1
|
Kilinç G, Boland R, Heemskerk MT, Spaink HP, Haks MC, van der Vaart M, Ottenhoff THM, Meijer AH, Saris A. Host-directed therapy with amiodarone in preclinical models restricts mycobacterial infection and enhances autophagy. Microbiol Spectr 2024; 12:e0016724. [PMID: 38916320 PMCID: PMC11302041 DOI: 10.1128/spectrum.00167-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/23/2024] [Indexed: 06/26/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) as well as nontuberculous mycobacteria are intracellular pathogens whose treatment is extensive and increasingly impaired due to the rise of mycobacterial drug resistance. The loss of antibiotic efficacy has raised interest in the identification of host-directed therapeutics (HDT) to develop novel treatment strategies for mycobacterial infections. In this study, we identified amiodarone as a potential HDT candidate that inhibited both intracellular Mtb and Mycobacterium avium in primary human macrophages without directly impairing bacterial growth, thereby confirming that amiodarone acts in a host-mediated manner. Moreover, amiodarone induced the formation of (auto)phagosomes and enhanced autophagic targeting of mycobacteria in macrophages. The induction of autophagy by amiodarone is likely due to enhanced transcriptional regulation, as the nuclear intensity of the transcription factor EB, the master regulator of autophagy and lysosomal biogenesis, was strongly increased. Furthermore, blocking lysosomal degradation with bafilomycin impaired the host-beneficial effect of amiodarone. Finally, amiodarone induced autophagy and reduced bacterial burden in a zebrafish embryo model of tuberculosis, thereby confirming the HDT activity of amiodarone in vivo. In conclusion, we have identified amiodarone as an autophagy-inducing antimycobacterial HDT that improves host control of mycobacterial infections. IMPORTANCE Due to the global rise in antibiotic resistance, there is a strong need for alternative treatment strategies against intracellular bacterial infections, including Mycobacterium tuberculosis (Mtb) and non-tuberculous mycobacteria. Stimulating host defense mechanisms by host-directed therapy (HDT) is a promising approach for treating mycobacterial infections. This study identified amiodarone, an antiarrhythmic agent, as a potential HDT candidate that inhibits the survival of Mtb and Mycobacterium avium in primary human macrophages. The antimycobacterial effect of amiodarone was confirmed in an in vivo tuberculosis model based on Mycobacterium marinum infection of zebrafish embryos. Furthermore, amiodarone induced autophagy and inhibition of the autophagic flux effectively impaired the host-protective effect of amiodarone, supporting that activation of the host (auto)phagolysosomal pathway is essential for the mechanism of action of amiodarone. In conclusion, we have identified amiodarone as an autophagy-inducing HDT that improves host control of a wide range of mycobacteria.
Collapse
Affiliation(s)
- Gül Kilinç
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Ralf Boland
- Institute of Biology Leiden, Leiden University, Leiden, the Netherlands
| | - Matthias T. Heemskerk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Herman P. Spaink
- Institute of Biology Leiden, Leiden University, Leiden, the Netherlands
| | - Mariëlle C. Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Anno Saris
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
2
|
Yokota Y, Takaki K, Baba K, Sasaki S, Hirano H, Osada H, Kataoka T. Amiodarone inhibits the Toll-like receptor 3-mediated nuclear factor κB signaling pathway by blocking organelle acidification. Biochem Biophys Res Commun 2024; 708:149801. [PMID: 38531219 DOI: 10.1016/j.bbrc.2024.149801] [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: 02/27/2024] [Accepted: 03/18/2024] [Indexed: 03/28/2024]
Abstract
Toll-like receptor (TLR) agonists or pro-inflammatory cytokines converge to activate the nuclear factor κB (NF-κB) signaling pathway, which provokes inflammatory responses. In the present study, we identified amiodarone hydrochloride as a selective inhibitor of the TLR3-mediated NF-κB signaling pathway by screening the RIKEN NPDepo Chemical Library. In human umbilical vein endothelial cells (HUVEC), amiodarone selectively inhibited the expression of intercellular adhesion molecule-1 (ICAM-1) induced by polyinosinic-polycytidylic acid (Poly(I:C)), but not tumor necrosis factor-α, interleukin-1α, or lipopolysaccharide. In response to a Poly(I:C) stimulation, amiodarone at 20 μM reduced the up-regulation of mRNA expression encoding ICAM-1, vascular cell adhesion molecule-1, and E-selectin. The nuclear translocation of the NF-κB subunit RelA was inhibited by amiodarone at 15-20 μM in Poly(I:C)-stimulated HUVEC. Amiodarone diminished the fluorescent dots of LysoTracker® Red DND-99 scattered over the cytoplasm of HUVEC. Therefore, the present study revealed that amiodarone selectively inhibited the TLR3-mediated NF-κB signaling pathway by blocking the acidification of intracellular organelles.
Collapse
Affiliation(s)
- Yuka Yokota
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Keiko Takaki
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Kosuke Baba
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Saki Sasaki
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Hiroyuki Hirano
- Chemical Resource Development Research Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama, 351-0198, Japan
| | - Hiroyuki Osada
- Chemical Resource Development Research Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama, 351-0198, Japan; Department of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Takao Kataoka
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan; Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan.
| |
Collapse
|
3
|
Ghafouri E, Bigdeli M, Khalafiyan A, Amirkhani Z, Ghanbari R, Hasan A, Khanahmad H, Boshtam M, Makvandi P. Unmasking the complex roles of hypocalcemia in cancer, COVID-19, and sepsis: Engineered nanodelivery and diagnosis. ENVIRONMENTAL RESEARCH 2023; 238:116979. [PMID: 37660871 DOI: 10.1016/j.envres.2023.116979] [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: 07/10/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/05/2023]
Abstract
Calcium (Ca2+) homeostasis is essential for maintaining physiological processes in the body. Disruptions in Ca2+ signaling can lead to various pathological conditions including inflammation, fibrosis, impaired immune function, and accelerated senescence. Hypocalcemia, a common symptom in diseases such as acute respiratory distress syndrome (ARDS), cancer, septic shock, and COVID-19, can have both potential protective and detrimental effects. This article explores the multifaceted role of Ca2+ dysregulation in inflammation, fibrosis, impaired immune function, and accelerated senescence, contributing to disease severity. Targeting Ca2+ signaling pathways may provide opportunities to develop novel therapeutics for age-related diseases and combat viral infections. However, the role of Ca2+ in viral infections is complex, and evidence suggests that hypocalcemia may have a protective effect against certain viruses, while changes in Ca2+ homeostasis can influence susceptibility to viral infections. The effectiveness and safety of Ca2+ supplements in COVID-19 patients remain a subject of ongoing research and debate. Further investigations are needed to understand the intricate interplay between Ca2+ signaling and disease pathogenesis.
Collapse
Affiliation(s)
- Elham Ghafouri
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Anis Khalafiyan
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zohre Amirkhani
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roham Ghanbari
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| |
Collapse
|
4
|
Strizzi S, Bernardo L, D'Ursi P, Urbinati C, Bianco A, Limanaqi F, Manconi A, Milanesi M, Macchi A, Di Silvestre D, Cavalleri A, Pareschi G, Rusnati M, Clerici M, Mauri P, Biasin M. An innovative strategy to investigate microbial protein modifications in a reliable fast and sensitive way: A therapy oriented proof of concept based on UV-C irradiation of SARS-CoV-2 spike protein. Pharmacol Res 2023; 194:106862. [PMID: 37479104 DOI: 10.1016/j.phrs.2023.106862] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/04/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
The characterization of modifications of microbial proteins is of primary importance to dissect pathogen lifecycle mechanisms and could be useful in identifying therapeutic targets. Attempts to solve this issue yielded only partial and non-exhaustive results. We developed a multidisciplinary approach by coupling in vitro infection assay, mass spectrometry (MS), protein 3D modelling, and surface plasma resonance (SPR). As a proof of concept, the effect of low UV-C (273 nm) irradiation on SARS-CoV-2 spike (S) protein was investigated. Following UV-C exposure, MS analysis identified, among other modifications, the disruption of a disulphide bond within the conserved S2 subunit of S protein. Computational analyses revealed that this bond breakage associates with an allosteric effect resulting in the generation of a closed conformation with a reduced ability to bind the ACE2 receptor. The UV-C-induced reduced affinity of S protein for ACE2 was further confirmed by SPR analyses and in vitro infection assays. This comprehensive approach pinpoints the S2 domain of S protein as a potential therapeutic target to prevent SARS-CoV-2 infection. Notably, this workflow could be used to screen a wide variety of microbial protein domains, resulting in a precise molecular fingerprint and providing new insights to adequately address future epidemics.
Collapse
Affiliation(s)
- Sergio Strizzi
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, 20122 Milan, Italy
| | - Letizia Bernardo
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), 20054 Segrate, MI, Italy
| | - Pasqualina D'Ursi
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), 20054 Segrate, MI, Italy
| | - Chiara Urbinati
- Unit of Macromolecular Interaction Analysis, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Andrea Bianco
- Italian National Institute for Astrophysics (INAF) - Brera Astronomical Observatory, Via E. Bianchi, 46, Merate, 23807 Lecco, Italy
| | - Fiona Limanaqi
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, 20122 Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza, 20122 Milan, Italy
| | - Andrea Manconi
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), 20054 Segrate, MI, Italy
| | - Maria Milanesi
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), 20054 Segrate, MI, Italy; Unit of Macromolecular Interaction Analysis, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Alberto Macchi
- Italian National Institute for Astrophysics (INAF) - Brera Astronomical Observatory, Via E. Bianchi, 46, Merate, 23807 Lecco, Italy
| | - Dario Di Silvestre
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), 20054 Segrate, MI, Italy
| | - Adalberto Cavalleri
- Epidemiology and Prevention Unit, IRCCS Foundation, Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milan, Italy
| | - Giovanni Pareschi
- Italian National Institute for Astrophysics (INAF) - Brera Astronomical Observatory, Via E. Bianchi, 46, Merate, 23807 Lecco, Italy
| | - Marco Rusnati
- Unit of Macromolecular Interaction Analysis, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza, 20122 Milan, Italy; Don C. Gnocchi Foundation, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation, Via A. Capecelatro 66, 20148 Milan, íItaly
| | - PierLuigi Mauri
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), 20054 Segrate, MI, Italy; Interdisciplinary Research Center "Health Science", Sant'Anna School of Advanced Studies, 56127 Pisa, Italy.
| | - Mara Biasin
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, 20122 Milan, Italy
| |
Collapse
|
5
|
Lim SP. Targeting SARS-CoV-2 and host cell receptor interactions. Antiviral Res 2023; 210:105514. [PMID: 36581047 PMCID: PMC9792186 DOI: 10.1016/j.antiviral.2022.105514] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Despite the availability of vaccines and therapeutics, continual genetic alterations render the severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) a persistent threat, particularly for the immunocompromised and elderly. Through interactions of its spike (S) protein with different receptors and coreceptors on host cell surfaces, the virus enters the cell either via fusion with the plasma membrane or through endocytosis. Angiotensin-converting enzyme 2 (ACE2) has been identified as a key receptor utilized by SARS-CoV-2 and related human coronaviruses to mediate cell entry in the lung airways. Auxiliary SARS-CoV-2 entry receptors such as ASGPR1, Kremen protein 1, integrins have also been reported. In this review, therapeutic approaches to block SARS-CoV-2 and host cell receptor interactions are discussed.
Collapse
Affiliation(s)
- Siew Pheng Lim
- Experimental Drug Development Centre (EDDC), A*STAR, 10, Biopolis Road, #05-01, Chromos, 138670, Singapore.
| |
Collapse
|
6
|
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
|
7
|
Korade Z, Tallman KA, Kim HYH, Balog M, Genaro-Mattos TC, Pattnaik A, Mirnics K, Pattnaik AK, Porter NA. Dose-Response Effects of 7-Dehydrocholesterol Reductase Inhibitors on Sterol Profiles and Vesicular Stomatitis Virus Replication. ACS Pharmacol Transl Sci 2022; 5:1086-1096. [PMID: 36407960 PMCID: PMC9667548 DOI: 10.1021/acsptsci.2c00051] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Indexed: 11/29/2022]
Abstract
Cholesterol is ubiquitous in cells; it plays a critical role in membrane structure and transport as well as in intracellular trafficking processes. There are suggestions that cholesterol metabolism is linked to innate immunity with inhibitors of DHCR7, the last enzyme in the cholesterol pathway, suggested to have potential as viral therapeutics nearly a decade ago. In fact, there are a number of highly prescribed pharmaceuticals that are off-target inhibitors of DHCR7, causing increased cellular levels of 7-dehydrodesmosterol (7-DHD) and 7-dehydrocholesterol (7-DHC). We report here dose-response studies of six such inhibitors on late-stage cholesterol biosynthesis in Neuro2a cells as well as their effect on infection of vesicular stomatitis virus (VSV). Four of the test compounds are FDA-approved drugs (cariprazine, trazodone, metoprolol, and tamoxifen), one (ifenprodil) has been the object of a recent Phase 2b COVID trial, and one (AY9944) is an experimental compound that has seen extensive use as a DHCR7 inhibitor. The three FDA-approved drugs inhibit replication of a GFP-tagged VSV with efficacies that mirror their effect on DHCR7. Ifenprodil and AY9944 have complex inhibitory profiles, acting on both DHCR7 and DHCR14, while tamoxifen does not inhibit DHCR7 and is toxic to Neuro2a at concentrations where it inhibits the Δ7-Δ8 isomerase of the cholesterol pathway. VSV itself affects the sterol profile in Neuro2a cells, showing a dose-response increase of dehydrolathosterol and lathosterol, the substrates for DHCR7, with a corresponding decrease in desmosterol and cholesterol. 7-DHD and 7-DHC are orders of magnitude more vulnerable to free radical chain oxidation than other sterols as well as polyunsaturated fatty esters, and the effect of these sterols on viral infection is likely a reflection of this fact of Nature.
Collapse
Affiliation(s)
- Zeljka Korade
- Department
of Pediatrics, Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Keri A. Tallman
- Department
of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Hye-Young H. Kim
- Department
of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Marta Balog
- Munroe-Meyer
Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska 68105, United States
- Department
of Medical Biology and Genetics, Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek 31000, Croatia
| | - Thiago C. Genaro-Mattos
- Munroe-Meyer
Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska 68105, United States
| | - Aryamav Pattnaik
- Nebraska
Center for Virology and School of Veterinary Medicine and Biomedical
Sciences, University of Nebraska-Lincoln, Lincoln 68583, United States
| | - Károly Mirnics
- Munroe-Meyer
Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska 68105, United States
| | - Asit K. Pattnaik
- Nebraska
Center for Virology and School of Veterinary Medicine and Biomedical
Sciences, University of Nebraska-Lincoln, Lincoln 68583, United States
| | - Ned A. Porter
- Department
of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
8
|
Jamal QMS. Antiviral Potential of Plants against COVID-19 during Outbreaks-An Update. Int J Mol Sci 2022; 23:13564. [PMID: 36362351 PMCID: PMC9655040 DOI: 10.3390/ijms232113564] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/06/2022] [Accepted: 11/02/2022] [Indexed: 12/01/2023] Open
Abstract
Several human diseases are caused by viruses, including cancer, Type I diabetes, Alzheimer's disease, and hepatocellular carcinoma. In the past, people have suffered greatly from viral diseases such as polio, mumps, measles, dengue fever, SARS, MERS, AIDS, chikungunya fever, encephalitis, and influenza. Recently, COVID-19 has become a pandemic in most parts of the world. Although vaccines are available to fight the infection, their safety and clinical trial data are still questionable. Social distancing, isolation, the use of sanitizer, and personal productive strategies have been implemented to prevent the spread of the virus. Moreover, the search for a potential therapeutic molecule is ongoing. Based on experiences with outbreaks of SARS and MERS, many research studies reveal the potential of medicinal herbs/plants or chemical compounds extracted from them to counteract the effects of these viral diseases. COVID-19's current status includes a decrease in infection rates as a result of large-scale vaccination program implementation by several countries. But it is still very close and needs to boost people's natural immunity in a cost-effective way through phytomedicines because many underdeveloped countries do not have their own vaccination facilities. In this article, phytomedicines as plant parts or plant-derived metabolites that can affect the entry of a virus or its infectiousness inside hosts are described. Finally, it is concluded that the therapeutic potential of medicinal plants must be analyzed and evaluated entirely in the control of COVID-19 in cases of uncontrollable SARS infection.
Collapse
Affiliation(s)
- Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah 52741, Saudi Arabia
| |
Collapse
|
9
|
Hullin-Matsuda F, Colosetti P, Rabia M, Luquain-Costaz C, Delton I. Exosomal lipids from membrane organization to biomarkers: Focus on an endolysosomal-specific lipid. Biochimie 2022; 203:77-92. [DOI: 10.1016/j.biochi.2022.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022]
|
10
|
Antiviral Effects of ABMA and DABMA against Influenza Virus In Vitro and In Vivo via Regulating the Endolysosomal Pathway and Autophagy. Int J Mol Sci 2022; 23:ijms23073940. [PMID: 35409297 PMCID: PMC8999625 DOI: 10.3390/ijms23073940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
Influenza virus is an acute and highly contagious respiratory pathogen that causes great concern to public health and for which there is a need for extensive drug discovery. The small chemical compound ABMA and its analog DABMA, containing an adamantane or a dimethyl-adamantane group, respectively, have been demonstrated to inhibit multiple toxins (diphtheria toxin, Clostridium difficile toxin B, Clostridium sordellii lethal toxin) and viruses (Ebola, rabies virus, HSV-2) by acting on the host’s vesicle trafficking. Here, we showed that ABMA and DABMA have antiviral effects against both amantadine-sensitive influenza virus subtypes (H1N1 and H3N2), amantadine-resistant subtypes (H3N2), and influenza B virus with EC50 values ranging from 2.83 to 7.36 µM (ABMA) and 1.82 to 6.73 µM (DABMA), respectively. ABMA and DABMA inhibited the replication of influenza virus genomic RNA and protein synthesis by interfering with the entry stage of the virus. Molecular docking evaluation together with activity against amantadine-resistant influenza virus strains suggested that ABMA and DABMA were not acting as M2 ion channel blockers. Subsequently, we found that early internalized H1N1 virions were retained in accumulated late endosome compartments after ABMA treatment. Additionally, ABMA disrupted the early stages of the H1N1 life cycle or viral RNA synthesis by interfering with autophagy. ABMA and DABMA protected mice from an intranasal H1N1 challenge with an improved survival rate of 67%. The present study suggests that ABMA and DABMA are potential antiviral leads for the development of a host-directed treatment against influenza virus infection.
Collapse
|
11
|
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: 33] [Impact Index Per Article: 16.5] [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
|
12
|
Tarantino N, Della Rocca DG, Zou F, Lin A, Natale A, Di Biase L. Prevalence, Outcomes, and Management of Ventricular Arrhythmias in COVID-19 Patients. Card Electrophysiol Clin 2022; 14:11-20. [PMID: 35221078 PMCID: PMC8554003 DOI: 10.1016/j.ccep.2021.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We review the current data on epidemiology, the clinical significance, the pathophysiologic mechanisms, and the treatment of VAs in the setting of COVID-19. VAs prevail in 0.15% to 8% of hospitalized patients, but only sustained and rapid tachyarrhythmias are purportedly associated with a significant increase in mortality. Multiple factors can elicit VAs, which are ultimately deemed to be a marker of severe systemic disease rather than a distinct cardiac condition. Even though the electrophysiologist plays a determinant role in the secondary prevention of VAs, a multidisciplinary approach is indispensable for primary prophylaxis and acute management.
Collapse
Affiliation(s)
- Nicola Tarantino
- Montefiore Medical Center, 111 E 210th street, Bronx, NY 10467, USA
| | - Domenico G Della Rocca
- Texas Cardiac Arrhythmia Institute at St. David's Medical Center, 3000 N I-35, Suite 720, Austin, TX 78705, USA
| | - Fengwei Zou
- Montefiore Medical Center, 111 E 210th street, Bronx, NY 10467, USA
| | - Aung Lin
- Montefiore Medical Center, 111 E 210th street, Bronx, NY 10467, USA
| | - Andrea Natale
- Texas Cardiac Arrhythmia Institute at St. David's Medical Center, 3000 N I-35, Suite 720, Austin, TX 78705, USA; Scripps Interventional Car, 9834 Genesee Ave, La Jolla, CA 92037, USA; Health Education Campus, 9501 Euclid Ave, Cleveland, OH 44106, USA
| | - Luigi Di Biase
- Montefiore Medical Center, 111 E 210th street, Bronx, NY 10467, USA; Texas Cardiac Arrhythmia Institute at St. David's Medical Center, 3000 N I-35, Suite 720, Austin, TX 78705, USA.
| |
Collapse
|
13
|
Korn D, Pervitsky V, Bobrowski T, Alves VM, Schmitt C, Bizon C, Baker N, Chirkova R, Cherkasov A, Muratov E, Tropsha A. COVID-19 Knowledge Extractor (COKE): A Curated Repository of Drug-Target Associations Extracted from the CORD-19 Corpus of Scientific Publications on COVID-19. J Chem Inf Model 2021; 61:5734-5741. [PMID: 34783553 PMCID: PMC8610010 DOI: 10.1021/acs.jcim.1c01285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Indexed: 12/31/2022]
Abstract
The COVID-19 pandemic has catalyzed a widespread effort to identify drug candidates and biological targets of relevance to SARS-COV-2 infection, which resulted in large numbers of publications on this subject. We have built the COVID-19 Knowledge Extractor (COKE), a web application to extract, curate, and annotate essential drug-target relationships from the research literature on COVID-19. SciBiteAI ontological tagging of the COVID Open Research Data set (CORD-19), a repository of COVID-19 scientific publications, was employed to identify drug-target relationships. Entity identifiers were resolved through lookup routines using UniProt and DrugBank. A custom algorithm was used to identify co-occurrences of the target protein and drug terms, and confidence scores were calculated for each entity pair. COKE processing of the current CORD-19 database identified about 3000 drug-protein pairs, including 29 unique proteins and 500 investigational, experimental, and approved drugs. Some of these drugs are presently undergoing clinical trials for COVID-19. The COKE repository and web application can serve as a useful resource for drug repurposing against SARS-CoV-2. COKE is freely available at https://coke.mml.unc.edu/, and the code is available at https://github.com/DnlRKorn/CoKE.
Collapse
Affiliation(s)
- Daniel Korn
- Department of Computer Science, The
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
27599, United States
- Laboratory for Molecular Modeling, Division of
Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy,
The University of North Carolina at Chapel Hill, Chapel Hill,
North Carolina 27599, United States
| | - Vera Pervitsky
- Laboratory for Molecular Modeling, Division of
Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy,
The University of North Carolina at Chapel Hill, Chapel Hill,
North Carolina 27599, United States
| | - Tesia Bobrowski
- Laboratory for Molecular Modeling, Division of
Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy,
The University of North Carolina at Chapel Hill, Chapel Hill,
North Carolina 27599, United States
| | - Vinicius M. Alves
- Office of Data Science, National Toxicology Program,
NIEHS, Morrisville, North Carolina 27560, United
States
| | - Charles Schmitt
- Office of Data Science, National Toxicology Program,
NIEHS, Morrisville, North Carolina 27560, United
States
| | - Chris Bizon
- Renaissance Computing Institute, The
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
27599, United States
| | - Nancy Baker
- ParlezChem, 123 W. Union
Street, Hillsborough, North Carolina 27278, United States
| | - Rada Chirkova
- Department of Computer Science, North Carolina
State University, Raleigh, North Carolina 27606-5550, United
States
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of
British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Eugene Muratov
- Laboratory for Molecular Modeling, Division of
Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy,
The University of North Carolina at Chapel Hill, Chapel Hill,
North Carolina 27599, United States
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of
Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy,
The University of North Carolina at Chapel Hill, Chapel Hill,
North Carolina 27599, United States
| |
Collapse
|
14
|
Le Corre P, Loas G. Repurposing functional inhibitors of acid sphingomyelinase (fiasmas): an opportunity against SARS-CoV-2 infection? J Clin Pharm Ther 2021; 46:1213-1219. [PMID: 33645763 PMCID: PMC8014536 DOI: 10.1111/jcpt.13390] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/09/2021] [Accepted: 02/09/2021] [Indexed: 12/31/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Infection by SARS-CoV-2, the virus responsible of COVID-19, is associated with limited treatment options. The purpose of this study was to evaluate the rationale for repurposing functional inhibitors of acid sphingomyelinase (FIASMAs), several of which are approved medicines, for the treatment of SAR-CoV-2 infections. COMMENT We propose and discuss the FIASMAs' lysosomotropism as a possible explanation for their observed in vitro activities against viruses, and more specifically against infections caused by coronaviruses such as SARS-CoV-2. Successful in vitro-to-in vivo translation of FIASMAs requires that their pharmacokinetics (dosing regimen and drug-drug interactions) are matched with viral kinetics. WHAT IS NEW AND CONCLUSION Drug repurposing to ensure rapid patient access to effective treatment has garnered much attention in this era of the COVID-19 pandemic. The observed lysosomotropic activity of small-molecule FIASMA compounds suggests that their repurposing as potential drugs against SARS-CoV-2 is promising.
Collapse
Affiliation(s)
- Pascal Le Corre
- Pôle PharmacieService Hospitalo‐Universitaire de PharmacieCHU de RennesRennesFrance
- Univ RennesCHU RennesInsermEHESP, Irset (Institut de recherche en santé environnement et travail) ‐ UMR_S 1085RennesFrance
- Laboratoire de Biopharmacie et Pharmacie CliniqueFaculté de PharmacieUniversité de Rennes 1RennesFrance
| | - Gwenolé Loas
- Department of PsychiatryHôpital ErasmeUniversité libre de Bruxelles (ULB)BrusselsBelgium
- Research Unit (ULB 266)Hôpital ErasmeUniversité libre de Bruxelles (ULB)BrusselsBelgium
| |
Collapse
|
15
|
Heterogeneous graph attention networks for drug virus association prediction. Methods 2021; 198:11-18. [PMID: 34419588 PMCID: PMC8376526 DOI: 10.1016/j.ymeth.2021.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/11/2021] [Indexed: 12/11/2022] Open
Abstract
Coronavirus Disease-19 (COVID-19) has lead global epidemics with high morbidity and mortality. However, there are currently no proven effective drugs targeting COVID-19. Identifying drug-virus associations can not only provide insights into the understanding of drug-virus interaction mechanism, but also guide and facilitate the screening of compound candidates for antiviral drug discovery. Since conventional experiment methods are time-consuming, laborious and expensive, computational methods to identify potential drug candidates for viruses (e.g., COVID-19) provide an alternative strategy. In this work, we propose a novel framework of Heterogeneous Graph Attention Networks for Drug-Virus Association predictions, named HGATDVA. First, we fully incorporate multiple sources of biomedical data, e.g., drug chemical information, virus genome sequences and viral protein sequences, to construct abundant features for drugs and viruses. Second, we construct two drug-virus heterogeneous graphs. For each graph, we design a self-enhanced graph attention network (SGAT) to explicitly model the dependency between a node and its local neighbors and derive the graph-specific representations for nodes. Third, we further develop a neural network architecture with tri-aggregator to aggregate the graph-specific representations to generate the final node representations. Extensive experiments were conducted on two datasets, i.e., DrugVirus and MDAD, and the results demonstrated that our model outperformed 7 state-of-the-art methods. Case study on SARS-CoV-2 validated the effectiveness of our model in identifying potential drugs for viruses.
Collapse
|
16
|
Yang L, Pei RJ, Li H, Ma XN, Zhou Y, Zhu FH, He PL, Tang W, Zhang YC, Xiong J, Xiao SQ, Tong XK, Zhang B, Zuo JP. Identification of SARS-CoV-2 entry inhibitors among already approved drugs. Acta Pharmacol Sin 2021; 42:1347-1353. [PMID: 33116249 PMCID: PMC7594953 DOI: 10.1038/s41401-020-00556-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
To discover effective drugs for COVID-19 treatment amongst already clinically approved drugs, we developed a high throughput screening assay for SARS-CoV-2 virus entry inhibitors using SARS2-S pseudotyped virus. An approved drug library of 1800 small molecular drugs was screened for SARS2 entry inhibitors and 15 active drugs were identified as specific SARS2-S pseudovirus entry inhibitors. Antiviral tests using native SARS-CoV-2 virus in Vero E6 cells confirmed that 7 of these drugs (clemastine, amiodarone, trimeprazine, bosutinib, toremifene, flupenthixol, and azelastine) significantly inhibited SARS2 replication, reducing supernatant viral RNA load with a promising level of activity. Three of the drugs were classified as histamine receptor antagonists with clemastine showing the strongest anti-SARS2 activity (EC50 = 0.95 ± 0.83 µM). Our work suggests that these 7 drugs could enter into further in vivo studies and clinical investigations for COVID-19 treatment.
Collapse
|
17
|
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: 2.0] [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
|
18
|
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: 1.3] [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
|
19
|
Rai PK, Mueed Z, Chowdhury A, Deval R, Kumar D, Kamal MA, Negi YS, Pareek S, Poddar NK. Current Overviews on COVID-19 Management Strategies. Curr Pharm Biotechnol 2021; 23:361-387. [PMID: 33966618 DOI: 10.2174/1389201022666210509022313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/02/2021] [Accepted: 02/08/2021] [Indexed: 02/08/2023]
Abstract
The coronavirus pandemic has hit the world lately and caused acute respiratory syndrome in humans. The causative agent of the disease was soon brought to focus by scientists as SARS-CoV-2 and later called a novel coronavirus by the general public. Due to the severity and rapid spread of the disease, WHO classifies the COVID-19 pandemic as the 6th public health emergency even after taking efforts like worldwide quarantine and restrictions. Since only symptomatic treatment is available, the best way to control the spread of the virus is by taking preventive measures. Various types of antigen/antibody detection kits and diagnostic methods are available for the diagnosis of COVID-19 patients. In recent years, various phytochemicals and repurposing drugs are showing a broad range of anti-viral activities with different modes of action have been identified. Repurposing drugs such as arbidol, hydroxychloroquine, chloroquine, lopinavir, favipiravir, remdesivir, hexamethylene amiloride, and dexamethasone, tocilizumab, interferon-β, neutralizing antibodies exhibit in vitro anti-coronaviral properties by inhibiting multiple processes in the virus life cycle. Various research groups are involved in drug trials and vaccine development. Plant-based anti-viral compounds such as baicalin, calanolides, curcumin, oxymatrine, matrine, and resveratrol exhibit different modes of action against a wide range of positive/negative sense-RNA/DNA virus, and future researches need to be conducted to ascertain their role, use in managing SARS-CoV-2. Thus, this article is an attempt to review the current understanding of COVID-19 acute respiratory disease and summarize its clinical features with their prospective control and various aspects of the therapeutic approach.
Collapse
Affiliation(s)
- Pankaj Kumar Rai
- Department of Biotechnology, Invertis University, Bareilly, Uttar Pradesh, India
| | - Zeba Mueed
- Department of Biotechnology, Invertis University, Bareilly, Uttar Pradesh, India
| | - Abhiroop Chowdhury
- School of Environment & Sustainability, O.P. Jindal Global University, Sonipat, Haryana, India
| | - Ravi Deval
- Department of Biotechnology, Invertis University, Bareilly, Uttar Pradesh, India
| | - Dinesh Kumar
- Department of Biotechnology, Invertis University, Bareilly, Uttar Pradesh, India
| | - Mohammad A Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan. China
| | - Yogeshwar Singh Negi
- Department of Biosciences, Manipal University Jaipur, DehmiKalan, Jaipur-Ajmer Expressway, Jaipur-303007, Rajasthan, India
| | - Shubhra Pareek
- Department of Chemistry, Manipal University Jaipur, DehmiKalan, Jaipur-Ajmer Expressway, Jaipur-303007, Rajasthan, India
| | - Nitesh Kumar Poddar
- Department of Biosciences, Manipal University Jaipur, DehmiKalan, Jaipur-Ajmer Expressway, Jaipur-303007, Rajasthan, India
| |
Collapse
|
20
|
Shaath H, Alajez NM. Identification of PBMC-based molecular signature associational with COVID-19 disease severity. Heliyon 2021; 7:e06866. [PMID: 33898797 PMCID: PMC8057768 DOI: 10.1016/j.heliyon.2021.e06866] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/10/2021] [Accepted: 04/16/2021] [Indexed: 12/18/2022] Open
Abstract
The longevity of COVID-19 as a global pandemic, and the devastating effects it has had on certain subsets of individuals thus far has highlighted the importance of identifying blood-based biomarkers associated with disease severity. We employed computational and transcriptome analyses of publicly available datasets from PBMCs from 126 patients with COVID-19 admitted to ICU (n = 50), COVID-19 not admitted to ICU (n = 50), non-COVID-19 admitted to ICU (n = 16) and non-COVID-19 not admitted to ICU (n = 10), and utilized the Gencode V33 assembly to analyze protein coding mRNA and long noncoding RNA (lncRNA) transcriptomes in the context of disease severity. Our data identified several aberrantly expressed mRNA and lncRNA based biomarkers associated with SARS-CoV-2 severity, which in turn significantly affected canonical, upstream, and disease functions in each group of patients. Immune, interferon, and antiviral responses were severely suppressed in COVID-19 patients admitted to ICU versus those who were not admitted to ICU. Our data suggests a possible therapeutic approach for severe COVID-19 through administration of interferon therapy. Delving further into these biomarkers, roles and their implications on the onset and disease severity of COVID-19 could play a crucial role in patient stratification and identifying varied therapeutic options with diverse clinical implications.
Collapse
Affiliation(s)
- Hibah Shaath
- College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Nehad M. Alajez
- College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| |
Collapse
|
21
|
Darquennes G, Le Corre P, Le Moine O, Loas G. Association between Functional Inhibitors of Acid Sphingomyelinase (FIASMAs) and Reduced Risk of Death in COVID-19 Patients: A Retrospective Cohort Study. Pharmaceuticals (Basel) 2021; 14:226. [PMID: 33799977 PMCID: PMC7998809 DOI: 10.3390/ph14030226] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
Given the current scarcity of curative treatment of COVID-19, the search for an effective treatment modality among all available medications has become a priority. This study aimed at investigating the role of functional inhibitors of acid sphingomyelinase (FIASMAs) on in-hospital COVID-19 mortality. In this retrospective cohort study, we included adult in-patients with laboratory-confirmed COVID-19 between 1 March 2020 and 31 August 2020 with definite outcomes (discharged hospital or deceased) from Erasme Hospital (Brussels, Belgium). We used univariate and multivariate logistic regression models to explore the risk factors associated with in-hospital mortality. We included 350 patients (205 males, 145 females) with a mean age of 63.24 years (SD = 17.4, range: 21-96 years). Seventy-two patients died in the hospital and 278 were discharged. The four most common comorbidities were hypertension (184, 52.6%), chronic cardiac disease (110, 31.4%), obesity (96, 27.8%) and diabetes (95, 27.1%). Ninety-three participants (26.6%) received a long-term prescription for FIASMAs. Among these, 60 (64.5%) received amlodipine. For FIASMAs status, multivariable regression showed increasing odds ratio (OR) for in-hospital deaths associated with older age (OR 1.05, 95% CI: 1.02-1.07; p = 0.00015), and higher prevalence of malignant neoplasm (OR 2.09, 95% CI: 1.03-4.22; p = 0.039). Nonsignificant decreasing OR (0.53, 95% CI: 0.27-1.04; p = 0.064) was reported for FIASMA status. For amlodipine status, multivariable regression revealed increasing OR of in-hospital deaths associated with older age (OR 1.04, 95% CI: 1.02-1.07; p = 0.0009), higher prevalence of hypertension (OR 2.78, 95% CI: 1.33-5.79; p = 0.0062) and higher prevalence of malignant neoplasm (OR 2.71, 95% CI: 1.23-5.97; p = 0.013), then secondarily decreasing OR of in-hospital death associated with long-term treatment with amlodipine (OR 0.24, 95% CI: 0.09-0.62; p = 0.0031). Chronic treatment with amlodipine could be significantly associated with low mortality of COVID-19 in-patients.
Collapse
Affiliation(s)
- Gil Darquennes
- Department of Psychiatry, Hôpital Erasme, Université libre de Bruxelles (ULB), 1050 Brussels, Belgium;
- Research Unit (ULB 266), Hôpital Erasme, Université libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - Pascal Le Corre
- Pôle Pharmacie, Service Hospitalo-Universitaire de Pharmacie, CHU de Rennes, 35033 Rennes, France;
- Irset (Institut de Recherche en Santé, Environnement et Travail)—UMR_S 1085, University of Rennes, CHU Rennes, INSERM, EHESP, 35000 Rennes, France
- Laboratoire de Biopharmacie et Pharmacie Clinique, Faculté de Pharmacie, Université de Rennes 1, 35043 Rennes, France
| | - Olivier Le Moine
- Department of Medical Gastro-Enterology, Hôpital Erasme, Université libre de Bruxelles (ULB), 1050 Brussels, Belgium;
| | - Gwenolé Loas
- Department of Psychiatry, Hôpital Erasme, Université libre de Bruxelles (ULB), 1050 Brussels, Belgium;
- Research Unit (ULB 266), Hôpital Erasme, Université libre de Bruxelles (ULB), 1050 Brussels, Belgium
| |
Collapse
|
22
|
Lonare S, Hazarika M, Das CK. Antiviral effect of amiodarone and its possible role in COVID-19: An appraisal. Indian J Pharmacol 2021; 52:533-534. [PMID: 33666199 PMCID: PMC8092184 DOI: 10.4103/ijp.ijp_714_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Snehal Lonare
- Department of Pharmacology and Toxicology, GADVASU, Ludhiana, Punjab, India
| | - Mythili Hazarika
- Department of Psychiatry, Guwahati Medical College Hospital, Affiliated to Srimanta Sankaradeva University of Health Sciences, Assam, India
| | - Chandra Kr Das
- Associate Professor of Clinical Psychology, Gauhati Medical College and Hospital Affiliated to Srimanta Sankaradeva University of Health Sciences, Assam, India
| |
Collapse
|
23
|
Agostinis P, Vianello V, Dereani E, Caruso G, Montessoro B, Taurian M, Di Chiara A, Patruno V, Cavallin F, Fadini GP, Aldovini A, Baritussio A, Curcio F, Tascini C, Baritussio A. Response to a massive SARS-CoV-2 infection in a nursing home transformed into a caring center. Aging Clin Exp Res 2021; 33:443-450. [PMID: 33506312 PMCID: PMC7840424 DOI: 10.1007/s40520-020-01784-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/30/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND The best policy to follow when nursing homes are massively hit by SARS-CoV2 is unclear. AIM To describe COVID-19 containment in a nursing home transformed into a caring center. METHODS Physicians and nurses were recruited. The facility was reorganized and connected with the laboratory of the reference hospital. Ultrasound was used to diagnose pneumonia. Patients needing intensive care were transferred to the reference hospital. Hydroxychloroquine/azithromycin/enoxaparin were used initially, while amiodarone/enoxaparin were used at a later phase. Under both regimens, methylprednisolone was added for severe cases. Prophylaxis was done with hydroxychloroquine initially and then with amiodarone. PERIOD COVERED March 22-July 31, 2020. RESULTS The facility was reorganized in two days. Ninety-two guests of the 121 (76%) and 25 personnel of 118 (21.1%) became swab test positive. Seven swab test negative patients who developed symptoms were considered to have COVID-19. Twenty-seven patients died, 23 swab test positive, 5 of whom after full recovery. Four patients needing intensive care were transferred (3 died). Mortality, peaking in April 2020, was correlated with symptoms, comorbidities, dyspnea, fatigue, stupor/coma, high neutrophil to lymphocyte ratio, C-reactive protein, interleukin-6, pro-calcitonin, and high oxygen need (p ≤ 0.001 for all). Among swab-positive staff, 3 had pneumonia and recovered. Although no comparison could be made between different treatment and prophylaxis strategies, potentially useful suggestions emerged. Mortality compared well with that of nursing homes of the same area not transformed into care centers. CONCLUSION Nursing homes massively hit by SARS-CoV-2 can become caring centers for patients not needing intensive care.
Collapse
Affiliation(s)
- Paolo Agostinis
- Department of Medicine, Ospedale Sant'Antonio Abate, Azienda Sanitaria Universitaria Friuli Centrale, Tolmezzo, Italy
| | - Valentina Vianello
- Department of General Surgery, Ospedale Santa Maria della Misericordia, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | | | - Giuseppe Caruso
- Special Continuity Care Units, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Beatrice Montessoro
- Special Continuity Care Units, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Marco Taurian
- Special Continuity Care Units, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Antonio Di Chiara
- Department of Medicine, Ospedale Sant'Antonio Abate, Azienda Sanitaria Universitaria Friuli Centrale, Tolmezzo, Italy
| | - Vincenzo Patruno
- Department of Pulmonology and Respiratory Medicine, Ospedale Santa Maria della Misericordia, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | | | - Gian Paolo Fadini
- Department of Medicine, Section of Diabetes and Metabolic Diseases, University of Padua, Padua, Italy
| | - Anna Aldovini
- Department of Medicine and Harvard Medical School, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Anna Baritussio
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Francesco Curcio
- Department of Medicine and Clinical Pathology Institute, University of Udine and Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Carlo Tascini
- Clinical Infectious Diseases, Ospedale Santa Maria Della Misericordia, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | | |
Collapse
|
24
|
Verma YK, Verma R, Tyagi N, Behl A, Kumar S, Gangenahalli GU. COVID-19 and its Therapeutics: Special Emphasis on Mesenchymal Stem Cells Based Therapy. Stem Cell Rev Rep 2021; 17:113-131. [PMID: 32920752 PMCID: PMC7486977 DOI: 10.1007/s12015-020-10037-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The novel virus, Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) caused the Corona Virus Disease-2019 (COVID-19) outbreak in Wuhan, Hubei province of China. This virus disseminated rapidly and reached to an unprecedented pandemic proportion in more than 213 nations with a large number of fatalities. The hypersecretion of pro-inflammatory cytokines is the main cause of mortality and morbidity due to COVID-19, therefore strategies that avert the cytokine storm may play a crucial role in abating the severity of COVID-19. This review highlights the minute details of SARS-CoV-2, its genomic organization, genomic variations within structural and non-structural proteins and viral progression mechanism in human beings. The approaches like antiviral strategies are discussed, including drugs that obstruct viral propagation and suppress the pro-inflammatory cytokines. This compilation emphasizes Mesenchymal Stem Cells (MSCs) based therapy alone or in combination with other therapeutics as an attractive curative approach for COVID-19 pandemic. The MSCs and its secretome, including antimicrobial peptides (AMPs) have various capabilities, for instance, immunomodulation, regeneration, antimicrobial properties, potential for attenuating the cytokine storm and bare minimum chances of being infected with SARS-CoV-2 virus. The immunomodulatory property of MSCs affects inflammatory state and regulates immune response during SARS-CoV-2 infection. However, as of now, there is no WHO-approved MSCs based therapy for the treatment of COVID-19 infection. Graphical abstract.
Collapse
Affiliation(s)
- Yogesh Kumar Verma
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences (INMAS), Brig. S.K. Mazumdar Road, Delhi, 110054, India
| | - Ranjan Verma
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences (INMAS), Brig. S.K. Mazumdar Road, Delhi, 110054, India
| | - Nishant Tyagi
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences (INMAS), Brig. S.K. Mazumdar Road, Delhi, 110054, India
| | - Amanpreet Behl
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences (INMAS), Brig. S.K. Mazumdar Road, Delhi, 110054, India
| | - Subodh Kumar
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences (INMAS), Brig. S.K. Mazumdar Road, Delhi, 110054, India
| | - Gurudutta U Gangenahalli
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences (INMAS), Brig. S.K. Mazumdar Road, Delhi, 110054, India.
| |
Collapse
|
25
|
Mészáros B, Sámano-Sánchez H, Alvarado-Valverde J, Čalyševa J, Martínez-Pérez E, Alves R, Shields DC, Kumar M, Rippmann F, Chemes LB, Gibson TJ. Short linear motif candidates in the cell entry system used by SARS-CoV-2 and their potential therapeutic implications. Sci Signal 2021; 14:eabd0334. [PMID: 33436497 PMCID: PMC7928535 DOI: 10.1126/scisignal.abd0334] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
The first reported receptor for SARS-CoV-2 on host cells was the angiotensin-converting enzyme 2 (ACE2). However, the viral spike protein also has an RGD motif, suggesting that cell surface integrins may be co-receptors. We examined the sequences of ACE2 and integrins with the Eukaryotic Linear Motif (ELM) resource and identified candidate short linear motifs (SLiMs) in their short, unstructured, cytosolic tails with potential roles in endocytosis, membrane dynamics, autophagy, cytoskeleton, and cell signaling. These SLiM candidates are highly conserved in vertebrates and may interact with the μ2 subunit of the endocytosis-associated AP2 adaptor complex, as well as with various protein domains (namely, I-BAR, LC3, PDZ, PTB, and SH2) found in human signaling and regulatory proteins. Several motifs overlap in the tail sequences, suggesting that they may act as molecular switches, such as in response to tyrosine phosphorylation status. Candidate LC3-interacting region (LIR) motifs are present in the tails of integrin β3 and ACE2, suggesting that these proteins could directly recruit autophagy components. Our findings identify several molecular links and testable hypotheses that could uncover mechanisms of SARS-CoV-2 attachment, entry, and replication against which it may be possible to develop host-directed therapies that dampen viral infection and disease progression. Several of these SLiMs have now been validated to mediate the predicted peptide interactions.
Collapse
Affiliation(s)
- Bálint Mészáros
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.
| | - Hugo Sámano-Sánchez
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Jesús Alvarado-Valverde
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences
| | - Jelena Čalyševa
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences
| | - Elizabeth Martínez-Pérez
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Laboratorio de bioinformática estructural, Fundación Instituto Leloir, C1405BWE Buenos Aires, Argentina
| | - Renato Alves
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Denis C Shields
- School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Manjeet Kumar
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.
| | - Friedrich Rippmann
- Computational Chemistry & Biology, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Lucía B Chemes
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", IIB-UNSAM, IIBIO-CONICET, Universidad Nacional de San Martín, CP1650 San Martín, Buenos Aires, Argentina.
| | - Toby J Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.
| |
Collapse
|
26
|
Pillaiyar T, Wendt LL, Manickam M, Easwaran M. The recent outbreaks of human coronaviruses: A medicinal chemistry perspective. Med Res Rev 2021; 41:72-135. [PMID: 32852058 PMCID: PMC7461420 DOI: 10.1002/med.21724] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/22/2020] [Accepted: 08/08/2020] [Indexed: 01/18/2023]
Abstract
Coronaviruses (CoVs) infect both humans and animals. In humans, CoVs can cause respiratory, kidney, heart, brain, and intestinal infections that can range from mild to lethal. Since the start of the 21st century, three β-coronaviruses have crossed the species barrier to infect humans: severe-acute respiratory syndrome (SARS)-CoV-1, Middle East respiratory syndrome (MERS)-CoV, and SARS-CoV-2 (2019-nCoV). These viruses are dangerous and can easily be transmitted from human to human. Therefore, the development of anticoronaviral therapies is urgently needed. However, to date, no approved vaccines or drugs against CoV infections are available. In this review, we focus on the medicinal chemistry efforts toward the development of antiviral agents against SARS-CoV-1, MERS-CoV, SARS-CoV-2, targeting biochemical events important for viral replication and its life cycle. These targets include the spike glycoprotein and its host-receptors for viral entry, proteases that are essential for cleaving polyproteins to produce functional proteins, and RNA-dependent RNA polymerase for viral RNA replication.
Collapse
Affiliation(s)
- Thanigaimalai Pillaiyar
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal ChemistryUniversity of BonnBonnGermany
| | - Lukas L. Wendt
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal ChemistryUniversity of BonnBonnGermany
| | - Manoj Manickam
- Department of ChemistryPSG Institute of Technology and Applied ResearchCoimbatoreTamil NaduIndia
| | - Maheswaran Easwaran
- Department of Biomedical EngineeringSethu Institute of TechnologyVirudhunagarTamilnaduIndia
| |
Collapse
|
27
|
Jiang B, Liang S, Liang G, Wei H. Could dantrolene be explored as a repurposed drug to treat COVID-19 patients by restoring intracellular calcium homeostasis? EUROPEAN REVIEW FOR MEDICAL AND PHARMACOLOGICAL SCIENCES 2020; 24:10228-10238. [PMID: 33090434 DOI: 10.26355/eurrev_202010_23247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dantrolene, an FDA approved drug to treat malignant hyperthermia and muscle spasm, has been demonstrated to inhibit severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mediated toxicity of host cells. Ryanodine receptor overactivation and associated disruption of intracellular Ca2+ homeostasis play important roles in SARS-CoV-2 infection and replication of host cells. Dantrolene, as an inhibitor of RyRs, is expected to ameliorate these detrimental effects of SARS-CoV-2 in host cells. Additionally, dantrolene has also been shown to inhibit multiple cell or organ damage induced by hypoxia/ischemia, mitochondria damage, oxidative stresses, inflammation, impairment of autophagy and apoptosis, etc., which are often the causes of severity and mortality of COVID-19 patients. We have repurposed that dantrolene has a high potential at treating COVID-19 patients and reducing its morbidity and mortality.
Collapse
Affiliation(s)
- B Jiang
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA.
| | | | | | | |
Collapse
|
28
|
Morris G, Athan E, Walder K, Bortolasci CC, O'Neil A, Marx W, Berk M, Carvalho AF, Maes M, Puri BK. Can endolysosomal deacidification and inhibition of autophagy prevent severe COVID-19? Life Sci 2020; 262:118541. [PMID: 33035581 PMCID: PMC7537668 DOI: 10.1016/j.lfs.2020.118541] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023]
Abstract
The possibility is examined that immunomodulatory pharmacotherapy may be clinically useful in managing the pandemic coronavirus disease 2019 (COVID-19), known to result from infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a positive-sense single-stranded RNA virus. The dominant route of cell entry of the coronavirus is via phagocytosis, with ensconcement in endosomes thereafter proceeding via the endosomal pathway, involving transfer from early (EEs) to late endosomes (LEs) and ultimately into lysosomes via endolysosomal fusion. EE to LE transportation is a rate-limiting step for coronaviruses. Hence inhibition or dysregulation of endosomal trafficking could potentially inhibit SARS-CoV-2 replication. Furthermore, the acidic luminal pH of the endolysosomal system is critical for the activity of numerous pH-sensitive hydrolytic enzymes. Golgi sub-compartments and Golgi-derived secretory vesicles also depend on being mildly acidic for optimal function and structure. Activation of endosomal toll-like receptors by viral RNA can upregulate inflammatory mediators and contribute to a systemic inflammatory cytokine storm, associated with a worsened clinical outcome in COVID-19. Such endosomal toll-like receptors could be inhibited by the use of pharmacological agents which increase endosomal pH, thereby reducing the activity of acid-dependent endosomal proteases required for their activity and/or assembly, leading to suppression of antigen-presenting cell activity, decreased autoantibody secretion, decreased nuclear factor-kappa B activity and decreased pro-inflammatory cytokine production. It is also noteworthy that SARS-CoV-2 inhibits autophagy, predisposing infected cells to apoptosis. It is therefore also suggested that further pharmacological inhibition of autophagy might encourage the apoptotic clearance of SARS-CoV-2-infected cells.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Eugene Athan
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Department of Infectious Disease, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Victoria, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Wolf Marx
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry, the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Maes
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | | |
Collapse
|
29
|
Korn D, Pervitsky V, Bobrowski T, Alves VM, Schmitt C, Bizon C, Baker N, Chirkova R, Cherkasov A, Muratov E, Tropsha A. COVID-19 Knowledge Extractor (COKE): A Tool and a Web Portal to Extract Drug - Target Protein Associations from the CORD-19 Corpus of Scientific Publications on COVID-19. CHEMRXIV : THE PREPRINT SERVER FOR CHEMISTRY 2020:13289222. [PMID: 33269341 PMCID: PMC7709174 DOI: 10.26434/chemrxiv.13289222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Revised: 11/26/2020] [Indexed: 12/02/2022]
Abstract
Objective: The COVID-19 pandemic has catalyzed a widespread effort to identify drug candidates and biological targets of relevance to SARS-COV-2 infection, which resulted in large numbers of publications on this subject. We have built the COVID-19 Knowledge Extractor (COKE), a web application to extract, curate, and annotate essential drug-target relationships from the research literature on COVID-19 to assist drug repurposing efforts. Materials and Methods: SciBiteAI ontological tagging of the COVID Open Research Dataset (CORD-19), a repository of COVID-19 scientific publications, was employed to identify drug-target relationships. Entity identifiers were resolved through lookup routines using UniProt and DrugBank. A custom algorithm was used to identify co-occurrences of protein and drug terms, and confidence scores were calculated for each entity pair. Results: COKE processing of the current CORD-19 database identified about 3,000 drug-protein pairs, including 29 unique proteins and 500 investigational, experimental, and approved drugs. Some of these drugs are presently undergoing clinical trials for COVID-19. Discussion: The rapidly evolving situation concerning the COVID-19 pandemic has resulted in a dramatic growth of publications on this subject in a short period. These circumstances call for methods that can condense the literature into the key concepts and relationships necessary for insights into SARS-CoV-2 drug repurposing. Conclusion: The COKE repository and web application deliver key drug - target protein relationships to researchers studying SARS-CoV-2. COKE portal may provide comprehensive and critical information on studies concerning drug repurposing against COVID-19. COKE is freely available at https://coke.mml.unc.edu/ and the code is available at https://github.com/DnlRKorn/CoKE.
Collapse
Affiliation(s)
- Daniel Korn
- Department of Computer Science, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Vera Pervitsky
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Tesia Bobrowski
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Vinicius M. Alves
- Office of Data Science, National Toxicology Program, NIEHS, Morrisville, NC, 27560, USA
| | - Charles Schmitt
- Office of Data Science, National Toxicology Program, NIEHS, Morrisville, NC, 27560, USA
| | - Chris Bizon
- Renaissance Computing Institute, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7568, USA
| | - Nancy Baker
- ParlezChem, 123 W Union Street, Hillsborough, NC, 27278, USA
| | - Rada Chirkova
- Department of Computer Science, North Carolina State University, Raleigh, NC, 27606-5550
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Eugene Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
30
|
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: 15.8] [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
|
31
|
Luquain-Costaz C, Rabia M, Hullin-Matsuda F, Delton I. Bis(monoacylglycero)phosphate, an important actor in the host endocytic machinery hijacked by SARS-CoV-2 and related viruses. Biochimie 2020; 179:247-256. [PMID: 33159981 PMCID: PMC7642752 DOI: 10.1016/j.biochi.2020.10.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
Viruses, including the novel coronavirus SARS-CoV-2, redirect infected cell metabolism to their own purposes. After binding to its receptor angiotensin-converting enzyme 2 (ACE2) on the cell surface, the SARS-CoV-2 is taken up by receptor-mediated endocytosis ending in the acidic endolysosomal compartment. The virus hijacks the endosomal machinery leading to fusion of viral and endosomal membranes and release of the viral RNA into the cytosol. This mini-review specifically highlights the membrane lipid organization of the endosomal system focusing on the unconventional and late endosome/lysosome-specific phospholipid, bis(monoacylglycero)phosphate (BMP). BMP is enriched in alveolar macrophages of lung, one of the target tissue of SARS-CoV-2. This review details the BMP structure, its unsaturated fatty acid composition and fusogenic properties that are essential for the highly dynamic formation of the intraluminal vesicles inside the endosomes. Interestingly, BMP is necessary for infection and replication of enveloped RNA virus such as SARS-CoV-1 and Dengue virus. We also emphasize the role of BMP in lipid sorting and degradation, especially cholesterol transport in cooperation with Niemann Pick type C proteins (NPC 1 and 2) and with some oxysterol-binding protein (OSBP)-related proteins (ORPs) as well as in sphingolipid degradation. Interestingly, numerous virus infection required NPC1 as well as ORPs along the endocytic pathway. Furthermore, BMP content is increased during pathological endosomal lipid accumulation in various lysosomal storage disorders. This is particularly important knowing the high percentage of patients with metabolic disorders among the SARS-CoV-2 infected patients presenting severe forms of COVID-19.
Collapse
Affiliation(s)
- Céline Luquain-Costaz
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Maxence Rabia
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | | | - Isabelle Delton
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France.
| |
Collapse
|
32
|
Dubey A, Dahiya S, Rouse BT, Sehrawat S. Perspective: Reducing SARS-CoV2 Infectivity and Its Associated Immunopathology. Front Immunol 2020; 11:581076. [PMID: 33193385 PMCID: PMC7642257 DOI: 10.3389/fimmu.2020.581076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022] Open
Abstract
COVID-19 has become difficult to contain in our interconnected world. In this article, we discuss some approaches that could reduce the consequences of COVID-19. We elaborate upon the utility of camelid single-domain antibodies (sdAbs), also referred to as nanobodies, which are naturally poised to neutralize viruses without enhancing its infectivity. Smaller sized sdAbs can be easily selected using microbes or the subcellular organelle display methods and can neutralize SARS-CoV2 infectivity. We also discuss issues related to their production using scalable platforms. The favorable outcome of the infection is evident in patients when the inflammatory response is adequately curtailed. Therefore, we discuss approaches to mitigate hyperinflammatory reactions initiated by SARS-CoV2 but orchestrated by immune mediators.
Collapse
Affiliation(s)
- Abhishek Dubey
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Surbhi Dahiya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Barry T. Rouse
- Biomedical and Diagnostic Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN, United States
| | - Sharvan Sehrawat
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| |
Collapse
|
33
|
Sabarimurugan S, Dharmarajan A, Warrier S, Subramanian M, Swaminathan R. Comprehensive review on the prevailing COVID-19 therapeutics and the potential of repurposing SARS-CoV-1 candidate drugs to target SARS-CoV-2 as a fast-track treatment and prevention option. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1247. [PMID: 33178779 PMCID: PMC7607133 DOI: 10.21037/atm-20-4071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The recent seemingly uncontrollable pandemic caused by the novel severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) has been able to spread quickly due to the non-availability of effective antivirals or vaccines. The virus has structural and non-structural proteins that are considered as possible targets. Receptor recognition is the critical determinant and preliminary phase of viral infection to enter the host cell and causes tissue tropism. We have conducted a comprehensive review of relevant publication on in vitro, in silico, in vivo and clinical evaluation of drug candidates ranging from broad-spectrum antivirals to natural molecules targeted towards viral spike protein in addition to evaluate their suitability as therapies based on an analysis of the similarities between SARS-CoV-1 and SARS-CoV-2. In general, antiviral targets are based on two strategies, either targeting the host or the host’s immune cell. We have reviewed the available details on the SARS-CoV-2 strain’s host-viral binding sites entry mechanism, alongside recently tested effective antivirals. The hypothesis of this review may provide clear insight for researchers and physicians who are struggling to narrow down scientific options to control the current pandemic. Overall, we found that the promising efficacious drug candidates reported against SARS-CoV-1 could be considered for drug repurposing; this might help to identify a potential drug for therapeutic measures and development of vaccine for COVID-19.
Collapse
Affiliation(s)
- Shanthi Sabarimurugan
- School of Biomedical Sciences, The University of Western Australia, Perth, Australia
| | - Arun Dharmarajan
- CHIRI, School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA, Australia.,Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, India.,Department of Human Sciences, Faculty of Life Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Sudha Warrier
- CHIRI, School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA, Australia.,School of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore, India
| | - Maheswari Subramanian
- Department of Microbiology, Centre for Research and PG Studies, Indian Academy of Degree College (Aut), Bangalore, India
| | - Rajarajan Swaminathan
- Centre for Drug Design, Discovery and Development, SRM University, Sonipat, Haryana, New Delhi, India
| |
Collapse
|
34
|
Sanchis-Gomar F, Lavie CJ, Morin DP, Perez-Quilis C, Laukkanen JA, Perez MV. Amiodarone in the COVID-19 Era: Treatment for Symptomatic Patients Only, or Drug to Prevent Infection? Am J Cardiovasc Drugs 2020; 20:413-418. [PMID: 32737841 PMCID: PMC7394926 DOI: 10.1007/s40256-020-00429-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Amiodarone, one of the most widely prescribed antiarrhythmic drugs to treat both ventricular and supraventricular arrhythmias, has been identified as a candidate drug for use against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We present the rationale of using amiodarone in the COVID-19 scenario, as well as whether or not amiodarone administration represents a potential strategy to prevent SARS-CoV-2 infection, rather than simply used to treat patients already symptomatic and/or with severe coronavirus disease 2019 (COVID-19), based on current evidence.
Collapse
Affiliation(s)
- Fabian Sanchis-Gomar
- Department of Physiology, Faculty of Medicine, INCLIVA Biomedical Research Institute, University of Valencia, Av. Blasco Ibáñez, 15, 46010, Valencia, Spain.
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School, The University of Queensland School of Medicine, New Orleans, LA, USA
| | - Daniel P Morin
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School, The University of Queensland School of Medicine, New Orleans, LA, USA
| | - Carme Perez-Quilis
- Department of Physiology, Faculty of Medicine, INCLIVA Biomedical Research Institute, University of Valencia, Av. Blasco Ibáñez, 15, 46010, Valencia, Spain
| | - Jari A Laukkanen
- Institute of Clinical Medicine, Department of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Central Finland Health Care District, Jyvaskyla, Finland
| | - Marco V Perez
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
35
|
Vallamkondu J, John A, Wani WY, Ramadevi SP, Jella KK, Reddy PH, Kandimalla R. SARS-CoV-2 pathophysiology and assessment of coronaviruses in CNS diseases with a focus on therapeutic targets. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165889. [PMID: 32603829 PMCID: PMC7320676 DOI: 10.1016/j.bbadis.2020.165889] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 02/06/2023]
Abstract
The novel Coronavirus disease of 2019 (nCOV-19) is a viral outbreak noted first in Wuhan, China. This disease is caused by Severe Acute Respiratory Syndrome (SARS) Coronavirus (CoV)-2. In the past, other members of the coronavirus family, such as SARS and Middle East Respiratory Syndrome (MERS), have made an impact in China and the Arabian peninsula respectively. Both SARS and COVID-19 share similar symptoms such as fever, cough, and difficulty in breathing that can become fatal in later stages. However, SARS and MERS infections were epidemic diseases constrained to limited regions. By March 2020 the SARS-CoV-2 had spread across the globe and on March 11th, 2020 the World Health Organization (WHO) declared COVID-19 as pandemic disease. In severe SARS-CoV-2 infection, many patients succumbed to pneumonia. Higher rates of deaths were seen in older patients who had co-morbidities such as diabetes mellitus, hypertension, cardiovascular disease (CVD), and dementia. In this review paper, we discuss the effect of SARS-CoV-2 on CNS diseases, such as Alzheimer's-like dementia, and diabetes mellitus. We also focus on the virus genome, pathophysiology, theranostics, and autophagy mechanisms. We will assess the multiorgan failure reported in advanced stages of SARS-CoV-2 infection. Our paper will provide mechanistic clues and therapeutic targets for physicians and investigators to combat COVID-19.
Collapse
Affiliation(s)
| | - Albin John
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Willayat Yousuf Wani
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | | | | | - P Hemachandra Reddy
- Professor of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Internal Medicine, Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, Telangana, India; Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India.
| |
Collapse
|
36
|
Beeraka NM, Sadhu SP, Madhunapantula SV, Rao Pragada R, Svistunov AA, Nikolenko VN, Mikhaleva LM, Aliev G. Strategies for Targeting SARS CoV-2: Small Molecule Inhibitors-The Current Status. Front Immunol 2020; 11:552925. [PMID: 33072093 PMCID: PMC7531039 DOI: 10.3389/fimmu.2020.552925] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/18/2020] [Indexed: 01/08/2023] Open
Abstract
Severe Acute Respiratory Syndrome-Corona Virus-2 (SARS-CoV-2) induced Coronavirus Disease - 19 (COVID-19) cases have been increasing at an alarming rate (7.4 million positive cases as on June 11 2020), causing high mortality (4,17,956 deaths as on June 11 2020) and economic loss (a 3.2% shrink in global economy in 2020) across 212 countries globally. The clinical manifestations of this disease are pneumonia, lung injury, inflammation, and severe acute respiratory syndrome (SARS). Currently, there is no vaccine or effective pharmacological agents available for the prevention/treatment of SARS-CoV2 infections. Moreover, development of a suitable vaccine is a challenging task due to antibody-dependent enhancement (ADE) and Th-2 immunopathology, which aggravates infection with SARS-CoV-2. Furthermore, the emerging SARS-CoV-2 strain exhibits several distinct genomic and structural patterns compared to other coronavirus strains, making the development of a suitable vaccine even more difficult. Therefore, the identification of novel small molecule inhibitors (NSMIs) that can interfere with viral entry or viral propagation is of special interest and is vital in managing already infected cases. SARS-CoV-2 infection is mediated by the binding of viral Spike proteins (S-protein) to human cells through a 2-step process, which involves Angiotensin Converting Enzyme-2 (ACE2) and Transmembrane Serine Protease (TMPRSS)-2. Therefore, the development of novel inhibitors of ACE2/TMPRSS2 is likely to be beneficial in combating SARS-CoV-2 infections. However, the usage of ACE-2 inhibitors to block the SARS-CoV-2 viral entry requires additional studies as there are conflicting findings and severe health complications reported for these inhibitors in patients. Hence, the current interest is shifted toward the development of NSMIs, which includes natural antiviral phytochemicals and Nrf-2 activators to manage a SARS-CoV-2 infection. It is imperative to investigate the efficacy of existing antiviral phytochemicals and Nrf-2 activators to mitigate the SARS-CoV-2-mediated oxidative stress. Therefore, in this review, we have reviewed structural features of SARS-CoV-2 with special emphasis on key molecular targets and their known modulators that can be considered for the development of NSMIs.
Collapse
Affiliation(s)
- Narasimha M. Beeraka
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), JSS Academy of Higher Education & Research (JSS AHER), Mysore, India
| | - Surya P. Sadhu
- AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, India
| | - SubbaRao V. Madhunapantula
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), JSS Academy of Higher Education & Research (JSS AHER), Mysore, India
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore, India
| | | | - Andrey A. Svistunov
- I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Vladimir N. Nikolenko
- I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
- Department of Normal and Topographic Anatomy, M.V. Lomonosov Moscow State University, Moscow, Russia
| | | | - Gjumrakch Aliev
- Research Institute of Human Morphology, Moscow, Russia
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Moscow, Russia
- GALLY International Research Institute, San Antonio, TX, United States
| |
Collapse
|
37
|
Karamchandani K, Quintili A, Landis T, Bose S. Cardiac Arrhythmias in Critically Ill Patients With COVID-19: A Brief Review. J Cardiothorac Vasc Anesth 2020; 35:3789-3796. [PMID: 32888796 PMCID: PMC7418708 DOI: 10.1053/j.jvca.2020.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019, caused by severe acute respiratory syndrome coronavirus 2, is now a global pandemic affecting more than 12 million patients across 188 countries. A significant proportion of these patients require admission to intensive care units for acute hypoxic respiratory failure and are at an increased risk of developing cardiac arrhythmias. The presence of underlying comorbidities, pathophysiologic changes imposed by the disease, and concomitant polypharmacy, increase the likelihood of life-threatening arrhythmias in these patients. Supraventricular, as well as ventricular arrhythmias, are common and are associated with significant morbidity and mortality. It is important to understand the interplay of various causal factors while instituting strategies to mitigate the impact of modifiable risk factors. Furthermore, avoidance and early recognition of drug interactions, along with prompt treatment, might help improve outcomes in this vulnerable patient population.
Collapse
Affiliation(s)
- Kunal Karamchandani
- Department of Anesthesiology and Perioperative Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA.
| | - Ashley Quintili
- Department of Pharmacy, Penn State Health Milton S. Hershey Medical Center, Hershey, PA
| | - Terra Landis
- Department of Pharmacy, Penn State Health Milton S. Hershey Medical Center, Hershey, PA
| | - Somnath Bose
- Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
38
|
Su YB, Kuo MJ, Lin TY, Chien CS, Yang YP, Chou SJ, Leu HB. Cardiovascular manifestation and treatment in COVID-19. J Chin Med Assoc 2020; 83:704-709. [PMID: 32433342 PMCID: PMC7493780 DOI: 10.1097/jcma.0000000000000352] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
The novel coronavirus disease 2019 (COVID-19), with first presentation of atypical pneumonia, has spread rapidly from Wuhan, China, on December 12, 2019 to over 200 countries, caused 2 310 572 infected individuals and 158 691 mortalities, updated on April 19, 2020. Many studies have published timely to help global healthcare workers to understand and control the disease. Vulnerable patients with risk factors such as elderly, cardiovascular diseases (eg, hypertension, coronary disease, or cardiomyopathy), diabetes, and chronic kidney disease have worse outcomes after COVID-19 infection. COVID-19 could directly cause cardiovascular injuries such as pericarditis, myocarditis, myocardial infarction, heart failure, arrhythmias, or thromboembolic events, which urge cardiologists to be involved in the frontline to practice. Here, we provide a review of COVID-19 on cardiovascular system to assist clinical cardiologists to better understand the disease and being capable of providing comprehensive medical support.
Collapse
Affiliation(s)
- Yen-Bo Su
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ming-Jen Kuo
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ting-Yu Lin
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Chian-Shiu Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Shih-Jie Chou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Hsin-Bang Leu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan, ROC
- Healthcare and Service Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
39
|
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: 8.0] [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
|
40
|
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: 3.0] [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
|
41
|
Mitra P. Inhibiting fusion with cellular membrane system: therapeutic options to prevent severe acute respiratory syndrome coronavirus-2 infection. Am J Physiol Cell Physiol 2020; 319:C500-C509. [PMID: 32687406 PMCID: PMC7839238 DOI: 10.1152/ajpcell.00260.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV), an enveloped virus with a positive-sense single-stranded RNA genome, facilitates the host cell entry through intricate interactions with proteins and lipids of the cell membrane. The detailed molecular mechanism involves binding to the host cell receptor and fusion at the plasma membrane or after being trafficked to late endosomes under favorable environmental conditions. A crucial event in the process is the proteolytic cleavage of the viral spike protein by the host's endogenous proteases that releases the fusion peptide enabling fusion with the host cellular membrane system. The present review details the mechanism of viral fusion with the host and highlights the therapeutic options that prevent SARS-CoV-2 entry in humans.
Collapse
Affiliation(s)
- Prasenjit Mitra
- Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, Telangana, India
| |
Collapse
|
42
|
Rabia M, Leuzy V, Soulage C, Durand A, Fourmaux B, Errazuriz-Cerda E, Köffel R, Draeger A, Colosetti P, Jalabert A, Di Filippo M, Villard-Garon A, Bergerot C, Luquain-Costaz C, Moulin P, Rome S, Delton I, Hullin-Matsuda F. Bis(monoacylglycero)phosphate, a new lipid signature of endosome-derived extracellular vesicles. Biochimie 2020; 178:26-38. [PMID: 32659447 DOI: 10.1016/j.biochi.2020.07.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/29/2020] [Accepted: 07/08/2020] [Indexed: 01/05/2023]
Abstract
Bis(monoacylglycero)phosphate (BMP), also known as lysobisphosphatidic acid (LBPA), is a phospholipid specifically enriched in the late endosome-lysosome compartment playing a crucial role for the fate of endocytosed components. Due to its presence in extracellular fluids during diseases associated with endolysosomal dysfunction, it is considered as a possible biomarker of disorders such as genetic lysosomal storage diseases and cationic amphiphilic drug-induced phospholipidosis. However, there is no true validation of this biomarker in human studies, nor a clear identification of the carrier of this endolysosome-specific lipid in biofluids. The present study demonstrates that in absence of any sign of renal failure, BMP, especially all docosahexaenoyl containing species, are significantly increased in the urine of patients treated with the antiarrhythmic drug amiodarone. Such urinary BMP increase could reflect a generalized drug-induced perturbation of the endolysosome compartment as observed in vitro with amiodarone-treated human macrophages. Noteworthy, BMP was associated with extracellular vesicles (EVs) isolated from human urines and extracellular medium of human embryonic kidney HEK293 cells and co-localizing with classical EV protein markers CD63 and ALIX. In the context of drug-induced endolysosomal dysfunction, increased BMP-rich EV release could be useful to remove excess of undigested material. This first human pilot study not only reveals BMP as a urinary biomarker of amiodarone-induced endolysosomal dysfunction, but also highlights its utility to prove the endosomal origin of EVs, also named as exosomes. This peculiar lipid already known as a canonical late endosome-lysosome marker, may be thus considered as a new lipid marker of urinary exosomes.
Collapse
Affiliation(s)
- Maxence Rabia
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Valentin Leuzy
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Christophe Soulage
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Annie Durand
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Baptiste Fourmaux
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France; Functional Lipidomics Platform, CarMeN Laboratory / IMBL-INSA Lyon, 69621, Villeurbanne Cedex, France
| | | | - René Köffel
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Annette Draeger
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Pascal Colosetti
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Audrey Jalabert
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Mathilde Di Filippo
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France; Department of Biochemistry and Molecular Biology, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Bron, France
| | - Audrey Villard-Garon
- Department of Endocrinology, Hôpital Cardiovasculaire Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | - Cyrille Bergerot
- Department of Cardiology, Hôpital Cardiovasculaire Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | - Céline Luquain-Costaz
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Philippe Moulin
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France; Department of Endocrinology, Hôpital Cardiovasculaire Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | - Sophie Rome
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Isabelle Delton
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | | |
Collapse
|
43
|
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: 1.8] [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
|
44
|
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: 0.3] [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
|
45
|
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: 9.0] [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
|
46
|
SARS-CoV-2: An Update on Potential Antivirals in Light of SARS-CoV Antiviral Drug Discoveries. Vaccines (Basel) 2020; 8:vaccines8020335. [PMID: 32585913 PMCID: PMC7350231 DOI: 10.3390/vaccines8020335] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/04/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Coronaviruses (CoVs) are a group of RNA viruses that are associated with different diseases in animals, birds, and humans. Human CoVs (HCoVs) have long been known to be the causative agents of mild respiratory illnesses. However, two HCoVs associated with severe respiratory diseases are Severe Acute Respiratory Syndrome-CoV (SARS-CoV) and Middle East Respiratory Syndrome-CoV (MERS-CoV). Both viruses resulted in hundreds of deaths after spreading to several countries. Most recently, SARS-CoV-2 has emerged as the third HCoV causing severe respiratory distress syndrome and viral pneumonia (known as COVID-19) in patients from Wuhan, China, in December 2019. Soon after its discovery, SARS-CoV-2 spread to all countries, resulting in millions of cases and thousands of deaths. Since the emergence of SARS-CoV, many research groups have dedicated their resources to discovering effective antivirals that can treat such life-threatening infections. The rapid spread and high fatality rate of SARS-CoV-2 necessitate the quick discovery of effective antivirals to control this outbreak. Since SARS-CoV-2 shares 79% sequence identity with SARS-CoV, several anti-SARS-CoV drugs have shown promise in limiting SARS-CoV-2 replication in vitro and in vivo. In this review, we discuss antivirals described for SARS-CoV and provide an update on therapeutic strategies and antivirals against SARS-CoV-2. The control of the current outbreak will strongly depend on the discovery of effective and safe anti-SARS-CoV-2 drugs.
Collapse
|
47
|
Dinis-Oliveira RJ. COVID-19 research: pandemic versus "paperdemic", integrity, values and risks of the "speed science". Forensic Sci Res 2020; 5:174-187. [PMID: 32939434 PMCID: PMC7476615 DOI: 10.1080/20961790.2020.1767754] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 12/23/2022] Open
Abstract
Scientific integrity is a learned skill. When researchers and students learn integrity in laboratories or in the classroom, they are empowered to use similar principles in other aspects of their lives. This commentary reviews the concepts related to scientific integrity at a time when science faces important challenges related to the increase number of articles produced regarding research on coronavirus disease 2019 (COVID-19). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has ignited another parallel viral pandemic, with science ranging from robust studies to dishonest studies being conducted, posted, and shared at an unprecedented rate. A balance is needed between the benefits of the rapid access to new scientific data and the threat of causing panic or erroneous clinical decisions based on mistakes or misconduct. The truth is that the "scientific research has changed the world" but now, and more than ever, "it needs to change itself". A pandemic with a "paperdemic" will be even more complicated to manage if it progresses in an uncontrolled manner and is not properly scrutinized.
Collapse
Affiliation(s)
- Ricardo Jorge Dinis-Oliveira
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Sciences, IINFACTS-Institute of Research and Advanced Training in Health Sciences and Technologies, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
48
|
Pericàs JM, Hernandez-Meneses M, Sheahan TP, Quintana E, Ambrosioni J, Sandoval E, Falces C, Marcos MA, Tuset M, Vilella A, Moreno A, Miro JM. COVID-19: from epidemiology to treatment. Eur Heart J 2020; 41:2092-2112. [PMID: 32511724 PMCID: PMC7279517 DOI: 10.1093/eurheartj/ehaa462] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/21/2020] [Accepted: 05/14/2020] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has greatly impacted the daily clinical practice of cardiologists and cardiovascular surgeons. Preparedness of health workers and health services is crucial to tackle the enormous challenge posed by SARS-CoV-2 in wards, operating theatres, intensive care units, and interventionist laboratories. This Clinical Review provides an overview of COVID-19 and focuses on relevant aspects on prevention and management for specialists within the cardiovascular field.
Collapse
Affiliation(s)
- J M Pericàs
- Infectious Diseases Department, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - M Hernandez-Meneses
- Infectious Diseases Department, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - T P Sheahan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - E Quintana
- Cardiovascular Surgery Department, Hospital Clinic-IDIBAPS, Barcelona, Spain
| | - J Ambrosioni
- Infectious Diseases Department, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - E Sandoval
- Cardiovascular Surgery Department, Hospital Clinic-IDIBAPS, Barcelona, Spain
| | - C Falces
- Cardiology Department, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - M A Marcos
- Microbiology Service, Hospital Clinic-ISGlobal, University of Barcelona, Barcelona, Spain
| | - M Tuset
- Farmacy Department, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - A Vilella
- Preventive Medicine Service, Hospital Clinic-ISGlobal, University of Barcelona, Barcelona, Spain
| | - A Moreno
- Infectious Diseases Department, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - J M Miro
- Infectious Diseases Department, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| |
Collapse
|
49
|
Ghosh AK, Brindisi M, Shahabi D, Chapman ME, Mesecar AD. Drug Development and Medicinal Chemistry Efforts toward SARS-Coronavirus and Covid-19 Therapeutics. ChemMedChem 2020; 15:907-932. [PMID: 32324951 PMCID: PMC7264561 DOI: 10.1002/cmdc.202000223] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 infection is spreading at an alarming rate and has created an unprecedented health emergency around the globe. There is no effective vaccine or approved drug treatment against COVID-19 and other pathogenic coronaviruses. The development of antiviral agents is an urgent priority. Biochemical events critical to the coronavirus replication cycle provided a number of attractive targets for drug development. These include, spike protein for binding to host cell-surface receptors, proteolytic enzymes that are essential for processing polyproteins into mature viruses, and RNA-dependent RNA polymerase for RNA replication. There has been a lot of ground work for drug discovery and development against these targets. Also, high-throughput screening efforts have led to the identification of diverse lead structures, including natural product-derived molecules. This review highlights past and present drug discovery and medicinal-chemistry approaches against SARS-CoV, MERS-CoV and COVID-19 targets. The review hopes to stimulate further research and will be a useful guide to the development of effective therapies against COVID-19 and other pathogenic coronaviruses.
Collapse
Affiliation(s)
- Arun K. Ghosh
- Department of ChemistryPurdue UniversityWest LafayetteIN 47907USA
- Department of Medicinal Chemistry and Molecular PharmacolgyPurdue UniversityWest LafayetteIN 47907USA
| | - Margherita Brindisi
- Department of ChemistryPurdue UniversityWest LafayetteIN 47907USA
- Department of Excellence of PharmacyUniversity of Naples Federico II80131NaplesItaly
| | - Dana Shahabi
- Department of ChemistryPurdue UniversityWest LafayetteIN 47907USA
| | | | - Andrew D. Mesecar
- Department of ChemistryPurdue UniversityWest LafayetteIN 47907USA
- Department of BiochemistryPurdue UniversityWest LafayetteIN 47907USA
- Department of Biological SciencesPurdue UniversityWest LafayetteIN 47907USA
| |
Collapse
|
50
|
Ky B, Mann DL. COVID-19 Clinical Trials: A Primer for the Cardiovascular and Cardio-Oncology Communities. JACC CardioOncol 2020; 2:254-269. [PMID: 32313885 PMCID: PMC7164888 DOI: 10.1016/j.jaccao.2020.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease-2019 (COVID-19) pandemic has resulted in a proliferation of clinical trials designed to slow the spread of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). Many therapeutic agents that are being used to treat patients with COVID-19 are repurposed treatments for influenza, Ebola, or for malaria that were developed decades ago and are unlikely to be familiar to the cardiovascular and cardio-oncology communities. Here, we provide a foundation for cardiovascular and cardio-oncology physicians on the front line providing care to patients with COVID-19, so that they may better understand the emerging cardiovascular epidemiology and the biological rationale for the clinical trials that are ongoing for the treatment of patients with COVID-19.
Collapse
Key Words
- ACE, angiotensin-converting enzyme
- ACE2
- AT1R, angiotensin II type 1 receptor
- CI, confidence interval
- COVID-19
- COVID-19, coronavirus disease-2019
- CoV, coronavirus
- FDA, Food and Drug Administration
- IFN, interferon
- IL, interleukin
- IQR, interquartile range
- MERS, Middle East respiratory syndrome
- RAS, renin-angiotensin system
- RNA, ribonucleic acid
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory syndrome-coronavirus-2
- TMPRSS2, transmembrane protease serine 2
- clinical trials
- renin angiotensin system
- sACE2, soluble angiotensin-converting enzyme 2
Collapse
Affiliation(s)
- Bonnie Ky
- Department of Medicine, Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Douglas L. Mann
- Department of Medicine, Division of Cardiology, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|