants and antipsychotic drugs with different primary mechanisms of action were tested in ACE2/TMPRSS2-expressing human embryonic kidney cells against the infection by SARS-CoV-2 spike protein-dependent pseudoviruses. Some of these compounds were also tested in human lung epithelial cell line, Calu-1, against the first wave (B.1) lineage of SARS-CoV-2 and the variants of concern, B.1.1.7, B.1.351, and B.1.617.2. Several clinically used antidepressants, including fluoxetine, citalopram, reboxetine, imipramine, as well as antipsychotic compounds chlorpromazine, flupenthixol, and pimozide inhibited the infection by pseudotyped viruses with minimal effects on cell viability. The antiviral action of several of these drugs was verified in Calu-1 cells against the B.1 lineage of SARS-CoV-2. By contrast, the anticonvulsant carbamazepine, and novel antidepressants ketamine, known as anesthetic at high doses, and its derivatives as well as MAO and phosphodiesterase inhibitors phenelzine and rolipram, respectively, showed no activity in the pseudovirus model. Furthermore, fluoxetine remained effective against pseudoviruses with common receptor binding domain mutations, N501Y, K417N, and E484K, as well as B.1.1.7 (alpha), B.1.351 (beta), and B.1.617.2 (delta) variants of SARS-CoV-2. Our study confirms previous data and extends information on the repurposing of these drugs to counteract SARS-CoV-2 infection including different variants of concern, however, extensive clinical studies must be performed to confirm our in vitro findings.
Collapse
Affiliation(s)
- Senem Merve Fred
- Neuroscience Center–HiLIFE, University of Helsinki, Helsinki, Finland
| | - Suvi Kuivanen
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Hasan Ugurlu
- Department of Virology, University of Helsinki, Helsinki, Finland
| | | | - Lev Levanov
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Kalle Saksela
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eero Castrén
- Neuroscience Center–HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
35
|
De Hert M, Mazereel V, Stroobants M, De Picker L, Van Assche K, Detraux J. COVID-19-Related Mortality Risk in People With Severe Mental Illness: A Systematic and Critical Review. Front Psychiatry 2022; 12:798554. [PMID: 35095612 PMCID: PMC8793909 DOI: 10.3389/fpsyt.2021.798554] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/03/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Increasing clinical evidence suggests that people with severe mental illness (SMI), including schizophrenia spectrum disorders, bipolar disorder (BD), and major depressive disorder (MDD), are at higher risk of dying from COVID-19. Several systematic reviews examining the association between psychiatric disorders and COVID-19-related mortality have recently been published. Although these reviews have been conducted thoroughly, certain methodological limitations may hinder the accuracy of their research findings. Methods: A systematic literature search, using the PubMed, Embase, Web of Science, and Scopus databases (from inception to July 23, 2021), was conducted for observational studies assessing the risk of death associated with COVID-19 infection in adult patients with pre-existing schizophrenia spectrum disorders, BD, or MDD. Methodological quality of the included studies was assessed using the Newcastle-Ottawa Scale (NOS). Results: Of 1,446 records screened, 13 articles investigating the rates of death in patients with pre-existing SMI were included in this systematic review. Quality assessment scores of the included studies ranged from moderate to high. Most results seem to indicate that patients with SMI, particularly patients with schizophrenia spectrum disorders, are at significantly higher risk of COVID-19-related mortality, as compared to patients without SMI. However, the extent of the variation in COVID-19-related mortality rates between studies including people with schizophrenia spectrum disorders was large because of a low level of precision of the estimated mortality outcome(s) in certain studies. Most studies on MDD and BD did not include specific information on the mood state or disease severity of patients. Due to a lack of data, it remains unknown to what extent patients with BD are at increased risk of COVID-19-related mortality. A variety of factors are likely to contribute to the increased mortality risk of COVID-19 in these patients. These include male sex, older age, somatic comorbidities (particularly cardiovascular diseases), as well as disease-specific characteristics. Conclusion: Methodological limitations hamper the accuracy of COVID-19-related mortality estimates for the main categories of SMIs. Nevertheless, evidence suggests that SMI is associated with excess COVID-19 mortality. Policy makers therefore must consider these vulnerable individuals as a high-risk group that should be given particular attention. This means that targeted interventions to maximize vaccination uptake among these patients are required to address the higher burden of COVID-19 infection in this already disadvantaged group.
Collapse
Affiliation(s)
- Marc De Hert
- Department of Neurosciences, Center for Clinical Psychiatry, University Psychiatric Center, KU Leuven, Kortenberg, Belgium
- Antwerp Health Law and Ethics Chair, University of Antwerp, Antwerp, Belgium
| | - Victor Mazereel
- Department of Neurosciences, Center for Clinical Psychiatry, University Psychiatric Center, KU Leuven, Kortenberg, Belgium
| | - Marc Stroobants
- Biomedical Library, University Psychiatric Center, KU Leuven, Kortenberg, Belgium
| | - Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium
- University Psychiatric Hospital Campus Duffel, Duffel, Belgium
| | - Kristof Van Assche
- Antwerp Health Law and Ethics Chair, University of Antwerp, Antwerp, Belgium
- Research Group Personal Rights and Property Rights, Faculty of Law, University of Antwerp, Antwerp, Belgium
| | - Johan Detraux
- Department of Neurosciences, Public Health Psychiatry, University Psychiatric Center, KU Leuven, Kortenberg, Belgium
| |
Collapse
|
36
|
Hoertel N, Sánchez-Rico M, Gulbins E, Kornhuber J, Carpinteiro A, Lenze EJ, Reiersen AM, Abellán M, de la Muela P, Vernet R, Blanco C, Cougoule C, Beeker N, Neuraz A, Gorwood P, Alvarado JM, Meneton P, Limosin F. Association Between FIASMAs and Reduced Risk of Intubation or Death in Individuals Hospitalized for Severe COVID-19: An Observational Multicenter Study. Clin Pharmacol Ther 2021. [PMID: 34050932 DOI: 10.1002/cpt.2317.10.1002/cpt.2317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Several medications commonly used for a number of medical conditions share a property of functional inhibition of acid sphingomyelinase (ASM), or FIASMA. Preclinical and clinical evidence suggest that the ASM/ceramide system may be central to severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection. We examined the potential usefulness of FIASMA use among patients hospitalized for severe coronavirus disease 2019 (COVID-19) in an observational multicenter study conducted at Greater Paris University hospitals. Of 2,846 adult patients hospitalized for severe COVID-19, 277 (9.7%) were taking an FIASMA medication at the time of their hospital admission. The primary end point was a composite of intubation and/or death. We compared this end point between patients taking vs. not taking an FIASMA medication in time-to-event analyses adjusted for sociodemographic characteristics and medical comorbidities. The primary analysis was a Cox regression model with inverse probability weighting (IPW). Over a mean follow-up of 9.2 days (SD = 12.5), the primary end point occurred in 104 patients (37.5%) receiving an FIASMA medication, and 1,060 patients (41.4%) who did not. Despite being significantly and substantially associated with older age and greater medical severity, FIASMA medication use was significantly associated with reduced likelihood of intubation or death in both crude (hazard ratio (HR) = 0.71, 95% confidence interval (CI) = 0.58-0.87, P < 0.001) and primary IPW (HR = 0.58, 95%CI = 0.46-0.72, P < 0.001) analyses. This association remained significant in multiple sensitivity analyses and was not specific to one particular FIASMA class or medication. These results show the potential importance of the ASM/ceramide system in COVID-19 and support the continuation of FIASMA medications in these patients. Double-blind controlled randomized clinical trials of these medications for COVID-19 are needed.
Collapse
Affiliation(s)
- Nicolas Hoertel
- Assistance Publique-Hopitaux de Paris, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM, UMR_S1266, Université de Paris, Paris, France
| | - Marina Sánchez-Rico
- Assistance Publique-Hopitaux de Paris, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM, UMR_S1266, Université de Paris, Paris, France.,Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Erich Gulbins
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, Essen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alexander Carpinteiro
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, Essen, Germany.,Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Eric J Lenze
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Angela M Reiersen
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Miriam Abellán
- Assistance Publique-Hopitaux de Paris, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM, UMR_S1266, Université de Paris, Paris, France
| | - Pedro de la Muela
- Assistance Publique-Hopitaux de Paris, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM, UMR_S1266, Université de Paris, Paris, France.,Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Raphaël Vernet
- Medical Informatics, Biostatistics and Public Health Department, Assistance Publique-Hopitaux de Paris, Centre-Université de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Carlos Blanco
- Division of Epidemiology, Services, and Prevention Research, National Institute on Drug Abuse, North Bethesda, Maryland, USA
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, Toulouse, France
| | - Nathanaël Beeker
- Unité de Recherche clinique, Hopital Cochin, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Antoine Neuraz
- INSERM, UMR_S 1138, Cordeliers Research Center, Université de Paris, Paris, France.,Department of Medical Informatics, Necker-Enfants Malades Hospital, Assistance Publique-Hopitaux de Paris, Centre-Université de Paris, Paris, France
| | - Philip Gorwood
- INSERM, U1266 (Institute of Psychiatry and Neuroscience of Paris), Université de Paris, Paris, France
| | - Jesús M Alvarado
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Pierre Meneton
- INSERM U1142 LIMICS, UMRS 1142, Sorbonne Universities, UPMC University of Paris 06, University of Paris 13, Paris, France
| | - Frédéric Limosin
- Assistance Publique-Hopitaux de Paris, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM, UMR_S1266, Université de Paris, Paris, France
| | | |
Collapse
|
37
|
Chan SW, Shafi T, Ford RC. Kite-Shaped Molecules Block SARS-CoV-2 Cell Entry at a Post-Attachment Step. Viruses 2021; 13:v13112306. [PMID: 34835112 PMCID: PMC8619434 DOI: 10.3390/v13112306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Anti-viral small molecules are currently lacking for treating coronavirus infection. The long development timescales for such drugs are a major problem, but could be shortened by repurposing existing drugs. We therefore screened a small library of FDA-approved compounds for potential severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antivirals using a pseudovirus system that allows a sensitive read-out of infectivity. A group of structurally-related compounds, showing moderate inhibitory activity with IC50 values in the 2–5 μM range, were identified. Further studies demonstrated that these “kite-shaped” molecules were surprisingly specific for SARS-CoV-1 and SARS-CoV-2 and that they acted early in the entry steps of the viral infectious cycle, but did not affect virus attachment to the cells. Moreover, the compounds were able to prevent infection in both kidney- and lung-derived human cell lines. The structural homology of the hits allowed the production of a well-defined pharmacophore that was found to be highly accurate in predicting the anti-viral activity of the compounds in the screen. We discuss the prospects of repurposing these existing drugs for treating current and future coronavirus outbreaks.
Collapse
|
38
|
Forrestall KL, Burley DE, Cash MK, Pottie IR, Darvesh S. Phenothiazines as dual inhibitors of SARS-CoV-2 main protease and COVID-19 inflammation. CAN J CHEM 2021. [DOI: 10.1139/cjc-2021-0139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
COVID-19, caused by the severe acute respiratory coronavirus 2 (SARS-CoV-2), currently has no treatment for acute infection. The main protease (Mpro) of SARS-CoV-2 is an essential enzyme for viral replication and an attractive target for disease intervention. The phenothiazine moiety has demonstrated drug versatility for biological systems, including inhibition of butyrylcholinesterase, a property important in the cholinesterase anti-inflammatory cascade. Nineteen phenothiazine drugs were investigated using in silico modelling techniques to predict binding energies and inhibition constants (Ki values) with SARS-CoV-2 Mpro. Because most side-effects of phenothiazines are due to interactions with various neurotransmitter receptors and transporters, phenothiazines with few such interactions were also investigated. All compounds were found to bind to the active site of SARS-CoV-2 Mpro and showed Ki values ranging from 1.30 to 52.4 µM in a rigid active site. Nine phenothiazines showed inhibition constants <10 µM. The compounds with limited interactions with neurotransmitter receptors and transporters showed micromolar (µM) Ki values. Docking results were compared with remdesivir and showed similar interactions with key residues Glu-166 and Gln-189 in the active site. This work has identified several phenothiazines with limited neurotransmitter receptor and transporter interactions and that may provide the dual action of inhibiting SARS-CoV-2 Mpro to prevent viral replication and promote the release of anti-inflammatory cytokines to curb viral-induced inflammation. These compounds are promising candidates for further investigation against SARS-CoV-2.
Collapse
Affiliation(s)
- Katrina L. Forrestall
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Darcy E. Burley
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Meghan K. Cash
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Ian R. Pottie
- Department of Chemistry and Physics, Faculty of Arts and Science, Mount Saint Vincent University, Halifax, NS B3M 2J6, Canada
- Department of Chemistry, Faculty of Science, Saint Mary’s University, Halifax, NS B3H 3C3, Canada
| | - Sultan Darvesh
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Chemistry and Physics, Faculty of Arts and Science, Mount Saint Vincent University, Halifax, NS B3M 2J6, Canada
- Department of Medicine (Neurology & Geriatric Medicine), Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
39
|
Tummino TA, Rezelj VV, Fischer B, Fischer A, O'Meara MJ, Monel B, Vallet T, White KM, Zhang Z, Alon A, Schadt H, O'Donnell HR, Lyu J, Rosales R, McGovern BL, Rathnasinghe R, Jangra S, Schotsaert M, Galarneau JR, Krogan NJ, Urban L, Shokat KM, Kruse AC, García-Sastre A, Schwartz O, Moretti F, Vignuzzi M, Pognan F, Shoichet BK. Drug-induced phospholipidosis confounds drug repurposing for SARS-CoV-2. Science 2021; 373:541-547. [PMID: 34326236 PMCID: PMC8501941 DOI: 10.1126/science.abi4708] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/15/2021] [Indexed: 01/16/2023]
Abstract
Repurposing drugs as treatments for COVID-19, the disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has drawn much attention. Beginning with sigma receptor ligands and expanding to other drugs from screening in the field, we became concerned that phospholipidosis was a shared mechanism underlying the antiviral activity of many repurposed drugs. For all of the 23 cationic amphiphilic drugs we tested, including hydroxychloroquine, azithromycin, amiodarone, and four others already in clinical trials, phospholipidosis was monotonically correlated with antiviral efficacy. Conversely, drugs active against the same targets that did not induce phospholipidosis were not antiviral. Phospholipidosis depends on the physicochemical properties of drugs and does not reflect specific target-based activities-rather, it may be considered a toxic confound in early drug discovery. Early detection of phospholipidosis could eliminate these artifacts, enabling a focus on molecules with therapeutic potential.
Collapse
Affiliation(s)
- Tia A Tummino
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA
- Graduate Program in Pharmaceutical Sciences and Pharmacogenomics, UCSF, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| | - Veronica V Rezelj
- Institut Pasteur, Viral Populations and Pathogenesis Unit, CNRS UMR 3569, 75724 Paris, Cedex 15, France
| | - Benoit Fischer
- Novartis Institutes for BioMedical Research, Preclinical Safety, Basel, Switzerland
| | - Audrey Fischer
- Novartis Institutes for BioMedical Research, Preclinical Safety, Basel, Switzerland
| | - Matthew J O'Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Blandine Monel
- Institut Pasteur, Virus and Immunity Unit, CNRS UMR 3569, 75724 Paris, Cedex 15, France
| | - Thomas Vallet
- Institut Pasteur, Viral Populations and Pathogenesis Unit, CNRS UMR 3569, 75724 Paris, Cedex 15, France
| | - Kris M White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ziyang Zhang
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA
- Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA
| | - Assaf Alon
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Heiko Schadt
- Novartis Institutes for BioMedical Research, Preclinical Safety, Basel, Switzerland
| | - Henry R O'Donnell
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Jiankun Lyu
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| | - Romel Rosales
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Briana L McGovern
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raveen Rathnasinghe
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sonia Jangra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-René Galarneau
- Novartis Institutes for BioMedical Research, Preclinical Safety, Cambridge, MA, USA
| | - Nevan J Krogan
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA
| | - Laszlo Urban
- Novartis Institutes for BioMedical Research, Preclinical Safety, Cambridge, MA, USA
| | - Kevan M Shokat
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA
- Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olivier Schwartz
- Institut Pasteur, Virus and Immunity Unit, CNRS UMR 3569, 75724 Paris, Cedex 15, France
| | - Francesca Moretti
- Novartis Institutes for BioMedical Research, Preclinical Safety, Basel, Switzerland.
| | - Marco Vignuzzi
- Institut Pasteur, Viral Populations and Pathogenesis Unit, CNRS UMR 3569, 75724 Paris, Cedex 15, France.
| | - Francois Pognan
- Novartis Institutes for BioMedical Research, Preclinical Safety, Basel, Switzerland.
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA.
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| |
Collapse
|
40
|
Loas G, Le Corre P. Update on Functional Inhibitors of Acid Sphingomyelinase (FIASMAs) in SARS-CoV-2 Infection. Pharmaceuticals (Basel) 2021; 14:691. [PMID: 34358117 PMCID: PMC8308787 DOI: 10.3390/ph14070691] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/11/2022] Open
Abstract
The SARS-CoV-2 outbreak is characterized by the need of the search for curative drugs for treatment. In this paper, we present an update of knowledge about the interest of the functional inhibitors of acid sphingomyelinase (FIASMAs) in SARS-CoV-2 infection. Forty-nine FIASMAs have been suggested in the treatment of SARS-CoV-2 infection using in silico, in vitro or in vivo studies. Further studies using large-sized, randomized and double-blinded controlled clinical trials are needed to evaluate FIASMAs in SARS-CoV-2 infection as off-label therapy.
Collapse
Affiliation(s)
- Gwenolé Loas
- Department of Psychiatry, Hôpital Erasme, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
- Research Unit (ULB 266), Hôpital Erasme, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - Pascal Le Corre
- Pôle Pharmacie, Service Hospitalo-Universitaire de Pharmacie, CHU de Rennes, 35033 Rennes, France;
- Irset (Institut de Recherche en Santé, Environnement et Travail)-Inserm UMR 1085, University of Rennes, CHU Rennes, INSERM, EHESP, 35000 Rennes, France
- Laboratoire de Biopharmacie et Pharmacie Clinique, Faculté de Pharmacie, Université de Rennes 1, 35043 Rennes, France
| |
Collapse
|
41
|
Brimson JM, Prasanth MI, Malar DS, Brimson S, Thitilertdecha P, Tencomnao T. Drugs that offer the potential to reduce hospitalization and mortality from SARS-CoV-2 infection: The possible role of the sigma-1 receptor and autophagy. Expert Opin Ther Targets 2021; 25:435-449. [PMID: 34236922 PMCID: PMC8290373 DOI: 10.1080/14728222.2021.1952987] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
Introduction: Despite the availability of new vaccines for SARS-CoV-2, there has been slow uptake and problems with supply in some parts of the world. Hence, there is still a necessity for drugs that can prevent hospitalization of patients and reduce the strain on health care systems. Drugs with sigma affinity potentially provide protection against the most severe symptoms of SARS-COV-2 and could prevent mortality via interactions with the sigma-1 receptor.Areas covered: This review examines the role of the sigma-1 receptor and autophagy in SARS-CoV-2 infections and how they may be linked. The authors reveal how sigma ligands may reduce the symptoms, complications, and deaths resulting from SARS-CoV-2 and offer insights on those patient cohorts that may benefit most from these drugs.Expert opinion: Drugs with sigma affinity potentially offer protection against the most severe symptoms of SARS-CoV-2 via interactions with the sigma-1 receptor. Agonists of the sigma-1 receptor may provide protection of the mitochondria, activate mitophagy to remove damaged and leaking mitochondria, prevent ER stress, manage calcium ion transport, and induce autophagy to prevent cell death in response to infection.
Collapse
Affiliation(s)
- James Michael Brimson
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Dicson Sheeja Malar
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
42
|
Chan S, Shafi T, Ford RC. Kite-shaped molecules block SARS-CoV-2 cell entry at a post-attachment step.. [DOI: 10.1101/2021.05.29.446272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
ABSTRACTAnti-viral small molecules are currently lacking for treating coronavirus infection. The long development timescales for such drugs are a major problem, but could be shortened by repurposing existing drugs. We therefore screened a small library of FDA-approved compounds for potential severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antivirals using a pseudovirus system that allows a sensitive read-out of infectivity. A group of structurally-related compounds, showing moderate inhibitory activity with IC50values in the 1-5µM range, were identified. Further studies demonstrated that these ‘kite-shaped’ molecules were surprisingly specific for SARS-CoV and SARS-CoV-2 and that they acted early in the entry steps of the viral infectious cycle, but did not affect virus attachment to the cells. Moreover the compounds were able to prevent infection in both kidney- and lung-derived human cell lines. The structural homology of the hits allowed the production of a well-defined pharmacophore that was found to be highly accurate in predicting the anti-viral activity of the compounds in the screen. We discuss the prospects of repurposing these existing drugs for treating current and future coronavirus outbreaks.
Collapse
|
43
|
Hoertel N, Sánchez-Rico M, Gulbins E, Kornhuber J, Carpinteiro A, Lenze EJ, Reiersen AM, Abellán M, de la Muela P, Vernet R, Blanco C, Cougoule C, Beeker N, Neuraz A, Gorwood P, Alvarado JM, Meneton P, Limosin F. Association Between FIASMAs and Reduced Risk of Intubation or Death in Individuals Hospitalized for Severe COVID-19: An Observational Multicenter Study. Clin Pharmacol Ther 2021; 110:1498-1511. [PMID: 34050932 PMCID: PMC8239599 DOI: 10.1002/cpt.2317] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/20/2021] [Indexed: 12/31/2022]
Abstract
Several medications commonly used for a number of medical conditions share a property of functional inhibition of acid sphingomyelinase (ASM), or FIASMA. Preclinical and clinical evidence suggest that the ASM/ceramide system may be central to severe acute respiratory syndrome‐coronavirus 2 (SARS‐CoV‐2) infection. We examined the potential usefulness of FIASMA use among patients hospitalized for severe coronavirus disease 2019 (COVID‐19) in an observational multicenter study conducted at Greater Paris University hospitals. Of 2,846 adult patients hospitalized for severe COVID‐19, 277 (9.7%) were taking an FIASMA medication at the time of their hospital admission. The primary end point was a composite of intubation and/or death. We compared this end point between patients taking vs. not taking an FIASMA medication in time‐to‐event analyses adjusted for sociodemographic characteristics and medical comorbidities. The primary analysis was a Cox regression model with inverse probability weighting (IPW). Over a mean follow‐up of 9.2 days (SD = 12.5), the primary end point occurred in 104 patients (37.5%) receiving an FIASMA medication, and 1,060 patients (41.4%) who did not. Despite being significantly and substantially associated with older age and greater medical severity, FIASMA medication use was significantly associated with reduced likelihood of intubation or death in both crude (hazard ratio (HR) = 0.71, 95% confidence interval (CI) = 0.58–0.87, P < 0.001) and primary IPW (HR = 0.58, 95%CI = 0.46–0.72, P < 0.001) analyses. This association remained significant in multiple sensitivity analyses and was not specific to one particular FIASMA class or medication. These results show the potential importance of the ASM/ceramide system in COVID‐19 and support the continuation of FIASMA medications in these patients. Double‐blind controlled randomized clinical trials of these medications for COVID‐19 are needed.
Collapse
Affiliation(s)
- Nicolas Hoertel
- Assistance Publique-Hopitaux de Paris, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM, UMR_S1266, Université de Paris, Paris, France
| | - Marina Sánchez-Rico
- Assistance Publique-Hopitaux de Paris, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM, UMR_S1266, Université de Paris, Paris, France.,Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Erich Gulbins
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, Essen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alexander Carpinteiro
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, Essen, Germany.,Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Eric J Lenze
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Angela M Reiersen
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Miriam Abellán
- Assistance Publique-Hopitaux de Paris, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM, UMR_S1266, Université de Paris, Paris, France
| | - Pedro de la Muela
- Assistance Publique-Hopitaux de Paris, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM, UMR_S1266, Université de Paris, Paris, France.,Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Raphaël Vernet
- Medical Informatics, Biostatistics and Public Health Department, Assistance Publique-Hopitaux de Paris, Centre-Université de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Carlos Blanco
- Division of Epidemiology, Services, and Prevention Research, National Institute on Drug Abuse, North Bethesda, Maryland, USA
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, Toulouse, France
| | - Nathanaël Beeker
- Unité de Recherche clinique, Hopital Cochin, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Antoine Neuraz
- INSERM, UMR_S 1138, Cordeliers Research Center, Université de Paris, Paris, France.,Department of Medical Informatics, Necker-Enfants Malades Hospital, Assistance Publique-Hopitaux de Paris, Centre-Université de Paris, Paris, France
| | - Philip Gorwood
- INSERM, U1266 (Institute of Psychiatry and Neuroscience of Paris), Université de Paris, Paris, France
| | - Jesús M Alvarado
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Pierre Meneton
- INSERM U1142 LIMICS, UMRS 1142, Sorbonne Universities, UPMC University of Paris 06, University of Paris 13, Paris, France
| | - Frédéric Limosin
- Assistance Publique-Hopitaux de Paris, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM, UMR_S1266, Université de Paris, Paris, France
| | | |
Collapse
|
44
|
Villoutreix BO, Krishnamoorthy R, Tamouza R, Leboyer M, Beaune P. Chemoinformatic Analysis of Psychotropic and Antihistaminic Drugs in the Light of Experimental Anti-SARS-CoV-2 Activities. Adv Appl Bioinform Chem 2021; 14:71-85. [PMID: 33880039 PMCID: PMC8051956 DOI: 10.2147/aabc.s304649] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022] Open
Abstract
Introduction There is an urgent need to identify therapies that prevent SARS-CoV-2 infection and improve the outcome of COVID-19 patients. Objective Based upon clinical observations, we proposed that some psychotropic and antihistaminic drugs could protect psychiatric patients from SARS-CoV-2 infection. This observation is investigated in the light of experimental in vitro data on SARS-CoV-2. Methods SARS-CoV-2 high-throughput screening results are available at the NCATS COVID-19 portal. We investigated the in vitro anti-viral activity of many psychotropic and antihistaminic drugs using chemoinformatics approaches. Results and Discussion We analyze our clinical observations in the light of SARS-CoV-2 experimental screening results and propose that several cationic amphiphilic psychotropic and antihistaminic drugs could protect people from SARS-CoV-2 infection; some of these molecules have very limited adverse effects and could be used as prophylactic drugs. Other cationic amphiphilic drugs used in other disease areas are also highlighted. Recent analyses of patient electronic health records reported by several research groups indicate that some of these molecules could be of interest at different stages of the disease progression. In addition, recently reported drug combination studies further suggest that it might be valuable to associate several cationic amphiphilic drugs. Taken together, these observations underline the need for clinical trials to fully evaluate the potentials of these molecules, some fitting in the so-called category of broad-spectrum antiviral agents. Repositioning orally available drugs that have moderate side effects and should act on molecular mechanisms less prone to drug resistance would indeed be of utmost importance to deal with COVID-19.
Collapse
Affiliation(s)
- Bruno O Villoutreix
- INSERM U1141, NeuroDiderot, Université de Paris, Hôpital Robert-Debré, Paris, F-75019, France
| | - Rajagopal Krishnamoorthy
- Université Paris Est Créteil, INSERM U955, IMRB, Laboratoire Neuropsychiatrie Translationnelle, AP-HP, Département Medico-Universitaire de Psychiatrie et d'Addictologie (DMU ADAPT), Hôpital Henri Mondor, Fondation FondaMental, Créteil, F-94010, France
| | - Ryad Tamouza
- Université Paris Est Créteil, INSERM U955, IMRB, Laboratoire Neuropsychiatrie Translationnelle, AP-HP, Département Medico-Universitaire de Psychiatrie et d'Addictologie (DMU ADAPT), Hôpital Henri Mondor, Fondation FondaMental, Créteil, F-94010, France
| | - Marion Leboyer
- Université Paris Est Créteil, INSERM U955, IMRB, Laboratoire Neuropsychiatrie Translationnelle, AP-HP, Département Medico-Universitaire de Psychiatrie et d'Addictologie (DMU ADAPT), Hôpital Henri Mondor, Fondation FondaMental, Créteil, F-94010, France
| | - Philippe Beaune
- INSERM U1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, 75006, France
| |
Collapse
|
45
|
Kitagawa T, Matsumoto A, Terashima I, Uesono Y. Antimalarial Quinacrine and Chloroquine Lose Their Activity by Decreasing Cationic Amphiphilic Structure with a Slight Decrease in pH. J Med Chem 2021; 64:3885-3896. [PMID: 33775096 DOI: 10.1021/acs.jmedchem.0c02056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Quinacrine (QC) and chloroquine (CQ) have antimicrobial and antiviral activities as well as antimalarial activity, although the mechanisms remain unknown. QC increased the antimicrobial activity against yeast exponentially with a pH-dependent increase in the cationic amphiphilic drug (CAD) structure. CAD-QC localized in the yeast membranes and induced glucose starvation by noncompetitively inhibiting glucose uptake as antipsychotic chlorpromazine (CPZ) did. An exponential increase in antimicrobial activity with pH-dependent CAD formation was also observed for CQ, indicating that the CAD structure is crucial for its pharmacological activity. A decrease in CAD structure with a slight decrease in pH from 7.4 greatly reduced their effects; namely, these drugs would inefficiently act on falciparum malaria and COVID-19 pneumonia patients with acidosis, resulting in resistance. The decrease in CAD structure at physiological pH was not observed for quinine, primaquine, or mefloquine. Therefore, restoring the normal blood pH or using pH-insensitive quinoline drugs might be effective for these infectious diseases with acidosis.
Collapse
Affiliation(s)
- Tomohisa Kitagawa
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Atsushi Matsumoto
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ichiro Terashima
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yukifumi Uesono
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
46
|
Eugene AR, Eugene B, Masiak M, Masiak JS. Head-to-Head Comparison of Sedation and Somnolence Among 37 Antipsychotics in Schizophrenia, Bipolar Disorder, Major Depression, Autism Spectrum Disorders, Delirium, and Repurposed in COVID-19, Infectious Diseases, and Oncology From the FAERS, 2004-2020. Front Pharmacol 2021; 12:621691. [PMID: 33841149 PMCID: PMC8027114 DOI: 10.3389/fphar.2021.621691] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/02/2021] [Indexed: 11/21/2022] Open
Abstract
Objective: Antipsychotic compounds are known to induce sedation somnolence and have expanded clinical indications beyond schizophrenia to regulatory approval in bipolar disorder, treatment-resistant depression, and is being repurposed in infectious diseases and oncology. However, the medical sciences literature lacks a comprehensive association between sedation and somnolence among a wide-range of antipsychotic compounds. The objective of this study is to assess the disproportionality of sedation and somnolence among thirty-seven typical and atypical antipsychotics. Materials and Methods: Patient adverse drug reactions (ADR) cases were obtained from the United States Food and Drug Administration Adverse Events Reporting System (FAERS) between January 01, 2004 and September 30, 2020 for a wide-array of clinical indications and off-label use of antipsychotics. An assessment of disproportionality were based on cases of sedation and somnolence and calculated using the case/non-case methodology. Statistical analysis resulting in the reporting odds-ratio (ROR) with corresponding 95% confidence intervals (95% CI) were conducted using the R statistical programming language. Results: Throughout the reporting period, there were a total of 9,373,236 cases with 99,251 specific ADRs reporting sedation and somnolence. Zuclopenthixol (n = 224) ROR = 13.3 (95% CI, 11.6–15.3) was most strongly associated of sedation and somnolence and haloperidol decanoate long-acting injection (LAI) was not statistically associated sedation and somnolence. Further, among atypical antipsychotic compounds, tiapride and asenapine were the top two compounds most strongly associated with sedation and somnolence. Comprehensively, the typical antipsychotics ROR = 5.05 (95%CI, 4.97–5.12) had a stronger association with sedation and somnolence when compared to atypical antipsychotics ROR = 4.65 (95%CI, 4.47–4.84). Conclusion: We conducted a head-to-head comparison of thirty-seven antipsychotics and ranked the compounds based on the association of sedation and somnolence from ADR data collected throughout 16 years from the FAERS. The results are informative and with recent interests in repurposing phenothiazine antipsychotics in infectious disease and oncology provides an informative assessment of the compounds during repurposing and in psychopharmacology.
Collapse
Affiliation(s)
- Andy R Eugene
- Independent Neurophysiology Unit, Department of Psychiatry, Medical University of Lublin, Lublin, Poland
| | | | | | - Jolanta Sylwia Masiak
- Independent Neurophysiology Unit, Department of Psychiatry, Medical University of Lublin, Lublin, Poland.,Medical Center, Lublin, Poland.,II Department of Psychiatry and Psychiatric Rehabilitation, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
47
|
Tummino TA, Rezelj VV, Fischer B, Fischer A, O'Meara MJ, Monel B, Vallet T, Zhang Z, Alon A, O'Donnell HR, Lyu J, Schadt H, White KM, Krogan NJ, Urban L, Shokat KM, Kruse AC, García-Sastre A, Schwartz O, Moretti F, Vignuzzi M, Pognan F, Shoichet BK. Phospholipidosis is a shared mechanism underlying the in vitro antiviral activity of many repurposed drugs against SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.03.23.436648. [PMID: 33791693 PMCID: PMC8010720 DOI: 10.1101/2021.03.23.436648] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Repurposing drugs as treatments for COVID-19 has drawn much attention. A common strategy has been to screen for established drugs, typically developed for other indications, that are antiviral in cells or organisms. Intriguingly, most of the drugs that have emerged from these campaigns, though diverse in structure, share a common physical property: cationic amphiphilicity. Provoked by the similarity of these repurposed drugs to those inducing phospholipidosis, a well-known drug side effect, we investigated phospholipidosis as a mechanism for antiviral activity. We tested 23 cationic amphiphilic drugs-including those from phenotypic screens and others that we ourselves had found-for induction of phospholipidosis in cell culture. We found that most of the repurposed drugs, which included hydroxychloroquine, azithromycin, amiodarone, and four others that have already progressed to clinical trials, induced phospholipidosis in the same concentration range as their antiviral activity; indeed, there was a strong monotonic correlation between antiviral efficacy and the magnitude of the phospholipidosis. Conversely, drugs active against the same targets that did not induce phospholipidosis were not antiviral. Phospholipidosis depends on the gross physical properties of drugs, and does not reflect specific target-based activities, rather it may be considered a confound in early drug discovery. Understanding its role in infection, and detecting its effects rapidly, will allow the community to better distinguish between drugs and lead compounds that more directly impact COVID-19 from the large proportion of molecules that manifest this confounding effect, saving much time, effort and cost.
Collapse
Affiliation(s)
- Tia A Tummino
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA
- Graduate Program in Pharmaceutical Sciences and Pharmacogenomics, UCSF, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| | - Veronica V Rezelj
- Institut Pasteur, Viral Populations and Pathogenesis Unit, CNRS UMR 3569, 75724 Paris, Cedex 15, France
| | - Benoit Fischer
- Novartis Institutes for BioMedical Research, Preclinical Safety, Basel, Switzerland
| | - Audrey Fischer
- Novartis Institutes for BioMedical Research, Preclinical Safety, Basel, Switzerland
| | - Matthew J O'Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Blandine Monel
- Institut Pasteur, Virus and Immunity Unit, CNRS UMR 3569, 75724 Paris, Cedex 15, France
| | - Thomas Vallet
- Institut Pasteur, Viral Populations and Pathogenesis Unit, CNRS UMR 3569, 75724 Paris, Cedex 15, France
| | - Ziyang Zhang
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA
- Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA
| | - Assaf Alon
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Henry R O'Donnell
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Jiankun Lyu
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| | - Heiko Schadt
- Novartis Institutes for BioMedical Research, Preclinical Safety, Basel, Switzerland
| | - Kris M White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nevan J Krogan
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA
| | - Laszlo Urban
- Novartis Institutes for BioMedical Research, Preclinical Safety, Cambridge, MA, USA
| | - Kevan M Shokat
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA
- Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olivier Schwartz
- Institut Pasteur, Virus and Immunity Unit, CNRS UMR 3569, 75724 Paris, Cedex 15, France
| | - Francesca Moretti
- Novartis Institutes for BioMedical Research, Preclinical Safety, Basel, Switzerland
| | - Marco Vignuzzi
- Institut Pasteur, Viral Populations and Pathogenesis Unit, CNRS UMR 3569, 75724 Paris, Cedex 15, France
| | - Francois Pognan
- Novartis Institutes for BioMedical Research, Preclinical Safety, Basel, Switzerland
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| |
Collapse
|
48
|
Gitahy Falcao Faria C, Weiner L, Petrignet J, Hingray C, Ruiz De Pellon Santamaria Á, Villoutreix BO, Beaune P, Leboyer M, Javelot H. Antihistamine and cationic amphiphilic drugs, old molecules as new tools against the COVID-19? Med Hypotheses 2021; 148:110508. [PMID: 33571758 PMCID: PMC7830196 DOI: 10.1016/j.mehy.2021.110508] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/06/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022]
Abstract
Several studies have reported that certain psychoactive drugs could have a protective effect against SARS-CoV-2. Herein, we propose that antihistamines (anti-H1) and cationic amphiphilic drugs (CAD), specifically, have the capacity to disrupt virus entry and replication. In addition, several of these molecules have limited side effects and as such could be promising prophylactic candidates against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Clara Gitahy Falcao Faria
- Institute of Psychiatry, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil; Clinique de Psychiatrie, CHU de Strasbourg, France
| | - Luisa Weiner
- Clinique de Psychiatrie, CHU de Strasbourg, France
| | - Julien Petrignet
- Laboratoire Synthèse et Isolement de Molécules BioActives (SIMBA, EA 7502), Université de Tours, Faculté des Sciences et Techniques, Tours, France
| | - Coraline Hingray
- Pôle Hospitalo-Universitaire de Psychiatrie d'Adultes du Grand Nancy, Centre Psychothérapique de Nancy, Laxou, CHU de Nancy, Département de Neurologie, Nancy, France
| | | | - Bruno O Villoutreix
- Université de Paris, NeuroDiderot, Inserm U1141, hôpital Robert-Debré, F-75019 Paris, France
| | - Philippe Beaune
- Inserm U 1138 Centre de recherche des Cordeliers 75006 Paris, Université de Paris, Paris, France
| | - Marion Leboyer
- Université Paris Est Créteil (UPEC), INSERM U955, IMRB, Laboratoire Neuro-psychiatrie translationnelle, AP-HP, Département Medico-Universitaire de Psychiatrie et d'Addictologie (DMU ADAPT), Hôpital Henri Mondor, Fondation FondaMental, F-94010 Créteil, France
| | - Hervé Javelot
- Etablissement Public de Santé Alsace Nord, Brumath, France Laboratoire de Toxicologie et Pharmacologie Neuro Cardiovasculaire, Université de Strasbourg, Strasbourg, France.
| |
Collapse
|