1
|
Kelly JA, Aida-Ficken V, McMullan LK, Chatterjee P, Shrivastava-Ranjan P, Marot S, Jenks MH, Lo MK, Montgomery JM, Spiropoulou CF, Flint M. Mechanisms of action of repurposed Ebola virus antivirals - the roles of phospholipidosis and cholesterol homeostasis. Antiviral Res 2025; 238:106167. [PMID: 40245950 DOI: 10.1016/j.antiviral.2025.106167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Cell-based drug repurposing screens have been a common approach to identifying compounds with antiviral properties. For Ebola virus (EBOV), such screens yield unexpectedly high hit rates. We investigated two mechanisms underlying the anti-EBOV activities of repurposed compounds. Phospholipidosis (PLD) is excessive accumulation of cellular lipids that confounds screens for SARS-CoV-2. We performed a meta-analysis of published screens and supplemented these with our own using infectious EBOV at biosafety level-4. A list of nearly 400 hit compounds from seven anti-EBOV screens was compiled. Most (63 %) of these hits were predicted to induce PLD, and their anti-EBOV activities broadly correlated with PLD induction. PLD-inducing compounds did not inhibit infection by several other highly pathogenic viruses, suggesting that PLD was not a confounding factor for screens against Lassa, Crimean-Congo hemorrhagic fever, and Rift Valley fever viruses. Of four cells lines tested, HeLa cells were the least susceptible to PLD induction. In addition to PLD, many of the hit compounds identified disrupt cholesterol homeostasis. Previous research found inhibition of cholesterol synthesis by statins blocked EBOV infection. To understand if compounds inhibiting this mechanism could contribute to high hit rates, we further examined this pathway. We identified multiple additional inhibitors of cholesterol biosynthesis, that also blocked EBOV infection, albeit with varying potency and cytotoxicity across cell lines. EBOV inhibitors that acted through this mechanism were suppressed by the addition of exogenous cholesterol. Our findings help define the effects that contribute to anti-EBOV activities and hence facilitate the selection of lead molecules suitable for subsequent development.
Collapse
Affiliation(s)
- Jamie A Kelly
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Virginia Aida-Ficken
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA; Auburn University College of Veterinary Medicine, Department of Pathobiology, Auburn, AL, USA
| | - Laura K McMullan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Payel Chatterjee
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Punya Shrivastava-Ranjan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Stéphane Marot
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - M Harley Jenks
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Michael K Lo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Joel M Montgomery
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA.
| | - Mike Flint
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA.
| |
Collapse
|
2
|
Durante D, Bott R, Cooper L, Owen C, Morsheimer KM, Patten J, Zielinski C, Peet NP, Davey RA, Gaisina IN, Rong L, Moore TW. N-Substituted Pyrrole-Based Heterocycles as Broad-Spectrum Filoviral Entry Inhibitors. J Med Chem 2024; 67:13737-13764. [PMID: 39169825 PMCID: PMC11812679 DOI: 10.1021/acs.jmedchem.4c00527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Since the largest and most fatal Ebola virus epidemic during 2014-2016, there have been several consecutive filoviral outbreaks in recent years, including those in 2021, 2022, and 2023. Ongoing outbreak prevalence and limited FDA-approved filoviral therapeutics emphasize the need for novel small molecule treatments. Here, we showcase the structure-activity relationship development of N-substituted pyrrole-based heterocycles and their potent, submicromolar entry inhibition against diverse filoviruses in a target-based pseudovirus assay. Inhibitor antiviral activity was validated using replication-competent Ebola, Sudan, and Marburg viruses. Mutational analysis was used to map the targeted region within the Ebola virus glycoprotein. Antiviral counter-screen and phospholipidosis assays were performed to demonstrate the reduced off-target activity of these filoviral entry inhibitors. Favorable antiviral potency, selectivity, and drug-like properties of the N-substituted pyrrole-based heterocycles support their potential as broad-spectrum antifiloviral treatments.
Collapse
Affiliation(s)
- Destiny Durante
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Ryan Bott
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Laura Cooper
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Callum Owen
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, MA 02118, United States
| | - Kimberly M. Morsheimer
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, MA 02118, United States
| | - J.J. Patten
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, MA 02118, United States
| | - Christian Zielinski
- UICentre: Drug Discovery, University of Illinois Chicago, Chicago, IL, 60612, United States
| | - Norton P. Peet
- Chicago BioSolutions Inc., Chicago, IL 60612, United States
| | - Robert A. Davey
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, MA 02118, United States
| | - Irina N. Gaisina
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, United States
- Chicago BioSolutions Inc., Chicago, IL 60612, United States
- UICentre: Drug Discovery, University of Illinois Chicago, Chicago, IL, 60612, United States
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, United States
- Chicago BioSolutions Inc., Chicago, IL 60612, United States
| | - Terry W. Moore
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, United States
- University of Illinois Cancer Center, University of Illinois Chicago, Chicago, IL 60612, United States
| |
Collapse
|
3
|
Tang W, Tang Z, Liu H, Lu J, Du Q, Tian H, Li J. Xanthohumol and echinocystic acid induces PSTVd tolerance in tomato. PLANT DIRECT 2024; 8:e612. [PMID: 38911016 PMCID: PMC11190350 DOI: 10.1002/pld3.612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 06/25/2024]
Abstract
Tomato is a popular vegetable worldwide; its production is highly threatened by infection with the potato spindle tuber viroid (PSTVd). We obtained the full-length genome sequence of previously conserved PSTVd and inoculated it on four genotypes of semi-cultivated tomatoes selected from a local tomato germplasm resource. SC-5, which is a PSTVd-resistant genotype, and SC-96, which is a PSTVd-sensitive genotype, were identified by detecting the fruit yield, plant growth, biomass accumulation, physiological indices, and PSTVd genome titer after PSTVd inoculation. A non-target metabolomics study was conducted on PSTVd-infected and control SC-5 to identify potential anti-PSTVd metabolites. The platform of liquid chromatography-mass spectrometry detected 158 or 123 differential regulated metabolites in modes of positive ion or negative ion. Principal component analysis revealed a clear separation of the global metabolite profile between PSTVd-infected leaves and control regardless of the detection mode. The potential anti-PSTVd compounds, xanthohumol, oxalicine B, indole-3-carbinol, and rosmarinic acid were significantly upregulated in positive ion mode, whereas echinocystic acid, chlorogenic acid, and 5-acetylsalicylic acid were upregulated in negative ion mode. Xanthohumol and echinocystic acid were detected as the most upregulated metabolites and were exogenously applied on PSTVd-diseased SC-96 seedlings. Both xanthohumol and echinocystic acid had instant and long-term inhibition effect on PSTVd titer. The highest reduction of disease symptom was induced by 2.6 mg/L of xanthohumol and 2.0 mg/L of echinocystic acid after 10 days of leaf spraying, respectively. A superior effect was seen on echinocystic acid than on xanthohumol. Our study provides a statistical basis for breeding anti-viroid tomato genotypes and creating plant-originating chemical preparations to prevent viroid disease.
Collapse
Affiliation(s)
- Wenkun Tang
- Vegetable Industry Research InstituteGuizhou UniversityGuiyangChina
- College of AgricultureGuizhou UniversityGuiyangChina
| | - Zhichao Tang
- Vegetable Industry Research InstituteGuizhou UniversityGuiyangChina
- College of AgricultureGuizhou UniversityGuiyangChina
| | - Haiyi Liu
- Vegetable Industry Research InstituteGuizhou UniversityGuiyangChina
- College of AgricultureGuizhou UniversityGuiyangChina
| | - Jinbiao Lu
- Vegetable Industry Research InstituteGuizhou UniversityGuiyangChina
- College of AgricultureGuizhou UniversityGuiyangChina
| | - Qianyun Du
- Guizhou Advanced Seed Industry GroupGuiyangChina
| | - Huan Tian
- Guizhou Advanced Seed Industry GroupGuiyangChina
| | - Jingwei Li
- Vegetable Industry Research InstituteGuizhou UniversityGuiyangChina
- College of AgricultureGuizhou UniversityGuiyangChina
| |
Collapse
|
4
|
Li S, Li H, Lian R, Xie J, Feng R. New perspective of small-molecule antiviral drugs development for RNA viruses. Virology 2024; 594:110042. [PMID: 38492519 DOI: 10.1016/j.virol.2024.110042] [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: 10/21/2023] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 03/18/2024]
Abstract
High variability and adaptability of RNA viruses allows them to spread between humans and animals, causing large-scale infectious diseases which seriously threat human and animal health and social development. At present, AIDS, viral hepatitis and other viral diseases with high incidence and low cure rate are still spreading around the world. The outbreaks of Ebola, Zika, dengue and in particular of the global pandemic of COVID-19 have presented serious challenges to the global public health system. The development of highly effective and broad-spectrum antiviral drugs is a substantial and urgent research subject to deal with the current RNA virus infection and the possible new viral infections in the future. In recent years, with the rapid development of modern disciplines such as artificial intelligence technology, bioinformatics, molecular biology, and structural biology, some new strategies and targets for antivirals development have emerged. Here we review the main strategies and new targets for developing small-molecule antiviral drugs against RNA viruses through the analysis of the new drug development progress against several highly pathogenic RNA viruses, to provide clues for development of future antivirals.
Collapse
Affiliation(s)
- Shasha Li
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China; Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Huixia Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Ruiya Lian
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China; Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Jingying Xie
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China; Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Ruofei Feng
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China.
| |
Collapse
|
5
|
Soni M, Tulsian K, Barot P, Vyas VK. Recent Advances in Therapeutic Approaches Against Ebola Virus Infection. RECENT ADVANCES IN ANTI-INFECTIVE DRUG DISCOVERY 2024; 19:276-299. [PMID: 38279760 DOI: 10.2174/0127724344267452231206061944] [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: 06/15/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Ebola virus (EBOV) is a genus of negative-strand RNA viruses belonging to the family Filoviradae that was first described in 1976 in the present-day Democratic Republic of the Congo. It has intermittently affected substantial human populations in West Africa and presents itself as a global health menace due to the high mortality rate of patients, high transmission rate, difficult patient management, and the emergence of complicated autoimmune disease-like conditions post-infection. OBJECTIVE EBOV or other EBOV-like species as a biochemical weapon pose a significant risk; hence, the need to develop both prophylactic and therapeutic medications to combat the virus is unquestionable. METHODS In this review work, we have compiled the literature pertaining to transmission, pathogenesis, immune response, and diagnosis of EBOV infection. We included detailed structural details of EBOV along with all the available therapeutics against EBOV disease. We have also highlighted current developments and recent advances in therapeutic approaches against Ebola virus disease (EVD). DISCUSSION The development of preventive vaccines against the virus is proving to be a successful effort as of now; however, problems concerning logistics, product stability, multi- dosing, and patient tracking are prominent in West Africa. Monoclonal antibodies that target EBOV proteins have also been developed and approved in the clinic; however, no small drug molecules that target these viral proteins have cleared clinical trials. An understanding of clinically approved vaccines and their shortcomings also serves an important purpose for researchers in vaccine design in choosing the right vector, antigen, and particular physicochemical properties that are critical for the vaccine's success against the virus across the world. CONCLUSION Our work brings together a comprehensive review of all available prophylactic and therapeutic medications developed and under development against the EBOV, which will serve as a guide for researchers in pursuing the most promising drug discovery strategies against the EBOV and also explore novel mechanisms of fighting against EBOV infection.
Collapse
Affiliation(s)
- Molisha Soni
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Kartik Tulsian
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Parv Barot
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Vivek Kumar Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
6
|
Shrivastava-Ranjan P, Jain S, Chatterjee P, Montgomery JM, Flint M, Albariño C, Spiropoulou CF. Development of a novel minigenome and recombinant VSV expressing Seoul hantavirus glycoprotein-based assays to identify anti-hantavirus therapeutics. Antiviral Res 2023; 214:105619. [PMID: 37142192 DOI: 10.1016/j.antiviral.2023.105619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
Seoul virus (SEOV) is an emerging global health threat that can cause hemorrhagic fever with renal syndrome (HFRS), which results in case fatality rates of ∼2%. There are no approved treatments for SEOV infections. We developed a cell-based assay system to identify potential antiviral compounds for SEOV and generated additional assays to characterize the mode of action of any promising antivirals. To test if candidate antivirals targeted SEOV glycoprotein-mediated entry, we developed a recombinant reporter vesicular stomatitis virus expressing SEOV glycoproteins. To facilitate the identification of candidate antiviral compounds targeting viral transcription/replication, we successfully generated the first reported minigenome system for SEOV. This SEOV minigenome (SEOV-MG) screening assay will also serve as a prototype assay for discovery of small molecules inhibiting replication of other hantaviruses, including Andes and Sin Nombre viruses. Ours is a proof-of-concept study in which we tested several compounds previously reported to have activity against other negative-strand RNA viruses using our newly developed hantavirus antiviral screening systems. These systems can be used under lower biocontainment conditions than those needed for infectious viruses, and identified several compounds with robust anti-SEOV activity. Our findings have important implications for the development of anti-hantavirus therapeutics.
Collapse
Affiliation(s)
- Punya Shrivastava-Ranjan
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Shilpi Jain
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Payel Chatterjee
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Joel M Montgomery
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mike Flint
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - César Albariño
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
7
|
Bílek R, Danzig V, Grimmichová T. Antiviral activity of amiodarone in SARS-CoV-2 disease. Physiol Res 2022. [DOI: 10.33549/physiolres.934974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Amiodarone seems to exhibit some antiviral activity in the disease caused by SARS-CoV-2. Here we have examined the SARS-CoV-2 disease course in the entire population of the Czech Republic and compared it with the course of the disease in patients treated with amiodarone in two major Prague’s hospitals. In the whole population of the Czech Republic SARS-CoV-2 infected 1665070 persons (15.6 %) out of 10694000 (100 %) between 1 April 2020 and 30 June 2021. In the same time period only 35 patients (3.4 %) treated with amiodarone were infected with SARS-CoV-2 virus out of 1032 patients (100 %) who received amiodarone. It appears that amiodarone can prevent SARS-CoV-2 virus infection by multiple mechanisms. In in-vitro experiments it exhibits SARS-CoV-2 virus replication inhibitions. Due to its anti-inflammatory and antioxidant properties, it may have beneficial effect on the complications caused by SARS-CoV-2 as well. Additionally, inorganic iodine released from amiodarone can be converted to hypoiodite (IO-), which has antiviral and antibacterial activity, and thus can affect the life cycle of the virus.
Collapse
Affiliation(s)
- R Bílek
- Institute of Endocrinology, Národní 8, 110 00 Prague 1, Czech Republic. ,
| | | | | |
Collapse
|
8
|
Russell T, Gangotia D, Barry G. Assessing the potential of repurposing ion channel inhibitors to treat emerging viral diseases and the role of this host factor in virus replication. Biomed Pharmacother 2022; 156:113850. [DOI: 10.1016/j.biopha.2022.113850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/25/2022] [Accepted: 10/06/2022] [Indexed: 12/03/2022] Open
|
9
|
BÍLEK R, DANZIG V, GRIMMICHOVÁ T. Antiviral activity of amiodarone in SARS-CoV-2 disease. Physiol Res 2022; 71:869-875. [PMID: 36426888 PMCID: PMC9814990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Amiodarone seems to exhibit some antiviral activity in the disease caused by SARS-CoV-2. Here we have examined the SARS-CoV-2 disease course in the entire population of the Czech Republic and compared it with the course of the disease in patients treated with amiodarone in two major Prague's hospitals. In the whole population of the Czech Republic SARS-CoV-2 infected 1665070 persons (15.6 %) out of 10694000 (100 %) between 1 April 2020 and 30 June 2021. In the same time period only 35 patients (3.4 %) treated with amiodarone were infected with SARS-CoV-2 virus out of 1032 patients (100 %) who received amiodarone. It appears that amiodarone can prevent SARS-CoV-2 virus infection by multiple mechanisms. In in-vitro experiments it exhibits SARS-CoV-2 virus replication inhibitions. Due to its anti-inflammatory and antioxidant properties, it may have beneficial effect on the complications caused by SARS-CoV-2 as well. Additionally, inorganic iodine released from amiodarone can be converted to hypoiodite (IO-), which has antiviral and antibacterial activity, and thus can affect the life cycle of the virus.
Collapse
Affiliation(s)
| | - Vilém DANZIG
- 2nd Department of Medicine, Cardiology and Angiology, Charles University, General University Hospital, Prague, Czech Republic
| | - Tereza GRIMMICHOVÁ
- Institute of Endocrinology, Prague, Czech Republic,The University Hospital Kralovske Vinohrady, Dept. of Internal Medicine, Division of Diabetology and Endocrinology, Prague, Czech Republic
| |
Collapse
|
10
|
Zhang Y, Huang Y, Xu Y. Antiviral Treatment Options for Severe Fever with Thrombocytopenia Syndrome Infections. Infect Dis Ther 2022; 11:1805-1819. [PMID: 36136218 PMCID: PMC9510271 DOI: 10.1007/s40121-022-00693-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/05/2022] [Indexed: 11/28/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome virus (SFTSV) is a tick-borne virus that produces severe fever with thrombocytopenia syndrome (SFTS). It is widespread in Japan, South Korea, and Central and Eastern China. The epidemic has developed rapidly through China in recent years. SFTS cases have been reported in 25 provinces in China, mainly distributed in rural areas in mountainous and hilly areas. The infection has a high case fatality rate and no specific treatments or vaccinations. Therefore, early diagnosis and treatment of SFTS infection is important to survival and disease control. In this article, we provide an overview on different aspects of SFTS with an emphasis on management, to explore the current treatment and prophylactic measures further.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Rd, Hefei, China
| | - Ying Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Rd, Hefei, China.
| | - Yuanhong Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Rd, Hefei, China.
| |
Collapse
|
11
|
Peng H, Ding C, Jiang L, Tang W, Liu Y, Zhao L, Yi Z, Ren H, Li C, He Y, Zheng X, Tang H, Chen Z, Qi Z, Zhao P. Discovery of potential anti-SARS-CoV-2 drugs based on large-scale screening in vitro and effect evaluation in vivo. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1181-1197. [PMID: 34962614 PMCID: PMC8713546 DOI: 10.1007/s11427-021-2031-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/17/2021] [Indexed: 12/22/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a global crisis. Clinical candidates with high efficacy, ready availability, and that do not develop resistance are in urgent need. Despite that screening to repurpose clinically approved drugs has provided a variety of hits shown to be effective against SARS-CoV-2 infection in cell culture, there are few confirmed antiviral candidates in vivo. In this study, 94 compounds showing high antiviral activity against SARS-CoV-2 in Vero E6 cells were identified from 2,580 FDA-approved small-molecule drugs. Among them, 24 compounds with low cytotoxicity were selected, and of these, 17 compounds also effectively suppressed SARS-CoV-2 infection in HeLa cells transduced with human ACE2. Six compounds disturb multiple processes of the SARS-CoV-2 life cycle. Their prophylactic efficacies were determined in vivo using Syrian hamsters challenged with SARS-CoV-2 infection. Seven compounds reduced weight loss and promoted weight regain of hamsters infected not only with the original strain but also the D614G variant. Except for cisatracurium, six compounds reduced hamster pulmonary viral load, and IL-6 and TNF-α mRNA when assayed at 4 d postinfection. In particular, sertraline, salinomycin, and gilteritinib showed similar protective effects as remdesivir in vivo and did not induce antiviral drug resistance after 10 serial passages of SARS-CoV-2 in vitro, suggesting promising application for COVID-19 treatment.
Collapse
Affiliation(s)
- Haoran Peng
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China
| | - Cuiling Ding
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China
| | - Liangliang Jiang
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China
| | - Wanda Tang
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China
| | - Yan Liu
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China
| | - Lanjuan Zhao
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China
| | - Zhigang Yi
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hao Ren
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China
| | - Chong Li
- Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200000, China
| | - Yanhua He
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China
| | - Xu Zheng
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China
| | - Hailin Tang
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China
| | - Zhihui Chen
- Department of Infectious Disease, Changhai Hospital, Shanghai, 200433, China.
| | - Zhongtian Qi
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China.
| | - Ping Zhao
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China.
| |
Collapse
|
12
|
Silva-Ramos CR, Montoya-Ruíz C, Faccini-Martínez ÁA, Rodas JD. An updated review and current challenges of Guanarito virus infection, Venezuelan hemorrhagic fever. Arch Virol 2022; 167:1727-1738. [PMID: 35579715 PMCID: PMC9110938 DOI: 10.1007/s00705-022-05453-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 03/08/2022] [Indexed: 11/30/2022]
Abstract
Guanarito virus (GTOV) is a member of the family Arenaviridae and has been designated a category A bioterrorism agent by the US Centers for Disease Control and Prevention. It is endemic to Venezuela's western region, and it is the etiological agent of "Venezuelan hemorrhagic fever" (VHF). Similar to other arenaviral hemorrhagic fevers, VHF is characterized by fever, mild hemorrhagic signs, nonspecific symptoms, thrombocytopenia, and leukopenia. Patients with severe disease usually develop signs of internal bleeding. Due to the absence of reference laboratories that can handle GTOV in endemic areas, diagnosis is primarily clinical and epidemiological. No antiviral therapies are available; thus, treatment includes only supportive analgesia and fluids. GTOV is transmitted by contact with the excreta of its rodent reservoir, Zygodontomys brevicauda. The main reasons for the emergence of the disease may be the increase in the human population, migration, and changes in land use patterns in rural areas. Social and environmental changes could make VHF an important cause of underdiagnosed acute febrile illnesses in regions near the endemic areas. Although there is evidence that GTOV circulates among rodents in different Venezuelan states, VHF cases have only been reported in the states of Portuguesa and Barinas. However, due to the increased frequency of invasions by humans into wildlife habitats, it is probable that VHF could become a public health problem in the nearby regions of Colombia and Brazil. The current Venezuelan political crisis is causing an increase in the migration of people and livestock, representing a risk for the redistribution and re-emergence of infectious diseases.
Collapse
Affiliation(s)
- Carlos Ramiro Silva-Ramos
- Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Carolina Montoya-Ruíz
- Facultad de Ciencias, Universidad Nacional de Colombia, Carrera 65, #59a, 110, Medellín, Antioquia, Colombia.
| | - Álvaro A Faccini-Martínez
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.,Comité de Medicina Tropical, Zoonosis y Medicina del Viajero, Asociación Colombiana de Infectología, Bogotá, Colombia
| | - Juan David Rodas
- Grupo de Investigación en Ciencias Veterinarias Centauro, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
13
|
di Filippo L, Doga M, Frara S, Giustina A. Hypocalcemia in COVID-19: Prevalence, clinical significance and therapeutic implications. Rev Endocr Metab Disord 2022; 23:299-308. [PMID: 33846867 PMCID: PMC8041474 DOI: 10.1007/s11154-021-09655-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/09/2021] [Indexed: 01/08/2023]
Abstract
COVID-19 extra-pulmonary features include several endocrine manifestations and these are becoming strongly clinically relevant in patients affected influencing disease severity and outcomes.At the beginning of COVID-19 pandemic no population data on calcium levels in patients affected were available and in April 2020 a first case of severe acute hypocalcemia in an Italian patient with SARS-CoV-2 infection was reported. Subsequently, several studies reported hypocalcemia as a highly prevalent biochemical abnormality in COVID-19 patients with a marked negative influence on disease severity, biochemical inflammation and thrombotic markers, and mortality. Also a high prevalence of vertebral fractures with worse respiratory impairment in patients affected and a widespread vitamin D deficiency have been frequently observed, suggesting an emerging "Osteo-Metabolic Phenotype" in COVID-19.To date, several potential pathophysiological factors have been hypothesized to play a role in determining hypocalcemia in COVID-19 including calcium dependent viral mechanisms of action, high prevalence of hypovitaminosis D in general population, chronic and acute malnutrition during critical illness and high levels of unbound and unsaturated fatty acids in inflammatory responses.Since hypocalcemia is a frequent biochemical finding in hospitalized COVID-19 patients possibly predicting worse outcomes and leading to acute cardiovascular and neurological complications if severe, it is reasonable to assess, monitor and, if indicated, replace calcium at first patient hospital evaluation and during hospitalization.
Collapse
Affiliation(s)
- Luigi di Filippo
- Institute of Endocrine and Metabolic Sciences, Università Vita-Salute San Raffaele, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Mauro Doga
- Institute of Endocrine and Metabolic Sciences, Università Vita-Salute San Raffaele, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Stefano Frara
- Institute of Endocrine and Metabolic Sciences, Università Vita-Salute San Raffaele, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Giustina
- Institute of Endocrine and Metabolic Sciences, Università Vita-Salute San Raffaele, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
14
|
Liu CH, Hu YT, Wong SH, Lin LT. Therapeutic Strategies against Ebola Virus Infection. Viruses 2022; 14:v14030579. [PMID: 35336986 PMCID: PMC8954160 DOI: 10.3390/v14030579] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 12/10/2022] Open
Abstract
Since the 2014–2016 epidemic, Ebola virus (EBOV) has spread to several countries and has become a major threat to global health. EBOV is a risk group 4 pathogen, which imposes significant obstacles for the development of countermeasures against the virus. Efforts have been made to develop anti-EBOV immunization and therapeutics, with three vaccines and two antibody-based therapeutics approved in recent years. Nonetheless, the high fatality of Ebola virus disease highlights the need to continuously develop antiviral strategies for the future management of EBOV outbreaks in conjunction with vaccination programs. This review aims to highlight potential EBOV therapeutics and their target(s) of inhibition, serving as a summary of the literature to inform readers of the novel candidates available in the continued search for EBOV antivirals.
Collapse
Affiliation(s)
- Ching-Hsuan Liu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Yee-Tung Hu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Shu Hui Wong
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Correspondence:
| |
Collapse
|
15
|
Saurav S, Tanwar J, Ahuja K, Motiani RK. Dysregulation of host cell calcium signaling during viral infections: Emerging paradigm with high clinical relevance. Mol Aspects Med 2021; 81:101004. [PMID: 34304899 PMCID: PMC8299155 DOI: 10.1016/j.mam.2021.101004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/18/2021] [Accepted: 07/16/2021] [Indexed: 12/22/2022]
Abstract
Viral infections are one of the leading causes of human illness. Viruses take over host cell signaling cascades for their replication and infection. Calcium (Ca2+) is a versatile and ubiquitous second messenger that modulates plethora of cellular functions. In last two decades, a critical role of host cell Ca2+ signaling in modulating viral infections has emerged. Furthermore, recent literature clearly implicates a vital role for the organellar Ca2+ dynamics (influx and efflux across organelles) in regulating virus entry, replication and severity of the infection. Therefore, it is not surprising that a number of viral infections including current SARS-CoV-2 driven COVID-19 pandemic are associated with dysregulated Ca2+ homeostasis. The focus of this review is to first discuss the role of host cell Ca2+ signaling in viral entry, replication and egress. We further deliberate on emerging literature demonstrating hijacking of the host cell Ca2+ dynamics by viruses. In particular, a variety of viruses including SARS-CoV-2 modulate lysosomal and cytosolic Ca2+ signaling for host cell entry and replication. Moreover, we delve into the recent studies, which have demonstrated the potential of several FDA-approved drugs targeting Ca2+ handling machinery in inhibiting viral infections. Importantly, we discuss the prospective of targeting intracellular Ca2+ signaling for better management and treatment of viral pathogenesis including COVID-19. Finally, we highlight the key outstanding questions in the field that demand critical and timely attention.
Collapse
Affiliation(s)
- Suman Saurav
- Laboratory of Calciomics and Systemic Pathophysiology, Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India
| | - Jyoti Tanwar
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi-110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Kriti Ahuja
- Laboratory of Calciomics and Systemic Pathophysiology, Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India
| | - Rajender K Motiani
- Laboratory of Calciomics and Systemic Pathophysiology, Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India.
| |
Collapse
|
16
|
Sikdar A, Gupta R, Boura E. Reviewing Antiviral Research Against Viruses Causing Human Diseases - A Structure Guided Approach. Curr Mol Pharmacol 2021; 15:306-337. [PMID: 34348638 DOI: 10.2174/1874467214666210804152836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 11/22/2022]
Abstract
The littlest of all the pathogens, viruses have continuously been the foremost strange microorganisms to consider. Viral Infections can cause extreme sicknesses as archived by the HIV/AIDS widespread or the later Ebola or Zika episodes. Apprehensive framework distortions are too regularly watched results of numerous viral contaminations. Besides, numerous infections are oncoviruses, which can trigger different sorts of cancer. Nearly every year a modern infection species rises debilitating the world populace with an annihilating episode. Subsequently, the need of creating antivirals to combat such rising infections. In any case, from the innovation of to begin with antiviral medicate Idoxuridine in 1962 to the revelation of Baloxavir marboxil (Xofluza) that was FDA-approved in 2018, the hone of creating antivirals has changed significantly. In this article, different auxiliary science strategies have been described that can be referral for therapeutics innovation.
Collapse
Affiliation(s)
- Arunima Sikdar
- Department of Hematology and Oncology, School of Medicine, The University of Tennessee Health Science Center, 920 Madison Ave, P.O.Box-38103, Memphis, Tennessee. United States
| | - Rupali Gupta
- Department of Neurology, Duke University Medical Center, Durham, North Carolina. United States
| | - Evzen Boura
- Department of Molecular Biology and Biochemistry, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, P.O. Box:16000, Prague. Czech Republic
| |
Collapse
|
17
|
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: 144] [Impact Index Per Article: 36.0] [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
|
18
|
Screening and Identification of Lujo Virus Inhibitors Using a Recombinant Reporter Virus Platform. Viruses 2021; 13:v13071255. [PMID: 34203149 PMCID: PMC8310135 DOI: 10.3390/v13071255] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022] Open
Abstract
Lujo virus (LUJV), a highly pathogenic arenavirus, was first identified in 2008 in Zambia. To aid the identification of effective therapeutics for LUJV, we developed a recombinant reporter virus system, confirming reporter LUJV comparability with wild-type virus and its utility in high-throughput antiviral screening assays. Using this system, we evaluated compounds with known and unknown efficacy against related arenaviruses, with the aim of identifying LUJV-specific and potential new pan-arenavirus antivirals. We identified six compounds demonstrating robust anti-LUJV activity, including several compounds with previously reported activity against other arenaviruses. These data provide critical evidence for developing broad-spectrum antivirals against high-consequence arenaviruses.
Collapse
|
19
|
Giri A, Srinivasan A, Sundar IK. COVID-19: Sleep, Circadian Rhythms and Immunity - Repurposing Drugs and Chronotherapeutics for SARS-CoV-2. Front Neurosci 2021; 15:674204. [PMID: 34220430 PMCID: PMC8249936 DOI: 10.3389/fnins.2021.674204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/05/2021] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has affected nearly 28 million people in the United States and has caused more than five hundred thousand deaths as of February 21, 2021. As the novel coronavirus continues to take its toll in the United States and all across the globe, particularly among the elderly (>65 years), clinicians and translational researchers are taking a closer look at the nexus of sleep, circadian rhythms and immunity that may contribute toward a more severe coronavirus disease-19 (COVID-19). SARS-CoV-2-induced multi-organ failure affects both central and peripheral organs, causing increased mortality in the elderly. However, whether differences in sleep, circadian rhythms, and immunity between older and younger individuals contribute to the age-related differences in systemic dysregulation of target organs observed in SARS-CoV-2 infection remain largely unknown. Current literature demonstrates the emerging role of sleep, circadian rhythms, and immunity in the development of chronic pulmonary diseases and respiratory infections in human and mouse models. The exact mechanism underlying acute respiratory distress syndrome (ARDS) and other cardiopulmonary complications in elderly patients in combination with associated comorbidities remain unclear. Nevertheless, understanding the critical role of sleep, circadian clock dysfunction in target organs, and immune status of patients with SARS-CoV-2 may provide novel insights into possible therapies. Chronotherapy is an emerging concept that is gaining attention in sleep medicine. Accumulating evidence suggests that nearly half of all physiological functions follow a strict daily rhythm. However, healthcare professionals rarely take implementing timed-administration of drugs into consideration. In this review, we summarize recent findings directly relating to the contributing roles of sleep, circadian rhythms and immune response in modulating infectious disease processes, and integrate chronotherapy in the discussion of the potential drugs that can be repurposed to improve the treatment and management of COVID-19.
Collapse
Affiliation(s)
| | | | - Isaac Kirubakaran Sundar
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
20
|
Wan W, Zhu S, Li S, Shang W, Zhang R, Li H, Liu W, Xiao G, Peng K, Zhang L. High-Throughput Screening of an FDA-Approved Drug Library Identifies Inhibitors against Arenaviruses and SARS-CoV-2. ACS Infect Dis 2021; 7:1409-1422. [PMID: 33183004 PMCID: PMC7671101 DOI: 10.1021/acsinfecdis.0c00486] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Indexed: 02/07/2023]
Abstract
Arenaviruses are a large family of enveloped negative-strand RNA viruses that include several causative agents of severe hemorrhagic fevers. Currently, there are no FDA-licensed drugs to treat arenavirus infection except for the off-labeled use of ribavirin. Here, we performed antiviral drug screening against the Old World arenavirus lymphocytic choriomeningitis virus (LCMV) using an FDA-approved drug library. Five drug candidates were identified, including mycophenolic acid, benidipine hydrochloride, clofazimine, dabrafenib, and apatinib, for having strong anti-LCMV effects. Further analysis indicated that benidipine hydrochloride inhibited LCMV membrane fusion, and an adaptive mutation on the LCMV glycoprotein D414 site was found to antagonize the anti-LCMV activity of benidipine hydrochloride. Mycophenolic acid inhibited LCMV replication by depleting GTP production. We also found mycophenolic acid, clofazimine, dabrafenib, and apatinib can inhibit the newly emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Owing to their FDA-approved status, these drug candidates can potentially be used rapidly in the clinical treatment of arenavirus and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Weiwei Wan
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of
Sciences, Wuhan, Hubei 430071, PR
China
- University of Chinese
Academy of Sciences, Beijing 100049, PR
China
| | - Shenglin Zhu
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of
Sciences, Wuhan, Hubei 430071, PR
China
| | - Shufen Li
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of
Sciences, Wuhan, Hubei 430071, PR
China
| | - Weijuan Shang
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of
Sciences, Wuhan, Hubei 430071, PR
China
| | - Ruxue Zhang
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of
Sciences, Wuhan, Hubei 430071, PR
China
| | - Hao Li
- Beijing Institute of
Microbiology and Epidemiology, State Key
Laboratory of Pathogen and Biosecurity, Beijing 100071, PR
China
| | - Wei Liu
- Beijing Institute of
Microbiology and Epidemiology, State Key
Laboratory of Pathogen and Biosecurity, Beijing 100071, PR
China
| | - Gengfu Xiao
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of
Sciences, Wuhan, Hubei 430071, PR
China
- University of Chinese
Academy of Sciences, Beijing 100049, PR
China
| | - Ke Peng
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of
Sciences, Wuhan, Hubei 430071, PR
China
- University of Chinese
Academy of Sciences, Beijing 100049, PR
China
| | - Leike Zhang
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of
Sciences, Wuhan, Hubei 430071, PR
China
- University of Chinese
Academy of Sciences, Beijing 100049, PR
China
| |
Collapse
|
21
|
Eh-Haj BM. Metabolic N-Dealkylation and N-Oxidation as Elucidators of the Role of Alkylamino Moieties in Drugs Acting at Various Receptors. Molecules 2021; 26:1917. [PMID: 33805491 PMCID: PMC8036657 DOI: 10.3390/molecules26071917] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 12/16/2022] Open
Abstract
Metabolic reactions that occur at alkylamino moieties may provide insight into the roles of these moieties when they are parts of drug molecules that act at different receptors. N-dealkylation of N,N-dialkylamino moieties has been associated with retaining, attenuation or loss of pharmacologic activities of metabolites compared to their parent drugs. Further, N-dealkylation has resulted in clinically used drugs, activation of prodrugs, change of receptor selectivity, and providing potential for developing fully-fledged drugs. While both secondary and tertiary alkylamino moieties (open chain aliphatic or heterocyclic) are metabolized by CYP450 isozymes oxidative N-dealkylation, only tertiary alkylamino moieties are subject to metabolic N-oxidation by Flavin-containing monooxygenase (FMO) to give N-oxide products. In this review, two aspects will be examined after surveying the metabolism of representative alkylamino-moieties-containing drugs that act at various receptors (i) the pharmacologic activities and relevant physicochemical properties (basicity and polarity) of the metabolites with respect to their parent drugs and (ii) the role of alkylamino moieties on the molecular docking of drugs in receptors. Such information is illuminative in structure-based drug design considering that fully-fledged metabolite drugs and metabolite prodrugs have been, respectively, developed from N-desalkyl and N-oxide metabolites.
Collapse
Affiliation(s)
- Babiker M Eh-Haj
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, University of Science and Technology of Fujairah, Emirate of Fujairah, Fujairah 2022, United Arab Emirates
| |
Collapse
|
22
|
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
|
23
|
Direct Intracellular Visualization of Ebola Virus-Receptor Interaction by In Situ Proximity Ligation. mBio 2021; 12:mBio.03100-20. [PMID: 33436438 PMCID: PMC7844541 DOI: 10.1128/mbio.03100-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Ebola virus causes episodic but increasingly frequent outbreaks of severe disease in Middle Africa, as shown by the recently overcome second largest outbreak on record in the Democratic Republic of Congo. Despite considerable effort, FDA-approved antifiloviral therapeutics or targeted interventions are not available yet. Ebola virus (EBOV) entry into host cells comprises stepwise and extensive interactions of the sole viral surface glycoprotein (GP) with multiple host factors. During the intricate process, following virus uptake and trafficking to late endosomal/lysosomal compartments, GP is proteolytically processed to cleaved GP (GPCL) by the endosomal proteases cathepsin B and L, unmasking GP’s receptor-binding site. Engagement of GPCL with the universal filoviral intracellular receptor Niemann-Pick C1 (NPC1) eventually culminates in fusion between viral and cellular membranes, cytoplasmic escape of the viral nucleocapsid, and subsequent infection. Mechanistic delineation of the indispensable GPCL-NPC1-binding step has been severely hampered by the unavailability of a robust cell-based assay assessing interaction of GPCL with full-length endosomal NPC1. Here, we describe a novel in situ assay to monitor GPCL-NPC1 engagement in intact, infected cells. Visualization of the subcellular localization of binding complexes is based on the principle of DNA-assisted, antibody-mediated proximity ligation. Virus-receptor binding monitored by proximity ligation was contingent on GP’s proteolytic cleavage and was sensitive to perturbations in the GPCL-NPC1 interface. Our assay also specifically decoupled detection of virus-receptor binding from steps post-receptor binding, such as membrane fusion and infection. Testing of multiple FDA-approved small-molecule inhibitors revealed that drug treatments inhibited virus entry and GPCL-NPC1 recognition by distinctive mechanisms. Together, here we present a newly established proximity ligation assay, which will allow us to dissect cellular and viral requirements for filovirus-receptor binding and to delineate the mechanisms of action of inhibitors on filovirus entry in a cell-based system.
Collapse
|
24
|
Krishnamoorthy P, Raj AS, Roy S, Kumar NS, Kumar H. Comparative transcriptome analysis of SARS-CoV, MERS-CoV, and SARS-CoV-2 to identify potential pathways for drug repurposing. Comput Biol Med 2021; 128:104123. [PMID: 33260034 PMCID: PMC7683955 DOI: 10.1016/j.compbiomed.2020.104123] [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: 07/22/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022]
Abstract
The ongoing COVID-19 pandemic caused by the coronavirus, SARS-CoV-2, has already caused in excess of 1.25 million deaths worldwide, and the number is increasing. Knowledge of the host transcriptional response against this virus and how the pathways are activated or suppressed compared to other human coronaviruses (SARS-CoV, MERS-CoV) that caused outbreaks previously can help in the identification of potential drugs for the treatment of COVID-19. Hence, we used time point meta-analysis to investigate available SARS-CoV and MERS-CoV in-vitro transcriptome datasets in order to identify the significant genes and pathways that are dysregulated at each time point. The subsequent over-representation analysis (ORA) revealed that several pathways are significantly dysregulated at each time point after both SARS-CoV and MERS-CoV infection. We also performed gene set enrichment analyses of SARS-CoV and MERS-CoV with that of SARS-CoV-2 at the same time point and cell line, the results of which revealed that common pathways are activated and suppressed in all three coronaviruses. Furthermore, an analysis of an in-vivo transcriptomic dataset of COVID-19 patients showed that similar pathways are enriched to those identified in the earlier analyses. Based on these findings, a drug repurposing analysis was performed to identify potential drug candidates for combating COVID-19.
Collapse
Affiliation(s)
- Pandikannan Krishnamoorthy
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, 462066, MP, India
| | - Athira S Raj
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, 462066, MP, India
| | - Swagnik Roy
- Microbiology Department, Zoram Medical College, Falkawn, Mizoram, 796005, India
| | | | - Himanshu Kumar
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, 462066, MP, India; Laboratory of Host Defense, WPI Immunology, Frontier Research Centre, Osaka University, Osaka, 5650871, Japan.
| |
Collapse
|
25
|
Bagheri A, Moezzi SMI, Mosaddeghi P, Nadimi Parashkouhi S, Fazel Hoseini SM, Badakhshan F, Negahdaripour M. Interferon-inducer antivirals: Potential candidates to combat COVID-19. Int Immunopharmacol 2020; 91:107245. [PMID: 33348292 PMCID: PMC7705326 DOI: 10.1016/j.intimp.2020.107245] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/19/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is an infective disease generated by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Given the pandemic urgency and lack of an effective cure for this disease, drug repurposing could open the way for finding a solution. Lots of investigations are ongoing to test the compounds already identified as antivirals. On the other hand, induction of type I interferons are found to play an important role in the generation of immune responses against SARS-CoV-2. Therefore, it was opined that the antivirals capable of triggering the interferons and their signaling pathway, could rationally be beneficial for treating COVID-19. On this basis, using a database of antivirals, called drugvirus, some antiviral agents were derived, followed by searches on their relevance to interferon induction. The examined list included drugs from different categories such as antibiotics, immunosuppressants, anti-cancers, non-steroidal anti-inflammatory drugs (NSAID), calcium channel blocker compounds, and some others. The results as briefed here, could help in finding potential drug candidates for COVID-19 treatment. However, their advantages and risks should be taken into account through precise studies, considering a systemic approach. Even though the adverse effects of some of these drugs may overweight their benefits, considering their mechanisms and structures may give a clue for designing novel drugs in the future. Furthermore, the antiviral effect and IFN-modifying mechanisms possessed by some of these drugs might lead to a synergistic effect against SARS-CoV-2, which deserve to be evaluated in further investigations.
Collapse
Affiliation(s)
- Ashkan Bagheri
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Iman Moezzi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pouria Mosaddeghi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sadra Nadimi Parashkouhi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mostafa Fazel Hoseini
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Badakhshan
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
26
|
Crespi B, Alcock J. Conflicts over calcium and the treatment of COVID-19. EVOLUTION MEDICINE AND PUBLIC HEALTH 2020; 9:149-156. [PMID: 33732462 PMCID: PMC7717197 DOI: 10.1093/emph/eoaa046] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022]
Abstract
Several recent studies have provided evidence that use of calcium channel blockers (CCBs), especially amlodipine and nifedipine, can reduce mortality from coronavirus disease 2019 (COVID-19). Moreover, hypocalcemia (a reduced level of serum ionized calcium) has been shown to be strongly positively associated with COVID-19 severity. Both effectiveness of CCBs as antiviral therapy, and positive associations of hypocalcemia with mortality, have been demonstrated for many other viruses as well. We evaluate these findings in the contexts of virus–host evolutionary conflicts over calcium metabolism, and hypocalcemia as either pathology, viral manipulation or host defence against pathogens. Considerable evidence supports the hypothesis that hypocalcemia represents a host defence. Indeed, hypocalcemia may exert antiviral effects in a similar manner as do CCBs, through interference with calcium metabolism in virus-infected cells. Prospective clinical studies that address the efficacy of CCBs and hypocalcemia should provide novel insights into the pathogenicity and treatment of COVID-19 and other viruses.
Collapse
Affiliation(s)
- Bernard Crespi
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Joe Alcock
- Department of Emergency Medicine, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
27
|
Vela JM. Repurposing Sigma-1 Receptor Ligands for COVID-19 Therapy? Front Pharmacol 2020; 11:582310. [PMID: 33364957 PMCID: PMC7751758 DOI: 10.3389/fphar.2020.582310] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/30/2020] [Indexed: 12/27/2022] Open
Abstract
Outbreaks of emerging infections, such as COVID-19 pandemic especially, confront health professionals with the unique challenge of treating patients. With no time to discover new drugs, repurposing of approved drugs or in clinical development is likely the only solution. Replication of coronaviruses (CoVs) occurs in a modified membranous compartment derived from the endoplasmic reticulum (ER), causes host cell ER stress and activates pathways to facilitate adaptation of the host cell machinery to viral needs. Accordingly, modulation of ER remodeling and ER stress response might be pivotal in elucidating CoV-host interactions and provide a rationale for new therapeutic, host-based antiviral approaches. The sigma-1 receptor (Sig-1R) is a ligand-operated, ER membrane-bound chaperone that acts as an upstream modulator of ER stress and thus a candidate host protein for host-based repurposing approaches to treat COVID-19 patients. Sig-1R ligands are frequently identified in in vitro drug repurposing screens aiming to identify antiviral compounds against CoVs, including severe acute respiratory syndrome CoV-2 (SARS-CoV-2). Sig-1R regulates key mechanisms of the adaptive host cell stress response and takes part in early steps of viral replication. It is enriched in lipid rafts and detergent-resistant ER membranes, where it colocalizes with viral replicase proteins. Indeed, the non-structural SARS-CoV-2 protein Nsp6 interacts with Sig-1R. The activity of Sig-1R ligands against COVID-19 remains to be specifically assessed in clinical trials. This review provides a rationale for targeting Sig-1R as a host-based drug repurposing approach to treat COVID-19 patients. Evidence gained using Sig-1R ligands in unbiased in vitro antiviral drug screens and the potential mechanisms underlying the modulatory effect of Sig-1R on the host cell response are discussed. Targeting Sig-1R is not expected to reduce dramatically established viral replication, but it might interfere with early steps of virus-induced host cell reprogramming, aid to slow down the course of infection, prevent the aggravation of the disease and/or allow a time window to mature a protective immune response. Sig-1R-based medicines could provide benefit not only as early intervention, preventive but also as adjuvant therapy.
Collapse
Affiliation(s)
- José Miguel Vela
- Drug Discovery and Preclinical Development, ESTEVE Pharmaceuticals, Barcelona, Spain
| |
Collapse
|
28
|
Lo MK, Albariño CG, Perry JK, Chang S, Tchesnokov EP, Guerrero L, Chakrabarti A, Shrivastava-Ranjan P, Chatterjee P, McMullan LK, Martin R, Jordan R, Götte M, Montgomery JM, Nichol ST, Flint M, Porter D, Spiropoulou CF. Remdesivir targets a structurally analogous region of the Ebola virus and SARS-CoV-2 polymerases. Proc Natl Acad Sci U S A 2020; 117:26946-26954. [PMID: 33028676 PMCID: PMC7604432 DOI: 10.1073/pnas.2012294117] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Remdesivir is a broad-spectrum antiviral nucleotide prodrug that has been clinically evaluated in Ebola virus patients and recently received emergency use authorization (EUA) for treatment of COVID-19. With approvals from the Federal Select Agent Program and the Centers for Disease Control and Prevention's Institutional Biosecurity Board, we characterized the resistance profile of remdesivir by serially passaging Ebola virus under remdesivir selection; we generated lineages with low-level reduced susceptibility to remdesivir after 35 passages. We found that a single amino acid substitution, F548S, in the Ebola virus polymerase conferred low-level reduced susceptibility to remdesivir. The F548 residue is highly conserved in filoviruses but should be subject to specific surveillance among novel filoviruses, in newly emerging variants in ongoing outbreaks, and also in Ebola virus patients undergoing remdesivir therapy. Homology modeling suggests that the Ebola virus polymerase F548 residue lies in the F-motif of the polymerase active site, a region that was previously identified as susceptible to resistance mutations in coronaviruses. Our data suggest that molecular surveillance of this region of the polymerase in remdesivir-treated COVID-19 patients is also warranted.
Collapse
Affiliation(s)
- Michael K Lo
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329;
| | - César G Albariño
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329
| | | | | | - Egor P Tchesnokov
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Lisa Guerrero
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329
| | - Ayan Chakrabarti
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329
| | - Punya Shrivastava-Ranjan
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329
| | - Payel Chatterjee
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329
| | - Laura K McMullan
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329
| | | | | | - Matthias Götte
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Joel M Montgomery
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329
| | - Stuart T Nichol
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329
| | - Mike Flint
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329
| | | | - Christina F Spiropoulou
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA 30329;
| |
Collapse
|
29
|
AbdelMassih AF, Ye J, Kamel A, Mishriky F, Ismail HA, Ragab HA, El Qadi L, Malak L, Abdu M, El-Husseiny M, Ashraf M, Hafez N, AlShehry N, El-Husseiny N, AbdelRaouf N, Shebl N, Hafez N, Youssef N, Afdal P, Hozaien R, Menshawey R, Saeed R, Fouda R. A multicenter consensus: A role of furin in the endothelial tropism in obese patients with COVID-19 infection. OBESITY MEDICINE 2020; 19:100281. [PMID: 32835124 PMCID: PMC7362855 DOI: 10.1016/j.obmed.2020.100281] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023]
Abstract
Furin, a cleavage enzyme, is increasingly recognized in the pathogenesis of metabolic syndrome. Its cleavage action is an essential activation step for the endothelial pathogenicity of several viruses including SARS-CoV-2. This Furin-mediated endothelial tropism seems to underlie the multi-organ system involvement of COVID-19; which is a feature that was not recognized in the older versions of coronaviridae. Obese and diabetic patients, males, and the elderly, have increased serum levels of Furin, with its increased cellular activity; this might explain why these subgroups are at an increased risk of COVID-19 related complications and deaths. In contrast, smoking decreases cellular levels of Furin, this finding may be at the origin of the decreased severity of COVID-19 in smokers. Chinese herbal derived luteolin is suggested to be putative Furin inhibitor, with previous success against Dengue Fever. Additionally, Furin intracellular levels are largely dependent on concentration of intracellular ions, notably sodium, potassium, and magnesium. Consequently, the use of ion channel inhibitors, such as Calcium Channel blockers or Potassium Channel blockers, can prevent cellular transfection early in the course of the illness. Nicotine patches and Colchicine have also been suggested as potential therapies due to Furin mediated inhibition of COVID-19.
Collapse
Affiliation(s)
- Antoine Fakhry AbdelMassih
- Pediatric Cardiology Unit, Pediatrics' Department, Faculty of Medicine, Cairo University, Egypt
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jianping Ye
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Aya Kamel
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Fady Mishriky
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Habiba-Allah Ismail
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Heba Amin Ragab
- Pediatric Cardiology Unit, Pediatrics' Department, Faculty of Medicine, Cairo University, Egypt
| | - Layla El Qadi
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Lauris Malak
- Pediatric Cardiology Unit, Pediatrics' Department, Faculty of Medicine, Cairo University, Egypt
| | - Mariam Abdu
- Pediatric Cardiology Unit, Pediatrics' Department, Faculty of Medicine, Cairo University, Egypt
| | - Miral El-Husseiny
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Mirette Ashraf
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Nada Hafez
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Nada AlShehry
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Nadine El-Husseiny
- Faculty of Dentistry, Cairo University, Egypt
- Pixagon Graphic Design Agency, Cairo, Egypt
| | - Nora AbdelRaouf
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Noura Shebl
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Nouran Hafez
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Nourhan Youssef
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Peter Afdal
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Rafeef Hozaien
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Rahma Menshawey
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Rana Saeed
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Egypt
| | - Raghda Fouda
- University of Irvine California, USA
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Pediatrics' Department, Faculty of Medicine, Cairo University, Egypt
| |
Collapse
|
30
|
A Virion-Based Assay for Glycoprotein Thermostability Reveals Key Determinants of Filovirus Entry and Its Inhibition. J Virol 2020; 94:JVI.00336-20. [PMID: 32611759 DOI: 10.1128/jvi.00336-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/26/2020] [Indexed: 11/20/2022] Open
Abstract
Ebola virus (EBOV) entry into cells is mediated by its spike glycoprotein (GP). Following attachment and internalization, virions traffic to late endosomes where GP is cleaved by host cysteine proteases. Cleaved GP then binds its cellular receptor, Niemann-Pick C1. In response to an unknown cellular trigger, GP undergoes conformational rearrangements that drive fusion of viral and endosomal membranes. The temperature-dependent stability (thermostability) of the prefusion conformers of class I viral fusion glycoproteins, including those of filovirus GPs, has provided insights into their propensity to undergo fusion-related rearrangements. However, previously described assays have relied on soluble glycoprotein ectodomains. Here, we developed a simple enzyme-linked immunosorbent assay (ELISA)-based assay that uses the temperature-dependent loss of conformational epitopes to measure thermostability of GP embedded in viral membranes. The base and glycan cap subdomains of all filovirus GPs tested suffered a concerted loss of prefusion conformation at elevated temperatures but did so at different temperature ranges, indicating virus-specific differences in thermostability. Despite these differences, all of these GPs displayed reduced thermostability upon cleavage to GP conformers (GPCL). Surprisingly, acid pH enhanced, rather than decreased, GP thermostability, suggesting it could enhance viral survival in hostile endo/lysosomal compartments. Finally, we confirmed and extended previous findings that some small-molecule inhibitors of filovirus entry destabilize EBOV GP and uncovered evidence that the most potent inhibitors act through multiple mechanisms. We establish the epitope-loss ELISA as a useful tool for studies of filovirus entry, engineering of GP variants with enhanced stability for use in vaccine development, and discovery of new stability-modulating antivirals.IMPORTANCE The development of Ebola virus countermeasures is challenged by our limited understanding of cell entry, especially at the step of membrane fusion. The surface-exposed viral protein, GP, mediates membrane fusion and undergoes major structural rearrangements during this process. The stability of GP at elevated temperatures (thermostability) can provide insights into its capacity to undergo these rearrangements. Here, we describe a new assay that uses GP-specific antibodies to measure GP thermostability under a variety of conditions relevant to viral entry. We show that proteolytic cleavage and acid pH have significant effects on GP thermostability that shed light on their respective roles in viral entry. We also show that the assay can be used to study how small-molecule entry inhibitors affect GP stability. This work provides a simple and readily accessible assay to engineer stabilized GP variants for antiviral vaccines and to discover and improve drugs that act by modulating GP stability.
Collapse
|
31
|
Chen L, Chen H, Dong S, Huang W, Chen L, Wei Y, Shi L, Li J, Zhu F, Zhu Z, Wang Y, Lv X, Yu X, Li H, Wei W, Zhang K, Zhu L, Qu C, Hong J, Hu C, Dong J, Qi R, Lu D, Wang H, Peng S, Hao G. The Effects of Chloroquine and Hydroxychloroquine on ACE2-Related Coronavirus Pathology and the Cardiovascular System: An Evidence-Based Review. FUNCTION 2020; 1:zqaa012. [PMID: 38626250 PMCID: PMC7454642 DOI: 10.1093/function/zqaa012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 01/08/2023] Open
Abstract
The ongoing pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a serious threat to global public health and there is currently no effective antiviral therapy. It has been suggested that chloroquine (CQ) and hydroxychloroquine (HCQ), which were primarily employed as prophylaxis and treatment for malaria, could be used to treat COVID-19. CQ and HCQ may be potential inhibitors of SARS-CoV-2 entry into host cells, which are mediated via the angiotensin-converting enzyme 2 (ACE2), and may also inhibit subsequent intracellular processes which lead to COVID-19, including damage to the cardiovascular (CV) system. However, paradoxically, CQ and HCQ have also been reported to cause damage to the CV system. In this review, we provide a critical examination of the published evidence. CQ and HCQ could potentially be useful drugs in the treatment of COVID-19 and other ACE2 involved virus infections, but the antiviral effects of CQ and HCQ need to be tested in more well-designed clinical randomized studies and their actions on the CV system need to be further elucidated. However, even if it were to turn out that CQ and HCQ are not useful drugs in practice, further studies of their mechanism of action could be helpful in improving our understanding of COVID-19 pathology.
Collapse
Affiliation(s)
- Li Chen
- Department of Medicine, Georgia Prevention Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Haiyan Chen
- Department of Endemic Disease, Guangzhou Center for Disease Control and Prevention, Guangzhou 510440, China
| | - Shan Dong
- Guangzhou First People’s Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou 510180, China
| | - Wei Huang
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Li Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yuan Wei
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sports University, Guangzhou 510500, China
| | - Liping Shi
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jinying Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Fengfeng Zhu
- Department of Hepatobiliary and Pancreas Surgery, The First Affiliated Hospital Of University of South China, Hengyang 421001, China
| | - Zhu Zhu
- Department of Hepatobiliary and Pancreas Surgery, The First Affiliated Hospital Of University of South China, Hengyang 421001, China
| | - Yiyang Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiuxiu Lv
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiaohui Yu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Hongmei Li
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Wei Wei
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Keke Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Lihong Zhu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Chen Qu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jian Hong
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Chaofeng Hu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jun Dong
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Renbin Qi
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Daxiang Lu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People’s Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Huadong Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People’s Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Shuang Peng
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Guang Hao
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
32
|
Al-Horani RA, Kar S, Aliter KF. Potential Anti-COVID-19 Therapeutics that Block the Early Stage of the Viral Life Cycle: Structures, Mechanisms, and Clinical Trials. Int J Mol Sci 2020; 21:E5224. [PMID: 32718020 PMCID: PMC7432953 DOI: 10.3390/ijms21155224] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
The ongoing pandemic of coronavirus disease-2019 (COVID-19) is being caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The disease continues to present significant challenges to the health care systems around the world. This is primarily because of the lack of vaccines to protect against the infection and the lack of highly effective therapeutics to prevent and/or treat the illness. Nevertheless, researchers have swiftly responded to the pandemic by advancing old and new potential therapeutics into clinical trials. In this review, we summarize potential anti-COVID-19 therapeutics that block the early stage of the viral life cycle. The review presents the structures, mechanisms, and reported results of clinical trials of potential therapeutics that have been listed in clinicaltrials.gov. Given the fact that some of these therapeutics are multi-acting molecules, other relevant mechanisms will also be described. The reviewed therapeutics include small molecules and macromolecules of sulfated polysaccharides, polypeptides, and monoclonal antibodies. The potential therapeutics target viral and/or host proteins or processes that facilitate the early stage of the viral infection. Frequent targets are the viral spike protein, the host angiotensin converting enzyme 2, the host transmembrane protease serine 2, and clathrin-mediated endocytosis process. Overall, the review aims at presenting update-to-date details, so as to enhance awareness of potential therapeutics, and thus, to catalyze their appropriate use in combating the pandemic.
Collapse
Affiliation(s)
- Rami A. Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| | - Srabani Kar
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| | - Kholoud F. Aliter
- Department of Chemistry, School of STEM, Dillard University, New Orleans, LA 70122, USA;
| |
Collapse
|
33
|
Gunesch AP, Zapatero-Belinchón FJ, Pinkert L, Steinmann E, Manns MP, Schneider G, Pietschmann T, Brönstrup M, von Hahn T. Filovirus Antiviral Activity of Cationic Amphiphilic Drugs Is Associated with Lipophilicity and Ability To Induce Phospholipidosis. Antimicrob Agents Chemother 2020; 64:e00143-20. [PMID: 32513799 PMCID: PMC7526846 DOI: 10.1128/aac.00143-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023] Open
Abstract
Several cationic amphiphilic drugs (CADs) have been found to inhibit cell entry of filoviruses and other enveloped viruses. Structurally unrelated CADs may have antiviral activity, yet the underlying common mechanism and structure-activity relationship are incompletely understood. We aimed to understand how widespread antiviral activity is among CADs and which structural and physico-chemical properties are linked to entry inhibition. We measured inhibition of Marburg virus pseudoparticle (MARVpp) cell entry by 45 heterogeneous and mostly FDA-approved CADs and cytotoxicity in EA.hy926 cells. We analyzed correlation of antiviral activity with four chemical properties: pKa, hydrophobicity (octanol/water partitioning coefficient; ClogP), molecular weight, and distance between the basic group and hydrophobic ring structures. Additionally, we quantified drug-induced phospholipidosis (DIPL) of a CAD subset by flow cytometry. Structurally similar compounds (derivatives) and those with similar chemical properties but unrelated structures (analogues) to those of strong inhibitors were obtained by two in silico similarity search approaches and tested for antiviral activity. Overall, 11 out of 45 (24%) CADs inhibited MARVpp by 40% or more. The strongest antiviral compounds were dronedarone, triparanol, and quinacrine. Structure-activity relationship studies revealed highly significant correlations between antiviral activity, hydrophobicity (ClogP > 4), and DIPL. Moreover, pKa and intramolecular distance between hydrophobic and hydrophilic moieties correlated with antiviral activity but to a lesser extent. We also showed that in contrast to analogues, derivatives had antiviral activity similar to that of the seed compound dronedarone. Overall, one-quarter of CADs inhibit MARVpp entry in vitro, and antiviral activity of CADs mostly relies on their hydrophobicity yet is promoted by the individual structure.
Collapse
Affiliation(s)
- Antonia P Gunesch
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover-Braunschweig Site, Braunschweig, Germany
- Institute of Experimental Virology, TWINCORE, Center for Experimental and Clinical Infection Research Hannover, Hannover, Germany
| | - Francisco J Zapatero-Belinchón
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover-Braunschweig Site, Braunschweig, Germany
- Institute of Experimental Virology, TWINCORE, Center for Experimental and Clinical Infection Research Hannover, Hannover, Germany
| | - Lukas Pinkert
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Eike Steinmann
- Department for Molecular and Medical Virology, Ruhr Universität Bochum, Bochum, Germany
| | - Michael P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover-Braunschweig Site, Braunschweig, Germany
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Eidgenössische Technische Hochschule, Zurich, Switzerland
| | - Thomas Pietschmann
- German Center for Infection Research, Hannover-Braunschweig Site, Braunschweig, Germany
- Institute of Experimental Virology, TWINCORE, Center for Experimental and Clinical Infection Research Hannover, Hannover, Germany
| | - Mark Brönstrup
- German Center for Infection Research, Hannover-Braunschweig Site, Braunschweig, Germany
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas von Hahn
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover-Braunschweig Site, Braunschweig, Germany
- Institute of Experimental Virology, TWINCORE, Center for Experimental and Clinical Infection Research Hannover, Hannover, Germany
- Department of Gastroenterology and Interventional Endoscopy, Asklepios Hospital Barmbek, Semmelweis University, Hamburg, Germany
| |
Collapse
|
34
|
Dash RN, Moharana AK, Subudhi BB. Sulfonamides: Antiviral Strategy for Neglected Tropical Disease Virus. CURR ORG CHEM 2020. [DOI: 10.2174/1385272824999200515094100] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The viral infections are a threat to the health system around the globe. Although
more than 60 antiviral drugs have been approved by the FDA, most of them are for the
management of few viruses like HIV, Hepatitis and Influenza. There is no antiviral for
many viruses including Dengue, Chikungunya and Japanese encephalitis. Many of these
neglected viruses are increasingly becoming global pathogens. Lack of broad spectrum of
action and the rapid rise of resistance and cross-resistance to existing antiviral have further
increased the challenge of antiviral development. Sulfonamide, as a privileged scaffold,
has been capitalized to develop several bioactive compounds and drugs. Accordingly, several
reviews have been published in recent times on bioactive sulfonamides. However,
there are not enough review reports of antiviral sulfonamides in the last five years. Sulfonamides
scaffolds have received sufficient attention for the development of non- nucleoside antivirals following
the emergence of cross-resistance to nucleoside inhibitors. Hybridization of bioactive pharmacophores
with sulfonamides has been used as a strategy to develop sulfonamide antivirals. This review is an effort to
analyze these attempts and evaluate their translational potential. Parameters including potency (IC50), toxicity
(CC50) and selectivity (CC50/IC50) have been used in this report to suggest the potential of sulfonamide derivatives
to progress further as antiviral. Since most of these antiviral properties are based on the in vitro results,
the drug-likeness of molecules has been predicted to propose in vivo potential. The structure-activity relationship
has been analyzed to encourage further optimization of antiviral properties.
Collapse
Affiliation(s)
- Rudra Narayan Dash
- Drug Development and Analysis Laboratory, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar-751029, Odisha, India
| | - Alok Kumar Moharana
- Drug Development and Analysis Laboratory, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar-751029, Odisha, India
| | - Bharat Bhusan Subudhi
- Drug Development and Analysis Laboratory, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar-751029, Odisha, India
| |
Collapse
|
35
|
Navarese EP, Musci RL, Frediani L, Gurbel PA, Kubica J. Ion channel inhibition against COVID-19: A novel target for clinical investigation. Cardiol J 2020; 27:421-424. [PMID: 32643141 DOI: 10.5603/cj.a2020.0090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Affiliation(s)
- Eliano P Navarese
- Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Rita L Musci
- Department of Cardiology, Azienda Ospedaliera Bonomo, Andria, Italy, Andria, Italy
| | - Lara Frediani
- Department of Cardiology, Azienda Usl Toscana Nord-Ovest Cardiologia UTIC-Ospedali Riuniti di Livorno, Italy, Livorno, Italy
| | - Paul A Gurbel
- Sinai Center for Cardiovascular Research, Sinai Hospital of Baltimore, LifeBridgehealth, Baltimore, MD, USA., Baltimore, United States
| | - Jacek Kubica
- Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland.
| |
Collapse
|
36
|
Ittzes B, Szentkiralyi E, Szabo Z, Batai IZ, Gyorffy O, Kovacs T, Batai I, Kerenyi M. Amiodarone that has antibacterial effect against human pathogens may represent a novel catheter lock. Acta Microbiol Immunol Hung 2020; 67:133-137. [PMID: 32634110 DOI: 10.1556/030.2020.01144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/09/2020] [Indexed: 11/19/2022]
Abstract
Infection is one of the most feared hospital-acquired complications. Infusion therapy is frequently administered through a central line. Infusions facilitating bacterial growth may be a source of central line-associated bloodstream infections. On the other hand, medications that kill bacteria may protect against this kind of infection and may be used as a catheter lock.In this study, we examined the impact of amiodarone on bacterial growth. Amiodarone is used for controlling cardiac arrhythmias and can be administered as an infusion for weeks. Standard microbiological methods have been used to study the growth of laboratory strains and clinical isolates of Staphylococcus aureus, Staphylococcus epidermidis, Pseudomonas aeruginosa, Escherichia coli, Klebsiella pneumoniae, and multidrug-resistant Acinetobacter baumannii in amiodarone. The minimum inhibitory concentration (MIC) of amiodarone was determined. Bacterial growth from in use amiodarone syringes and giving sets was also investigated.Most examined strains were killed within 1 min in amiodarone. The other strains were killed within 1 h. The MICs of amiodarone were <0.5-32 μg/mL.Amiodarone infusion is unlikely to be responsible for bloodstream infections as contaminating bacteria are killed within 1 h. Amiodarone may also protect against central line infections if used as a catheter lock.
Collapse
Affiliation(s)
- Balazs Ittzes
- 1Department of Medical Microbiology, Medical School, University of Pecs, Pecs, Hungary.,5Department of Anaesthesia and Intensive Therapy, North Devon District Hospital, Barnstaple, Devon, UK
| | - Eva Szentkiralyi
- 1Department of Medical Microbiology, Medical School, University of Pecs, Pecs, Hungary
| | - Zoltan Szabo
- 2Department of Anaesthesiology and Intensive Therapy, Medical School, University of Pecs, Pecs, Hungary
| | - Istvan Z Batai
- 3Department of Pharmacology and Pharmacotherapy, Medical School, University of Pecs, Pecs, Hungary
| | - Ors Gyorffy
- 1Department of Medical Microbiology, Medical School, University of Pecs, Pecs, Hungary.,4Department of Anaesthesiology and Intensive Therapy, St. Rafael Hospital, Zalaegerszeg, Hungary
| | - Tamas Kovacs
- 2Department of Anaesthesiology and Intensive Therapy, Medical School, University of Pecs, Pecs, Hungary.,4Department of Anaesthesiology and Intensive Therapy, St. Rafael Hospital, Zalaegerszeg, Hungary
| | - Istvan Batai
- 2Department of Anaesthesiology and Intensive Therapy, Medical School, University of Pecs, Pecs, Hungary
| | - Monika Kerenyi
- 1Department of Medical Microbiology, Medical School, University of Pecs, Pecs, Hungary
| |
Collapse
|
37
|
Sturley SL, Rajakumar T, Hammond N, Higaki K, Márka Z, Márka S, Munkacsi AB. Potential COVID-19 therapeutics from a rare disease: weaponizing lipid dysregulation to combat viral infectivity. J Lipid Res 2020; 61:972-982. [PMID: 32457038 PMCID: PMC7328045 DOI: 10.1194/jlr.r120000851] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/21/2020] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus (SARS-CoV)-2 has resulted in the death of more than 328,000 persons worldwide in the first 5 months of 2020. Herculean efforts to rapidly design and produce vaccines and other antiviral interventions are ongoing. However, newly evolving viral mutations, the prospect of only temporary immunity, and a long path to regulatory approval pose significant challenges and call for a common, readily available, and inexpensive treatment. Strategic drug repurposing combined with rapid testing of established molecular targets could provide a pause in disease progression. SARS-CoV-2 shares extensive structural and functional conservation with SARS-CoV-1, including engagement of the same host cell receptor (angiotensin-converting enzyme 2) localized in cholesterol-rich microdomains. These lipid-enveloped viruses encounter the endosomal/lysosomal host compartment in a critical step of infection and maturation. Niemann-Pick type C (NP-C) disease is a rare monogenic neurodegenerative disease caused by deficient efflux of lipids from the late endosome/lysosome (LE/L). The NP-C disease-causing gene (NPC1) has been strongly associated with viral infection, both as a filovirus receptor (e.g., Ebola) and through LE/L lipid trafficking. This suggests that NPC1 inhibitors or NP-C disease mimetics could serve as anti-SARS-CoV-2 agents. Fortunately, there are such clinically approved molecules that elicit antiviral activity in preclinical studies, without causing NP-C disease. Inhibition of NPC1 may impair viral SARS-CoV-2 infectivity via several lipid-dependent mechanisms, which disturb the microenvironment optimum for viral infectivity. We suggest that known mechanistic information on NPC1 could be utilized to identify existing and future drugs to treat COVID-19.
Collapse
MESH Headings
- Androstenes/therapeutic use
- Angiotensin-Converting Enzyme 2
- Anticholesteremic Agents/therapeutic use
- Antiviral Agents/therapeutic use
- Betacoronavirus/drug effects
- Betacoronavirus/metabolism
- Betacoronavirus/pathogenicity
- COVID-19
- Cholesterol/metabolism
- Coronavirus Infections/diagnosis
- Coronavirus Infections/drug therapy
- Coronavirus Infections/epidemiology
- Drug Repositioning/methods
- Humans
- Hydroxychloroquine/therapeutic use
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Lysosomes/drug effects
- Lysosomes/metabolism
- Lysosomes/virology
- Niemann-Pick C1 Protein
- Niemann-Pick Disease, Type C/drug therapy
- Niemann-Pick Disease, Type C/genetics
- Niemann-Pick Disease, Type C/metabolism
- Niemann-Pick Disease, Type C/pathology
- Pandemics
- Peptidyl-Dipeptidase A/genetics
- Peptidyl-Dipeptidase A/metabolism
- Pneumonia, Viral/diagnosis
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/epidemiology
- Protein Binding
- Receptors, Virus/antagonists & inhibitors
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- SARS-CoV-2
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/metabolism
Collapse
Affiliation(s)
| | - Tamayanthi Rajakumar
- School of Biological Sciences and Centre for
Biodiscovery, Victoria University of Wellington,
Wellington 6012, New Zealand
| | - Natalie Hammond
- School of Biological Sciences and Centre for
Biodiscovery, Victoria University of Wellington,
Wellington 6012, New Zealand
| | - Katsumi Higaki
- Division of Functional Genomics,
Tottori University, Yonago 683-8503,
Japan
| | - Zsuzsa Márka
- Department of Physics,
Columbia University, New York,
NY 10027
| | - Szabolcs Márka
- Department of Physics,
Columbia University, New York,
NY 10027
| | - Andrew B. Munkacsi
- School of Biological Sciences and Centre for
Biodiscovery, Victoria University of Wellington,
Wellington 6012, New Zealand
| |
Collapse
|
38
|
Castaldo N, Aimo A, Castiglione V, Padalino C, Emdin M, Tascini C. Safety and Efficacy of Amiodarone in a Patient With COVID-19. JACC Case Rep 2020; 2:1307-1310. [PMID: 32835273 PMCID: PMC7259916 DOI: 10.1016/j.jaccas.2020.04.053] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 12/25/2022]
Abstract
There is an urgent need for effective treatments for coronavirus disease-2019 (COVID-19). Amiodarone, like hydroxychloroquine, exerts antiviral actions by interfering with endocytosis and viral replication. Here, to our knowledge, we report the first case of a patient affected by respiratory failure related to COVID-19 who recovered after only supportive measures and a short amiodarone course. (Level of Difficulty: Beginner.).
Collapse
Affiliation(s)
- Nadia Castaldo
- Azienda Sanitaria Integrata del Friuli Centrale, Udine, Italy
| | - Alberto Aimo
- Institute of Life Science, Scuola Sant’Anna Pisa, Pisa, Italy
- Cardiology Division, University Hospital of Pisa, Pisa, Italy
- Address for correspondence: Dr. Alberto Aimo, Cardiology Division, University Hospital of Pisa, and Institute of Life Sciences, Scuola Superiore Sant’Anna, Piazza Martiri della Libertà, 33, 56124 Pisa, Italy.
| | - Vincenzo Castiglione
- Institute of Life Science, Scuola Sant’Anna Pisa, Pisa, Italy
- Cardiology Division, University Hospital of Pisa, Pisa, Italy
| | | | - Michele Emdin
- Institute of Life Science, Scuola Sant’Anna Pisa, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Carlo Tascini
- Azienda Sanitaria Integrata del Friuli Centrale, Udine, Italy
| |
Collapse
|
39
|
Dobson SJ, Mankouri J, Whitehouse A. Identification of potassium and calcium channel inhibitors as modulators of polyomavirus endosomal trafficking. Antiviral Res 2020; 179:104819. [PMID: 32389733 PMCID: PMC7205714 DOI: 10.1016/j.antiviral.2020.104819] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
During virus entry, members of the Polyomaviridae transit the endolysosomal network en route to the endoplasmic reticulum (ER), from which degraded capsids escape into the cytoplasm and enter the nucleus. Emerging evidence suggests that viruses require both endosomal acidification and the correct ionic balance of K+ and Ca2+ ions in endosomes for correct virus trafficking and genome release. Here, using two polyomaviruses with different capsid architectures, namely Simian virus 40 (SV40) and Merkel cell polyomavirus (MCPyV), we describe methods to rapidly quantify virus infection using IncuCyte ZOOM imaging analysis, and use this system to investigate the role of both K+ and Ca2+ channels during the early stages of virus entry. Using broad spectrum blockers of both K+ and Ca2+ channels to specifically target host cell ion channel functionality, we show that MCPyV, but not SV40 can be inhibited by K+ channel modulators, whilst both viruses are restricted by the broad spectrum Ca2+ channel inhibitor verapamil. Using a panel of more specific Ca2+ blockers, we show that both MCPyV and SV40 are dependent on the activity of two-pore Ca2+ channels (TPCs), as the TPC-specific blocker tetrandrine prevented capsid disassembly and nuclear transport required for virus entry. We therefore reveal a novel target to restrict the entry of polyomaviruses, which given the known role of TPCs during endolysosomal-ER fusion, is likely to be applicable to other viruses that transit this pathway. We describe novel high-throughput assays to study SV40 and MCPyV infection. MCPyV, but not SV40, is sensitive to K+ channel inhibition. Verapamil inhibits MCPyV and SV40 infection. Tetrandrine is a potent inhibitor of MCPyV and SV40 infection. Two-pore channel 1/2 activity is essential for polyomavirus entry.
Collapse
Affiliation(s)
- Samuel J Dobson
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, United Kingdom
| | - Jamel Mankouri
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, United Kingdom; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom.
| | - Adrian Whitehouse
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, United Kingdom; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
40
|
Affiliation(s)
- Alberto Aimo
- Institute of Life Science, Scuola Sant'Anna Pisa, Italy
| | | | - Michele Emdin
- Institute of Life Science, Scuola Sant'Anna Pisa, Italy.,Fondazione Toscana Gabriele Monasterio, Italy
| | - Carlo Tascini
- Azienda Sanitaria Integrata del Friuli Centrale, Italy
| |
Collapse
|
41
|
Nathan L, Lai AL, Millet JK, Straus MR, Freed JH, Whittaker GR, Daniel S. Calcium Ions Directly Interact with the Ebola Virus Fusion Peptide To Promote Structure-Function Changes That Enhance Infection. ACS Infect Dis 2020; 6:250-260. [PMID: 31746195 DOI: 10.1021/acsinfecdis.9b00296] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ebola virus disease is a serious global health concern given its periodic occurrence, high lethality, and the lack of approved therapeutics. Certain drugs that alter intracellular calcium, particularly in endolysosomes, have been shown to inhibit Ebola virus infection; however, the underlying mechanism is unknown. Here, we provide evidence that Zaire ebolavirus (EBOV) infection is promoted in the presence of calcium as a result of the direct interaction of calcium with the EBOV fusion peptide (FP). We identify the glycoprotein residues D522 and E540 in the FP as functionally critical to EBOV's interaction with calcium. We show using spectroscopic and biophysical assays that interactions of the fusion peptide with Ca2+ ions lead to lipid ordering in the host membrane during membrane fusion, and these changes are promoted at low pH and can be correlated with infectivity. We further demonstrate using circular dichroism spectroscopy that calcium interaction with the fusion peptide promotes α-helical structure of the fusion peptide, a conformational change that enhances membrane fusion, as validated using functional assays of membrane fusion. This study shows that calcium directly targets the Ebola virus fusion peptide and influences its conformation. As these residues are highly conserved across the Filoviridae, calcium's impact on fusion, and subsequently infectivity, is a key interaction that can be leveraged for developing strategies to defend against Ebola infection. This mechanistic insight provides a rationale for the use of calcium-interfering drugs already approved by the FDA as therapeutics against Ebola and enables further development of novel drugs to combat the virus.
Collapse
Affiliation(s)
- Lakshmi Nathan
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, New York 14853, United States
| | - Alex L. Lai
- Baker Laboratory, Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Jean Kaoru Millet
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Marco R. Straus
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Jack H. Freed
- Baker Laboratory, Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Gary R. Whittaker
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, New York 14853, United States
| |
Collapse
|
42
|
Host Calcium Channels and Pumps in Viral Infections. Cells 2019; 9:cells9010094. [PMID: 31905994 PMCID: PMC7016755 DOI: 10.3390/cells9010094] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 11/29/2022] Open
Abstract
Ca2+ is essential for virus entry, viral gene replication, virion maturation, and release. The alteration of host cells Ca2+ homeostasis is one of the strategies that viruses use to modulate host cells signal transduction mechanisms in their favor. Host calcium-permeable channels and pumps (including voltage-gated calcium channels, store-operated channels, receptor-operated channels, transient receptor potential ion channels, and Ca2+-ATPase) mediate Ca2+ across the plasma membrane or subcellular organelles, modulating intracellular free Ca2+. Therefore, these Ca2+ channels or pumps present important aspects of viral pathogenesis and virus–host interaction. It has been reported that viruses hijack host calcium channels or pumps, disturbing the cellular homeostatic balance of Ca2+. Such a disturbance benefits virus lifecycles while inducing host cells’ morbidity. Evidence has emerged that pharmacologically targeting the calcium channel or calcium release from the endoplasmic reticulum (ER) can obstruct virus lifecycles. Impeding virus-induced abnormal intracellular Ca2+ homeostasis is becoming a useful strategy in the development of potent antiviral drugs. In this present review, the recent identified cellular calcium channels and pumps as targets for virus attack are emphasized.
Collapse
|
43
|
DeWald LE, Johnson JC, Gerhardt DM, Torzewski LM, Postnikova E, Honko AN, Janosko K, Huzella L, Dowling WE, Eakin AE, Osborn BL, Gahagen J, Tang L, Green CE, Mirsalis JC, Holbrook MR, Jahrling PB, Dyall J, Hensley LE. In Vivo Activity of Amodiaquine against Ebola Virus Infection. Sci Rep 2019; 9:20199. [PMID: 31882748 PMCID: PMC6934550 DOI: 10.1038/s41598-019-56481-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022] Open
Abstract
During the Ebola virus disease (EVD) epidemic in Western Africa (2013‒2016), antimalarial treatment was administered to EVD patients due to the high coexisting malaria burden in accordance with World Health Organization guidelines. In an Ebola treatment center in Liberia, EVD patients receiving the combination antimalarial artesunate-amodiaquine had a lower risk of death compared to those treated with artemether-lumefantrine. As artemether and artesunate are derivatives of artemisinin, the beneficial anti-Ebola virus (EBOV) effect observed could possibly be attributed to the change from lumefantrine to amodiaquine. Amodiaquine is a widely used antimalarial in the countries that experience outbreaks of EVD and, therefore, holds promise as an approved drug that could be repurposed for treating EBOV infections. We investigated the potential anti-EBOV effect of amodiaquine in a well-characterized nonhuman primate model of EVD. Using a similar 3-day antimalarial dosing strategy as for human patients, plasma concentrations of amodiaquine in healthy animals were similar to those found in humans. However, the treatment regimen did not result in a survival benefit or decrease of disease signs in EBOV-infected animals. While amodiaquine on its own failed to demonstrate efficacy, we cannot exclude potential therapeutic value of amodiaquine when used in combination with artesunate or another antiviral.
Collapse
Affiliation(s)
- Lisa Evans DeWald
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
- Emergent BioSolutions Inc, Gaithersburg, MD, 20879, USA
| | - Joshua C Johnson
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
- AbViro LLC, Bethesda, MD, 20814, USA
| | - Dawn M Gerhardt
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
| | - Lisa M Torzewski
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
- Bioqual Inc, Rockville, MD, 20850, USA
| | - Elena Postnikova
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
| | - Anna N Honko
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Krisztina Janosko
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
| | - Louis Huzella
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
| | - William E Dowling
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Ann E Eakin
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Blaire L Osborn
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | | | - Liang Tang
- SRI International, Menlo Park, CA, 94025, USA
| | | | | | - Michael R Holbrook
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
| | - Peter B Jahrling
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
| | - Julie Dyall
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA.
| | - Lisa E Hensley
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
| |
Collapse
|
44
|
Lane TR, Massey C, Comer JE, Anantpadma M, Freundlich JS, Davey RA, Madrid PB, Ekins S. Repurposing the antimalarial pyronaridine tetraphosphate to protect against Ebola virus infection. PLoS Negl Trop Dis 2019; 13:e0007890. [PMID: 31751347 PMCID: PMC6894882 DOI: 10.1371/journal.pntd.0007890] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 12/05/2019] [Accepted: 10/29/2019] [Indexed: 12/28/2022] Open
Abstract
Recent outbreaks of the Ebola virus (EBOV) have focused attention on the dire need for antivirals to treat these patients. We identified pyronaridine tetraphosphate as a potential candidate as it is an approved drug in the European Union which is currently used in combination with artesunate as a treatment for malaria (EC50 between 420 nM—1.14 μM against EBOV in HeLa cells). Range-finding studies in mice directed us to a single 75 mg/kg i.p. dose 1 hr after infection which resulted in 100% survival and statistically significantly reduced viremia at study day 3 from a lethal challenge with mouse-adapted EBOV (maEBOV). Further, an EBOV window study suggested we could dose pyronaridine 2 or 24 hrs post-exposure to result in similar efficacy. Analysis of cytokine and chemokine panels suggests that pyronaridine may act as an immunomodulator during an EBOV infection. Our studies with pyronaridine clearly demonstrate potential utility for its repurposing as an antiviral against EBOV and merits further study in larger animal models with the added benefit of already being used as a treatment against malaria. To date there is no approved drug for Ebola Virus infection. Our approach has been to assess drugs that are already approved for other uses in various countries. Using computational models, we have previously identified three such drugs and demonstrated their activity against the Ebola virus in vitro. We now report on the in vitro absorption, metabolism, distribution, excretion and pharmacokinetic properties of one of these molecules, namely the antimalarial pyronaridine. We justify efficacy testing in the mouse model of ebola infection. We also demonstrate that a single dose of this drug is 100% effective against the virus. This study provides important preclinical evaluation of this already approved drug and justifies its selection for larger animal efficacy studies.
Collapse
Affiliation(s)
- Thomas R. Lane
- Collaborations Pharmaceuticals, Inc., Raleigh, NC, United States of America
| | - Christopher Massey
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Jason E. Comer
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
- Institutional Office of Regulated Nonclinical Studies, University of Texas Medical Branch, Galveston, TX, United States of America
- Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Manu Anantpadma
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, United States of America
| | - Joel S. Freundlich
- Departments of Pharmacology, Physiology, and Neuroscience & Medicine, Center for Emerging and Reemerging Pathogens, Rutgers University–New Jersey Medical School, NJ, United States of America
| | - Robert A. Davey
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, United States of America
| | | | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., Raleigh, NC, United States of America
- * E-mail:
| |
Collapse
|
45
|
Dyall J, Johnson JC, Hart BJ, Postnikova E, Cong Y, Zhou H, Gerhardt DM, Michelotti J, Honko AN, Kern S, DeWald LE, O'Loughlin KG, Green CE, Mirsalis JC, Bennett RS, Olinger GG, Jahrling PB, Hensley LE. In Vitro and In Vivo Activity of Amiodarone Against Ebola Virus. J Infect Dis 2019; 218:S592-S596. [PMID: 30016444 DOI: 10.1093/infdis/jiy345] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/10/2018] [Indexed: 12/18/2022] Open
Abstract
At the onset of the 2013-2016 epidemic of Ebola virus disease (EVD), no vaccine or antiviral medication was approved for treatment. Therefore, considerable efforts were directed towards the concept of drug repurposing or repositioning. Amiodarone, an approved multi-ion channel blocker for the treatment of cardiac arrhythmia, was reported to inhibit filovirus entry in vitro. Compassionate use of amiodarone in EVD patients indicated a possible survival benefit. In support of further clinical testing, we confirmed anti-Ebola virus activity of amiodarone in different cell types. Despite promising in vitro results, amiodarone failed to protect guinea pigs from a lethal dose of Ebola virus.
Collapse
Affiliation(s)
- Julie Dyall
- Integrated Research Facility, Frederick, Maryland
| | | | - Brit J Hart
- Integrated Research Facility, Frederick, Maryland
| | | | - Yu Cong
- Integrated Research Facility, Frederick, Maryland
| | | | | | | | - Anna N Honko
- Integrated Research Facility, Frederick, Maryland
| | - Steven Kern
- Bill & Melinda Gates Foundation, Seattle, Washington
| | | | | | | | | | | | | | - Peter B Jahrling
- Integrated Research Facility, Frederick, Maryland
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | | |
Collapse
|
46
|
Mirza MU, Vanmeert M, Ali A, Iman K, Froeyen M, Idrees M. Perspectives towards antiviral drug discovery against Ebola virus. J Med Virol 2019; 91:2029-2048. [PMID: 30431654 PMCID: PMC7166701 DOI: 10.1002/jmv.25357] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/04/2018] [Indexed: 12/18/2022]
Abstract
Ebola virus disease (EVD), caused by Ebola viruses, resulted in more than 11 500 deaths according to a recent 2018 WHO report. With mortality rates up to 90%, it is nowadays one of the most deadly infectious diseases. However, no Food and Drug Administration‐approved Ebola drugs or vaccines are available yet with the mainstay of therapy being supportive care. The high fatality rate and absence of effective treatment or vaccination make Ebola virus a category‐A biothreat pathogen. Fortunately, a series of investigational countermeasures have been developed to control and prevent this global threat. This review summarizes the recent therapeutic advances and ongoing research progress from research and development to clinical trials in the development of small‐molecule antiviral drugs, small‐interference RNA molecules, phosphorodiamidate morpholino oligomers, full‐length monoclonal antibodies, and vaccines. Moreover, difficulties are highlighted in the search for effective countermeasures against EVD with additional focus on the interplay between available in silico prediction methods and their evidenced potential in antiviral drug discovery.
Collapse
Affiliation(s)
- Muhammad Usman Mirza
- Department of Pharmaceutical Sciences, REGA Institute for Medical Research, Medicinal Chemistry, KU Leuven, Leuven, Belgium
| | - Michiel Vanmeert
- Department of Pharmaceutical Sciences, REGA Institute for Medical Research, Medicinal Chemistry, KU Leuven, Leuven, Belgium
| | - Amjad Ali
- Department of Genetics, Hazara University, Mansehra, Pakistan.,Molecular Virology Laboratory, Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Kanzal Iman
- Biomedical Informatics Research Laboratory (BIRL), Department of Biology, Lahore University of Management Sciences (LUMS), Lahore, Pakistan
| | - Matheus Froeyen
- Department of Pharmaceutical Sciences, REGA Institute for Medical Research, Medicinal Chemistry, KU Leuven, Leuven, Belgium
| | - Muhammad Idrees
- Molecular Virology Laboratory, Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan.,Hazara University Mansehra, Khyber Pakhtunkhwa Pakistan
| |
Collapse
|
47
|
Cherng JH, Hsu YJ, Liu CC, Tang SH, Sartika D, Chang SJ, Fan GY, Wu ST, Meng E. Activities of Ca 2+-related ion channels during the formation of kidney stones in an infection-induced urolithiasis rat model. Am J Physiol Renal Physiol 2019; 317:F1342-F1349. [PMID: 31509008 DOI: 10.1152/ajprenal.00199.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Bacterial infection has long been recognized to contribute to struvite urinary stone deposition; however, its contribution to the development of chronic kidney stones has not been extensively investigated. In the present study, we hypothesized another possible method of bacteria contributing to the formation of calcium oxalate (CaOx) that accounts for the biggest part of the kidney stone. Bacteria may play important roles by influencing renal Ca2+-related ion channel activities, resulting in chronic inflammation of the kidney along with rapid aggregation of stones. We examined the correlation among infection-promoted CaOx kidney stones and alterations in Ca2+-related ion channels in an animal model with experimentally induced Proteus mirabilis and foreign body infection. After the bladder was infected for 7 days, the data demonstrated that stones were presented and induced severe renal tubular breakage as well as altered levels of monocyte chemoattractant protein-1, cyclooxygenase-2, osteopontin, and transient receptor potential vanilloid member 5 expression, reflecting responses of kidney ion channels. Monocyte chemoattractant protein-1, osteopontin, and transient receptor potential vanilloid member 5 expression was significantly downregulated over time, indicating the chronic inflammation phase of the kidney and accelerated aggregation of CaOx crystals, respectively, whereas cyclooxygenase-2 exhibited no differences. These results indicated that bacterial infection is considerably correlated with an alteration in renal Ca2+-related ion channels and might support specific and targeted Ca2+-related ion channel-based therapeutics for urolithiasis and related inflammatory renal damage.
Collapse
Affiliation(s)
- Juin-Hong Cherng
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Department of Gerontological Health Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, Republic of China
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chuan-Chieh Liu
- Department of Cardiology, Cardinal Tien Hospital, Taipei, Taiwan, Republic of China
| | - Shou-Hung Tang
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Dewi Sartika
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Shu-Jen Chang
- Division of Rheumatology/Immunology/Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Gang-Yi Fan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Sheng-Tang Wu
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - En Meng
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
48
|
McMullan LK, Flint M, Chakrabarti A, Guerrero L, Lo MK, Porter D, Nichol ST, Spiropoulou CF, Albariño C. Characterisation of infectious Ebola virus from the ongoing outbreak to guide response activities in the Democratic Republic of the Congo: a phylogenetic and in vitro analysis. THE LANCET. INFECTIOUS DISEASES 2019; 19:1023-1032. [PMID: 31300330 PMCID: PMC11144275 DOI: 10.1016/s1473-3099(19)30291-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/28/2019] [Accepted: 06/03/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND The ongoing Ebola virus outbreak in the Ituri and North Kivu Provinces of the Democratic Republic of the Congo, which began in July, 2018, is the second largest ever recorded. Despite civil unrest, outbreak control measures and the administration of experimental therapies and a vaccine have been initiated. The aim of this study was to test the efficacy of candidate therapies and diagnostic tests with the outbreak strain Ituri Ebola virus. Lacking a virus isolate from this outbreak, a recombinant Ituri Ebola virus was compared with a similarly engineered Makona virus from the 2013-16 outbreak. METHODS Using Ebola virus sequences provided by organisations in DR Congo and a reverse genetics system, we generated an authentic Ebola virus from the ongoing outbreak in Ituri and North Kivu provinces. To relate this virus to other Ebola viruses in DR Congo, we did a phylogenetic analysis of representative complete Ebola virus genome sequences from previous outbreaks. We evaluated experimental therapies being tested in clinical trials in DR Congo, including remdesivir and ZMapp monoclonal antibodies, for their ability to inhibit the growth of infectious Ituri Ebola virus in cell culture. We also tested diagnostic assays for detection of the Ituri Ebola virus sequence. FINDINGS The phylogenetic analysis of whole-genome sequences from each Ebola virus outbreak suggests there are at least two Ebola virus strains in DR Congo, which have independently crossed into the human population. The Ituri Ebola strain initially grew slower than the Makona strain, yet reached similar mean yields of 3 × 107 50% tissue culture infectious dose by 72 h infection in Huh-7 cells. Ituri Ebola virus was similar to Makona in its susceptibility to inhibition by remdesivir and to neutralisation by monoclonal antibodies from ZMapp and other monoclonal antibodies. Remdesivir inhibited Ituri Ebola virus at a 50% effective concentration (EC50) of 12nM (with a selectivity index of 303) and Makona Ebola virus at 13nM (with a selectivity index of 279). The Zmapp monoclonal antibodies 2G4 and 4G7 neutralised Ituri Ebola virus with a mean EC50 of 0·24 μg/mL and 0·48 μg/mL, and Makona Ebola virus with a mean EC50 of 0·45 μg/mL and 0·2 μg/mL. The Xpert Ebola and US Centers for Disease Control and Prevention real-time RT-qPCR diagnostic assays detected Ituri and Makona Ebola virus sequences with similar sensitivities and efficiencies, despite primer site binding mismatches in the Ituri Ebola virus. INTERPRETATION Our findings provide a rationale for the continued testing of investigational therapies, confirm the effectiveness of the diagnostic assays used in the region, and establish a paradigm for the use of reverse genetics to inform response activities in an outbreak. FUNDING US Centers for Disease Control and Prevention.
Collapse
Affiliation(s)
- Laura K McMullan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Mike Flint
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Ayan Chakrabarti
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Lisa Guerrero
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Michael K Lo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - César Albariño
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
49
|
Arbidol and Other Low-Molecular-Weight Drugs That Inhibit Lassa and Ebola Viruses. J Virol 2019; 93:JVI.02185-18. [PMID: 30700611 DOI: 10.1128/jvi.02185-18] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/22/2019] [Indexed: 02/08/2023] Open
Abstract
Antiviral therapies that impede virus entry are attractive because they act on the first phase of the infectious cycle. Drugs that target pathways common to multiple viruses are particularly desirable when laboratory-based viral identification may be challenging, e.g., in an outbreak setting. We are interested in identifying drugs that block both Ebola virus (EBOV) and Lassa virus (LASV), two unrelated but highly pathogenic hemorrhagic fever viruses that have caused outbreaks in similar regions in Africa and share features of virus entry: use of cell surface attachment factors, macropinocytosis, endosomal receptors, and low pH to trigger fusion in late endosomes. Toward this goal, we directly compared the potency of eight drugs known to block EBOV entry with their potency as inhibitors of LASV entry. Five drugs (amodiaquine, apilimod, arbidol, niclosamide, and zoniporide) showed roughly equivalent degrees of inhibition of LASV and EBOV glycoprotein (GP)-bearing pseudoviruses; three (clomiphene, sertraline, and toremifene) were more potent against EBOV. We then focused on arbidol, which is licensed abroad as an anti-influenza drug and exhibits activity against a diverse array of clinically relevant viruses. We found that arbidol inhibits infection by authentic LASV, inhibits LASV GP-mediated cell-cell fusion and virus-cell fusion, and, reminiscent of its activity on influenza virus hemagglutinin, stabilizes LASV GP to low-pH exposure. Our findings suggest that arbidol inhibits LASV fusion, which may partly involve blocking conformational changes in LASV GP. We discuss our findings in terms of the potential to develop a drug cocktail that could inhibit both LASV and EBOV.IMPORTANCE Lassa and Ebola viruses continue to cause severe outbreaks in humans, yet there are only limited therapeutic options to treat the deadly hemorrhagic fever diseases they cause. Because of overlapping geographic occurrences and similarities in mode of entry into cells, we seek a practical drug or drug cocktail that could be used to treat infections by both viruses. Toward this goal, we directly compared eight drugs, approved or in clinical testing, for the ability to block entry mediated by the glycoproteins of both viruses. We identified five drugs with approximately equal potencies against both. Among these, we investigated the modes of action of arbidol, a drug licensed abroad to treat influenza infections. We found, as shown for influenza virus, that arbidol blocks fusion mediated by the Lassa virus glycoprotein. Our findings encourage the development of a combination of approved drugs to treat both Lassa and Ebola virus diseases.
Collapse
|
50
|
Edwards MR, Basler CF. Current status of small molecule drug development for Ebola virus and other filoviruses. Curr Opin Virol 2019; 35:42-56. [PMID: 31003196 PMCID: PMC6556423 DOI: 10.1016/j.coviro.2019.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/12/2019] [Indexed: 12/16/2022]
Abstract
The filovirus family includes some of the deadliest viruses known, including Ebola virus and Marburg virus. These viruses cause periodic outbreaks of severe disease that can be spread from person to person, making the filoviruses important public health threats. There remains a need for approved drugs that target all or most members of this virus family. Small molecule inhibitors that target conserved functions hold promise as pan-filovirus therapeutics. To date, compounds that effectively target virus entry, genome replication, gene expression, and virus egress have been described. The most advanced inhibitors are nucleoside analogs that target viral RNA synthesis reactions.
Collapse
Affiliation(s)
- Megan R Edwards
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, United States
| | - Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, United States.
| |
Collapse
|