1
|
Menacer R, Bouchekioua S, Meliani S, Belattar N. New combined Inverse-QSAR and molecular docking method for scaffold-based drug discovery. Comput Biol Med 2024; 180:108992. [PMID: 39128176 DOI: 10.1016/j.compbiomed.2024.108992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/14/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
Computer-aided drug discovery plays a vital role in developing novel medications for various diseases. The COVID-19 pandemic has heightened the need for innovative approaches to design lead compounds with the potential to become effective drugs. Specifically, designing promising inhibitors of the SARS-CoV-2 main protease (Mpro) is crucial, as it plays a key role in viral replication. Phytochemicals, primarily flavonoids and flavonols from medicinal plants, were screened. Fifty small molecules were selected for molecular docking analysis against SARS-CoV-2 Mpro (PDB ID: 6LU7). Binding energies and interactions were analyzed and compared to those of the anti-SARS-CoV-2 inhibitor Nirmatrelvir. Using these 50 structures as a training set, a QSAR model was built employing simple, reversible topological descriptors. An inverse-QSAR analysis was then performed on 2⁹ = 512 hydroxyl combinations at nine possible positions on the flavone and flavonol scaffold. The model predicted three novel, promising compounds exhibiting the most favorable binding energies (-8.5 kcal/mol) among the 512 possible hydroxyl combinations: 3,6,7,2',4'-pentahydroxyflavone (PF9), 6,7,2',4'-tetrahydroxyflavone (PF11), and 3,6,7,4'-tetrahydroxyflavone (PF15). Molecular dynamics (MD) simulations demonstrated the stability of the PF9/Mpro complex over 300 ns of simulation. These predicted structures, reported here for the first time, warrant synthesis and further evaluation of their biological activity through in vitro and in vivo studies.
Collapse
Affiliation(s)
- Rafik Menacer
- Centre de Recherche en Sciences Pharmaceutiques, Constantine, 25000, Algeria; Centre de Recherche Scientifique et Technique en Analyses Physico-Chimiques CRAPC, BP 384, Zone Industrielle, Bou-ismail, Tipaza, RP, 42004, Algeria.
| | - Saad Bouchekioua
- Centre de Recherche en Sciences Pharmaceutiques, Constantine, 25000, Algeria
| | - Saida Meliani
- Centre de Recherche en Sciences Pharmaceutiques, Constantine, 25000, Algeria
| | - Nadjah Belattar
- Centre de Recherche en Sciences Pharmaceutiques, Constantine, 25000, Algeria
| |
Collapse
|
2
|
Dai R, Gao H, Su R. Computer-aided drug design for virtual-screening and active-predicting of main protease (M pro) inhibitors against SARS-CoV-2. Front Pharmacol 2023; 14:1288363. [PMID: 38026989 PMCID: PMC10661973 DOI: 10.3389/fphar.2023.1288363] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: SARS-CoV-2 is a novel coronavirus with highly contagious and has posed a significant threat to global public health. The main protease (Mpro) is a promising target for antiviral drugs against SARS-CoV-2. Methods: In this study, we have used pharmacophore-based drug design technology to identify potential compounds from drug databases as Mpro inhibitors. Results: The procedure involves pharmacophore modeling, validation, and pharmacophore-based virtual screening, which identifies 257 compounds with promising inhibitory activity. Discussion: Molecular docking and non-bonding interactions between the targeted protein Mpro and compounds showed that ENA482732 was the best compound. These results provided a theoretical foundation for future studies of Mpro inhibitors against SARS-CoV-2.
Collapse
Affiliation(s)
| | - Hongwei Gao
- School of Life Science, Ludong University, Yantai, Shandong, China
| | | |
Collapse
|
3
|
Raza A, Chohan TA, Buabeid M, Arafa ESA, Chohan TA, Fatima B, Sultana K, Ullah MS, Murtaza G. Deep learning in drug discovery: a futuristic modality to materialize the large datasets for cheminformatics. J Biomol Struct Dyn 2023; 41:9177-9192. [PMID: 36305195 DOI: 10.1080/07391102.2022.2136244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/08/2022] [Indexed: 10/31/2022]
Abstract
Artificial intelligence (AI) development imitates the workings of the human brain to comprehend modern problems. The traditional approaches such as high throughput screening (HTS) and combinatorial chemistry are lengthy and expensive to the pharmaceutical industry as they can only handle a smaller dataset. Deep learning (DL) is a sophisticated AI method that uses a thorough comprehension of particular systems. The pharmaceutical industry is now adopting DL techniques to enhance the research and development process. Multi-oriented algorithms play a crucial role in the processing of QSAR analysis, de novo drug design, ADME evaluation, physicochemical analysis, preclinical development, followed by clinical trial data precision. In this study, we investigated the performance of several algorithms, including deep neural networks (DNN), convolutional neural networks (CNN) and multi-task learning (MTL), with the aim of generating high-quality, interpretable big and diverse databases for drug design and development. Studies have demonstrated that CNN, recurrent neural network and deep belief network are compatible, accurate and effective for the molecular description of pharmacodynamic properties. In Covid-19, existing pharmacological compounds has also been repurposed using DL models. In the absence of the Covid-19 vaccine, remdesivir and oseltamivir have been widely employed to treat severe SARS-CoV-2 infections. In conclusion, the results indicate the potential benefits of employing the DL strategies in the drug discovery process.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ali Raza
- Department of pharmaceutical chemistry, Faculty of Pharmacy, The University of Lahore, Pakistan
- Institute of Molecular Biology and Biochemistry, The University of Lahore, Pakistan
| | - Talha Ali Chohan
- Institute of Molecular Biology and Biochemistry, The University of Lahore, Pakistan
- Institute of Pharmaceutical Science, UVAS, Lahore, Pakistan
| | - Manal Buabeid
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - El-Shaima A Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | | | - Batool Fatima
- Department of biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Kishwar Sultana
- Department of pharmaceutical chemistry, Faculty of Pharmacy, The University of Lahore, Pakistan
| | - Malik Saad Ullah
- Department of Pharmacy, Government College University, Faisalabad, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Pakistan
| |
Collapse
|
4
|
Toigo L, Dos Santos Teodoro EI, Guidi AC, Gancedo NC, Petruco MV, Melo EB, Tonin FS, Fernandez-Llimos F, Chierrito D, de Mello JCP, de Medeiros Araújo DC, Sanches ACC. Flavonoid as possible therapeutic targets against COVID-19: a scoping review of in silico studies. Daru 2023; 31:51-68. [PMID: 37195402 PMCID: PMC10191091 DOI: 10.1007/s40199-023-00461-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/25/2023] [Indexed: 05/18/2023] Open
Abstract
OBJECTIVES This scoping review aims to present flavonoid compounds' promising effects and possible mechanisms of action on potential therapeutic targets in the SARS-CoV-2 infection process. METHODS A search of electronic databases such as PubMed and Scopus was carried out to evaluate the performance of substances from the flavonoid class at different stages of SARS-CoV-2 infection. RESULTS The search strategy yielded 382 articles after the exclusion of duplicates. During the screening process, 265 records were deemed as irrelevant. At the end of the full-text appraisal, 37 studies were considered eligible for data extraction and qualitative synthesis. All the studies used virtual molecular docking models to verify the affinity of compounds from the flavonoid class with crucial proteins in the replication cycle of the SARS-CoV-2 virus (Spike protein, PLpro, 3CLpro/ MPro, RdRP, and inhibition of the host's ACE II receptor). The flavonoids with more targets and lowest binding energies were: orientin, quercetin, epigallocatechin, narcissoside, silymarin, neohesperidin, delphinidin-3,5-diglucoside, and delphinidin-3-sambubioside-5-glucoside. CONCLUSION These studies allow us to provide a basis for in vitro and in vivo assays to assist in developing drugs for the treatment and prevention of COVID-19.
Collapse
Affiliation(s)
- Larissa Toigo
- Centro de Ciências Médicas e Farmacêuticas, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
| | | | - Ana Carolina Guidi
- Laboratório de Biologia Farmacêutica, Departamento de Farmácia, Universidade Estadual de Maringá, Maringá, Brazil
| | - Naiara Cássia Gancedo
- Laboratório de Biologia Farmacêutica, Departamento de Farmácia, Universidade Estadual de Maringá, Maringá, Brazil
| | - Marcus Vinícius Petruco
- Clínica de Reumatologia-Pneumologia Laboratório do Sono de Maringá e Hospital Bom Samaritano de Maringá, Maringá, Brazil
| | - Eduardo Borges Melo
- Centro de Ciências Médicas e Farmacêuticas, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
| | - Fernanda Stumpf Tonin
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Federal do Paraná, Curitiba, Brazil
- H&TRC- Health & Technology Research Center, ESTeSLEscola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisboa, Portugal
| | | | - Danielly Chierrito
- Centro de Ciências Médicas e Farmacêuticas, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
- Centro Universitário Ingá - UNINGÁ, Maringá, Brazil
| | - João Carlos Palazzo de Mello
- Laboratório de Biologia Farmacêutica, Departamento de Farmácia, Universidade Estadual de Maringá, Maringá, Brazil
| | | | | |
Collapse
|
5
|
Sarkar S, Gupta VK, Sharma S, Shen T, Gupta V, Mirzaei M, Graham SL, Chitranshi N. Computational refinement identifies functional destructive single nucleotide polymorphisms associated with human retinoid X receptor gene. J Biomol Struct Dyn 2023; 41:1458-1478. [PMID: 34971346 DOI: 10.1080/07391102.2021.2021991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Alterations in the nuclear retinoid X receptor (RXRs) signalling have been implicated in neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, stroke, multiple sclerosis and glaucoma. Single nucleotide polymorphisms (SNPs) are the main cause underlying single nucleic acid variations which in turn determine heterogeneity within various populations. These genetic polymorphisms have been suggested to associate with various degenerative disorders in population-wide analysis. This bioinformatics study was designed to investigate, search, retrieve and identify deleterious SNPs which may affect the structure and function of various RXR isoforms through a computational and molecular modelling approach. Amongst the 1,813 retrieved SNPs several were found to be deleterious with rs140464195_G139R, rs368400425_R358W and rs368586400_L383F RXRα mutant variants being the most detrimental ones causing changes in the interatomic interactions and decreasing the flexibility of the mutant proteins. Molecular genetics analysis identified seven missense mutations in RXRα/β/γ isoforms. Two novel mutations SNP IDs (rs1588299621 and rs1057519958) were identified in RXRα isoform. We used several in silico prediction tools such as SIFT, PolyPhen, I-Mutant, Protein Variation Effect Analyzer (PROVEAN), PANTHER, SNP&Go, PhD-SNP and SNPeffect to predict pathogenicity and protein stability associated with RXR mutations. The structural assessment by DynaMut tool revealed that hydrogen bonds were affected along with hydrophobic and carbonyl interactions resulting in reduced flexibility at the mutated residue positions but ultimately stabilizing the molecule as a whole. Summarizing, analysis of the missense mutations in RXR isoforms showed a mix of conclusive and inconclusive genotype-phenotype correlations suggesting the use of sophisticated computational analysis tools for studying RXR variants.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Soumalya Sarkar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Samridhi Sharma
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Ting Shen
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Melbourne, Australia
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Nitin Chitranshi
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
6
|
Unveiling the Potentiality of Shikonin Derivatives Inhibiting SARS-CoV-2 Main Protease by Molecular Dynamic Simulation Studies. Int J Mol Sci 2023; 24:ijms24043100. [PMID: 36834524 PMCID: PMC9963214 DOI: 10.3390/ijms24043100] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/13/2023] [Accepted: 01/28/2023] [Indexed: 02/09/2023] Open
Abstract
Shikonin, a phytochemical present in the roots of Lithospermum erythrorhizon, is well-known for its broad-spectrum activity against cancer, oxidative stress, inflammation, viruses, and anti-COVID-19 agents. A recent report based on a crystallographic study revealed a distinct conformation of shikonin binding to the SARS-CoV-2 main protease (Mpro), suggesting the possibility of designing potential inhibitors based on shikonin derivatives. The present study aimed to identify potential shikonin derivatives targeting the Mpro of COVID-19 by using molecular docking and molecular dynamics simulations. A total of 20 shikonin derivatives were screened, of which few derivatives showed higher binding affinity than shikonin. Following the MM-GBSA binding energy calculations using the docked structures, four derivatives were retained with the highest binding energy and subjected to molecular dynamics simulation. Molecular dynamics simulation studies suggested that alpha-methyl-n-butyl shikonin, beta-hydroxyisovaleryl shikonin, and lithospermidin-B interacted with two conserved residues, His41 and Cys145, through multiple bonding in the catalytic sites. This suggests that these residues may effectively suppress SARS-CoV-2 progression by inhibiting Mpro. Taken together, the present in silico study concluded that shikonin derivatives may play an influential role in Mpro inhibition.
Collapse
|
7
|
Lockdown Fatigue in Pediatric Respiratory Patients: Lessons from the First COVID-19 Year. CHILDREN (BASEL, SWITZERLAND) 2022; 9:children9121862. [PMID: 36553306 PMCID: PMC9776677 DOI: 10.3390/children9121862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/02/2022]
Abstract
Lockdown policies have been implemented worldwide to limit the spread of COVID-19. “Lockdown fatigue” is a state of exhaustion related to the accumulating effects of repeated lockdowns. The aim of the current study was to examine the effects of repeated lockdowns on children with respiratory disorders. Data on children aged 0−18 years old with respiratory disorders were collected by an electronic survey during the third lockdown in Israel. The retrieved information included demographics and baseline medical status, respiratory clinical status during the third lockdown compared to pre-lockdown periods, lockdown adherence, lifestyle modifications and caregiver emotional status. The results were compared to those of a similar questionnaire distributed during the first lockdown. A total of 234 patients (62% males, 37% females, mean age 6.8 years (confidence interval 6.1−7.5)) were enrolled. Respiratory symptoms and exacerbation frequency were reduced in 76 (35.5%) and 58 (27.4%) patients, respectively, compared to the pre-lockdown period and similar to the first lockdown (p = 0.840 and p = 0.063, respectively). However, compared to the first lockdown, the third lockdown was associated with greater use of reliever medications (p = 0.006), less use of inhaled corticosteroids as routine treatment (p = 0.027), and more pediatric emergency room visits and hospitalizations (p = 0.001 and p < 0.001, respectively). The results also showed an increase in sedentary lifestyle (e.g., reduced physical activity (p = 0.025), less healthy eating habits (p = 0.001)) and reduced adherence to lockdown policies (p < 0.001). These data show that the continuing stability of clinical improvement during lockdown was accompanied by deleterious sequelae that potentially indicate “lockdown fatigue” among children with respiratory disorders.
Collapse
|
8
|
Pitakbut T, Nguyen GN, Kayser O. Activity of THC, CBD, and CBN on Human ACE2 and SARS-CoV1/2 Main Protease to Understand Antiviral Defense Mechanism. PLANTA MEDICA 2022; 88:1047-1059. [PMID: 34638139 DOI: 10.1055/a-1581-3707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
THC, CBD, and CBN were reported as promising candidates against SARS-CoV2 infection, but the mechanism of action of these three cannabinoids is not understood. This study aims to determine the mechanism of action of THC, CBD, and CBN by selecting two essential targets that directly affect the coronavirus infections as viral main proteases and human angiotensin-converting enzyme2. Tested THC and CBD presented a dual-action action against both selected targets. Only CBD acted as a potent viral main protease inhibitor at the IC50 value of 1.86 ± 0.04 µM and exhibited only moderate activity against human angiotensin-converting enzyme2 at the IC50 value of 14.65 ± 0.47 µM. THC acted as a moderate inhibitor against both viral main protease and human angiotensin-converting enzymes2 at the IC50 value of 16.23 ± 1.71 µM and 11.47 ± 3.60 µM, respectively. Here, we discuss cannabinoid-associated antiviral activity mechanisms based on in silico docking studies and in vitro receptor binding studies.
Collapse
Affiliation(s)
- Thanet Pitakbut
- Technical Biochemistry, Faculty of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, Germany
| | - Gia-Nam Nguyen
- Technical Biochemistry, Faculty of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, Germany
- MINDbioscience GmbH, Dortmund, Germany
| | - Oliver Kayser
- Technical Biochemistry, Faculty of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, Germany
| |
Collapse
|
9
|
Understanding the mutational frequency in SARS-CoV-2 proteome using structural features. Comput Biol Med 2022; 147:105708. [PMID: 35714506 PMCID: PMC9173821 DOI: 10.1016/j.compbiomed.2022.105708] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/26/2022] [Accepted: 06/04/2022] [Indexed: 01/18/2023]
Abstract
The prolonged transmission of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus in the human population has led to demographic divergence and the emergence of several location-specific clusters of viral strains. Although the effect of mutation(s) on severity and survival of the virus is still unclear, it is evident that certain sites in the viral proteome are more/less prone to mutations. In fact, millions of SARS-CoV-2 sequences collected all over the world have provided us a unique opportunity to understand viral protein mutations and develop novel computational approaches to predict mutational patterns. In this study, we have classified the mutation sites into low and high mutability classes based on viral isolates count containing mutations. The physicochemical features and structural analysis of the SARS-CoV-2 proteins showed that features including residue type, surface accessibility, residue bulkiness, stability and sequence conservation at the mutation site were able to classify the low and high mutability sites. We further developed machine learning models using above-mentioned features, to predict low and high mutability sites at different selection thresholds (ranging 5-30% of topmost and bottommost mutated sites) and observed the improvement in performance as the selection threshold is reduced (prediction accuracy ranging from 65 to 77%). The analysis will be useful for early detection of variants of concern for the SARS-CoV-2, which can also be applied to other existing and emerging viruses for another pandemic prevention.
Collapse
|
10
|
Reina J, Iglesias C. [Nirmatrelvir plus ritonavir (Paxlovid) a potent SARS-CoV-2 3CLpro protease inhibitor combination]. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2022; 35:236-240. [PMID: 35183067 PMCID: PMC9134883 DOI: 10.37201/req/002.2022] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 12/17/2022]
Abstract
All coronavirus, including SARS-CoV-2, encode two proteases needed for the processing of PP1A and PP1AB polyproteins. The main protease 3CL (chemotripsine-like) gives rise to the formation of NSP11/16 proteins. The 3CL protease has been constituted as one of the possible therapeutic targets for the development of antiviral drugs against SARS-COV-2 due to its highly conserved sequence and structure among all coronaviruses. During the SARS-COV-1 pandemic, a hydroxymethyl ketone derivative (PF-00835231) was identified with an intense inhibitory activity against the 3CL protease. Subsequent chemical modifications gave rise to derivative PF-07321332 (nirmatrelvir) which has shown a high antiviral efficacy against SARS-COV-2. The company's data indicate that it is capable of reducing 89% the risk of hospitalization and death of patients infected with hardly adverse effects. Its effectiveness improves if it is administered orally in the first 24-48 hours and the duration of treatment has been established between 3-5 days. The commercial form has been associated with the antiviral ritonavir that has shown the metabolism of nirmatrelvir, lengthening its average life. This antiviral would be effective against current and future viral variants, since 3CL is not modified in them. The FDA approved this antiviral in November 2021 and EMA is in the final evaluation phase.
Collapse
Affiliation(s)
- J Reina
- Jordi Reina, Unidad de Virología, Servicio de Microbiología, Hospital Universitario Son Espases, Facultad de Medicina (UIB). Carretera Valldemossa 79, 07120 Palma de Mallorca. Spain.
| | | |
Collapse
|
11
|
Raj V, Lee JH, Shim JJ, Lee J. Antiviral activities of 4H-chromen-4-one scaffold-containing flavonoids against SARS-CoV-2 using computational and in vitro approaches. J Mol Liq 2022; 353:118775. [PMID: 35194277 PMCID: PMC8849861 DOI: 10.1016/j.molliq.2022.118775] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 10/14/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022]
Abstract
The widespread outbreak of the novel coronavirus called severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has caused the main health challenge worldwide. This pandemic has attracted the attention of the research communities in various fields, prompting efforts to discover rapid drug molecules for the treatment of the life-threatening COVID-19 disease. This study is aimed at investigating 4H-chromen-4-one scaffold-containing flavonoids that combat the SARS-CoV-2 virus using computational and in vitro approaches. Virtual screening studies of the molecule's library for 4H-chromen-4-one scaffold were performed with the recently reported coronavirus main protease (Mpro, also called 3CLpro) because it plays an essential role in the maturation and processing of the viral polyprotein. Based on the virtual screening, the top hit molecules such as isoginkgetin and afzelin molecules were selected for further estimating in vitro antiviral efficacies against SARS-CoV-2 in Vero cells. Additionally, these molecules were also docked with RNA-dependent RNA Polymerase (RdRp) to reveal the ligands-protein molecular interaction. In the in vitro study, isoginkgetin showed remarkable inhibition potency against the SARS-CoV-2 virus, with an IC50 value of 22.81 μM, compared to remdesivir, chloroquine, and lopinavir with IC50 values of 7.18, 11.63, and 11.49 μM, respectively. Furthermore, the complex stability of isoginkgetin with an active binding pocket of the SARS-CoV-2 Mpro and RdRp supports its inhibitory potency against the SARS-CoV-2. Thus, isoginkgetin is a potent leading drug candidate and needs to be used in in vivo trials for the treatment of SARS-CoV-2 infected patients.
Collapse
|
12
|
Structural and functional significance of the amino acid differences Val 35Thr, Ser 46Ala, Asn 65Ser, and Ala 94Ser in 3C-like proteinases from SARS-CoV-2 and SARS-CoV. Int J Biol Macromol 2021; 193:2113-2120. [PMID: 34774600 PMCID: PMC8580570 DOI: 10.1016/j.ijbiomac.2021.11.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/07/2021] [Accepted: 11/05/2021] [Indexed: 11/28/2022]
Abstract
Three dimensional structures of (chymo)trypsin-like proteinase (3CLpro) from SARS-CoV-2 and SARS-CoV differ at 8 positions. We previously found that the Val86Leu, Lys88Arg, Phe134His, and Asn180Lys mutations in these enzymes can change the orientation of the N- and C-terminal domains of 3CLpro relative to each other, which leads to a change in catalytic activity. This conclusion was derived from the comparison of the structural catalytic core in 169 (chymo)trypsin-like proteinases with the serine/cysteine fold. Val35Thr, Ser46Ala, Asn65Ser, Ala94Ser mutations were not included in that analysis, since they are located far from the catalytic tetrad. In the present work, the structural and functional roles of these variable amino acids at positions 35, 46, 65, and 94 in the 3CLpro sequences of SARS-CoV-2 and SARS-CoV have been established using a comparison of the same set of proteinases leading to the identification of new conservative elements. Comparative analysis showed that, in addition to interdomain mobility, which could modulate catalytic activity, the 3CLpro(s) can use for functional regulation an autolytic loop and the unique Asp33-Asn95 region (the Asp33-Asn95 Zone) in the N-terminal domain. Therefore, all 4 analyzed mutation sites are associated with the unique structure-functional features of the 3CLpro from SARS-CoV-2 and SARS-CoV. Strictly speaking, the presented structural results are hypothetical, since at present there is not a single experimental work on the identification and characterization of autolysis sites in these proteases.
Collapse
|
13
|
Yuan F, Wang L, Fang Y, Wang L. Global SNP analysis of 11,183 SARS-CoV-2 strains reveals high genetic diversity. Transbound Emerg Dis 2021; 68:3288-3304. [PMID: 33207070 PMCID: PMC7753349 DOI: 10.1111/tbed.13931] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/19/2020] [Accepted: 11/13/2020] [Indexed: 02/05/2023]
Abstract
Since first identified in December of 2019, COVID-19 has been quickly spreading to the world in few months and COVID-19 cases are still undergoing rapid surge in most countries worldwide. The causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), adapts and evolves rapidly in nature. With the availability of 16,092 SARS-CoV-2 full genomes in GISAID as of 13 May, we removed the poor-quality genomes and performed mutational profiling analysis for the remaining 11,183 viral genomes. Global analysis of all sequences identified all single nucleotide polymorphisms (SNPs) across the whole genome and critical SNPs with high mutation frequency that contributes to five-clade classification of global strains. A total of 119 SNPs were found with 74 non-synonymous mutations, 43 synonymous mutations and two mutations in intergenic regions. Analysis of geographic pattern of mutational profiling for the whole genome reveals differences between each continent. A transition mutation from C to T represents the most mutation types across the genome, suggesting rapid evolution and adaptation of the virus in host. Amino acid (AA) deletions and insertions found across the genome results in changes in viral protein length and potential function alteration. Mutational profiling for each gene was analysed, and results show that nucleocapsid gene demonstrates the highest mutational frequency, followed by Nsp2, Nsp3 and Spike gene. We further focused on non-synonymous mutational distributions on four key viral proteins, spike with 75 mutations, RNA-dependent-RNA-polymerase with 41 mutations, 3C-like protease with 22 mutations and Papain-like protease with 10 mutations. Results show that non-synonymous mutations on critical sites of these four proteins pose great challenge for development of anti-viral drugs and other countering measures. Overall, this study provides more understanding of genetic diversity/variability of SARS-CoV-2 and insights for development of anti-viral therapeutics.
Collapse
Affiliation(s)
- Fangfeng Yuan
- Department of PathobiologyCollege of Veterinary MedicineUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Liping Wang
- Department of Diagnostic Medicine and PathobiologyCollege of Veterinary MedicineKansas State UniversityManhattanKansasUSA
| | - Ying Fang
- Department of PathobiologyCollege of Veterinary MedicineUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Leyi Wang
- Veterinary Diagnostic Laboratory and Department of Veterinary Clinical MedicineCollege of Veterinary MedicineUniversity of IllinoisUrbanaIllinoisUSA
| |
Collapse
|
14
|
Aherfi S, Pradines B, Devaux C, Honore S, Colson P, Scola BL, Raoult D. Drug repurposing against SARS-CoV-1, SARS-CoV-2 and MERS-CoV. Future Microbiol 2021; 16:1341-1370. [PMID: 34755538 PMCID: PMC8579950 DOI: 10.2217/fmb-2021-0019] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Since the beginning of the COVID-19 pandemic, large in silico screening studies and numerous in vitro studies have assessed the antiviral activity of various drugs on SARS-CoV-2. In the context of health emergency, drug repurposing represents the most relevant strategy because of the reduced time for approval by international medicines agencies, the low cost of development and the well-known toxicity profile of such drugs. Herein, we aim to review drugs with in vitro antiviral activity against SARS-CoV-2, combined with molecular docking data and results from preliminary clinical studies. Finally, when considering all these previous findings, as well as the possibility of oral administration, 11 molecules consisting of nelfinavir, favipiravir, azithromycin, clofoctol, clofazimine, ivermectin, nitazoxanide, amodiaquine, heparin, chloroquine and hydroxychloroquine, show an interesting antiviral activity that could be exploited as possible drug candidates for COVID-19 treatment.
Collapse
Affiliation(s)
- Sarah Aherfi
- Aix-Marseille Université, Assistance Publique – Hôpitaux de Marseille (AP-HM), Marseille, 13005, France
- Institut Hospitalo-Universitaire (IHU) – Méditerranée Infection, Marseille, 13005, France
- Microbes, Evolution, Phylogeny & Infection (MEΦI), Marseille, 13005, France
| | - Bruno Pradines
- Institut Hospitalo-Universitaire (IHU) – Méditerranée Infection, Marseille, 13005, France
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, 13005, France
- Aix-Marseille Univ, IRD, SSA, AP-HM, VITROME, Marseille, 13005, France
- Centre national de référence du paludisme, Marseille, 13005, France
| | - Christian Devaux
- Institut Hospitalo-Universitaire (IHU) – Méditerranée Infection, Marseille, 13005, France
| | - Stéphane Honore
- Aix Marseille Université, Laboratoire de Pharmacie Clinique, Marseille, 13005, France
- AP-HM, hôpital Timone, service pharmacie, Marseille, 13005, France
| | - Philippe Colson
- Aix-Marseille Université, Assistance Publique – Hôpitaux de Marseille (AP-HM), Marseille, 13005, France
- Institut Hospitalo-Universitaire (IHU) – Méditerranée Infection, Marseille, 13005, France
- Microbes, Evolution, Phylogeny & Infection (MEΦI), Marseille, 13005, France
| | - Bernard La Scola
- Aix-Marseille Université, Assistance Publique – Hôpitaux de Marseille (AP-HM), Marseille, 13005, France
- Institut Hospitalo-Universitaire (IHU) – Méditerranée Infection, Marseille, 13005, France
- Microbes, Evolution, Phylogeny & Infection (MEΦI), Marseille, 13005, France
| | - Didier Raoult
- Aix-Marseille Université, Assistance Publique – Hôpitaux de Marseille (AP-HM), Marseille, 13005, France
- Institut Hospitalo-Universitaire (IHU) – Méditerranée Infection, Marseille, 13005, France
| |
Collapse
|
15
|
Sattari A, Ramazani A, Aghahosseini H. Repositioning therapeutics for COVID-19: virtual screening of the potent synthetic and natural compounds as SARS-CoV-2 3CLpro inhibitors. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2021. [PMCID: PMC7994965 DOI: 10.1007/s13738-021-02235-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The widespread transmission of SARS-CoV-2 has sparked alarm worldwide. Attaining the best drugs to treat COVID-19 at the shortest possible time is one of the most critical issues in this urgent situation. Molecular docking investigation of the therapeutic potential of marketed drugs is a fast and cost-effective approach to provide a solution to this problem. The recent research efforts have led to the resolving of the 3CLpro structure as a key protease in the lifecycle of coronavirus, which could facilitate in silico evaluation of drug candidates. Herein, the similarity between the SARS-CoV-2-3CL main protease and the other SARS-CoV receptors was evaluated via multiple sequence alignment and phylogenetic tree. The reported structure of the 3CLpro was considered as a target to identify potential inhibitors for treating COVID-19 using molecular docking based virtual screening protocol. Accordingly, a database of 50 synthetic compounds with various pharmacological usage such as antiviral, anti-inflammatory, anti-human immunodeficiency viruses, antimalarial, antibacterial, anticancer, and antioxidant including approved drugs and those undergoing clinical trials, and 40 natural compounds particularly those employed in traditional Iranian medicine was constructed. The output of multiple sequence alignment analysis showed that SARS-CoV-2 main protease shares a similarity of 96% with SARS-CoV. Also, the docking results indicated that the licofelone acyl glucuronide as an anti-inflammatory drug and delta-bilirubin as an antioxidant and anti-inflammatory agent as well as kappa-carrageenan conformer, beta-D-galactopyranosyl and calycosin 7-O-glucoside as natural compounds with minimal side-effects, according to in vitro studies, are good candidates to block the enzymatic activity of SARS-CoV-2 3CLpro. Moreover, the compound 1 with the highest negative binding energy is a chemical compound that due to its favorable interactions with the 3CLpro can be identified as a representative potential drug candidate for COVID-19.
Collapse
Affiliation(s)
- Ahmad Sattari
- Department of Chemistry, Faculty of Science, University of Zanjan, 45371-38791 Zanjan, Iran
| | - Ali Ramazani
- Department of Chemistry, Faculty of Science, University of Zanjan, 45371-38791 Zanjan, Iran
- Department of Biotechnology, Research Institute of Modern Biological Techniques (RIMBT), University of Zanjan, 45371-38791 Zanjan, Iran
| | - Hamideh Aghahosseini
- Department of Chemistry, Faculty of Science, University of Zanjan, 45371-38791 Zanjan, Iran
- Department of Biotechnology, Research Institute of Modern Biological Techniques (RIMBT), University of Zanjan, 45371-38791 Zanjan, Iran
| |
Collapse
|
16
|
Huijghebaert S, Hoste L, Vanham G. Essentials in saline pharmacology for nasal or respiratory hygiene in times of COVID-19. Eur J Clin Pharmacol 2021; 77:1275-1293. [PMID: 33772626 PMCID: PMC7998085 DOI: 10.1007/s00228-021-03102-3] [Citation(s) in RCA: 20] [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: 11/09/2020] [Accepted: 01/28/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE Nasal irrigation or nebulizing aerosol of isotonic or hypertonic saline is a traditional method for respiratory or nasal care. A recent small study in outpatients with COVID-19 without acute respiratory distress syndrome suggests substantial symptom resolution. We therefore analyzed pharmacological/pharmacodynamic effects of isotonic or hypertonic saline, relevant to SARS-CoV-2 infection and respiratory care. METHODS Mixed search method. RESULTS Due to its wetting properties, saline achieves an improved spreading of alveolar lining fluid and has been shown to reduce bio-aerosols and viral load. Saline provides moisture to respiratory epithelia and gels mucus, promotes ciliary beating, and improves mucociliary clearance. Coronaviruses and SARS-CoV-2 damage ciliated epithelium in the nose and airways. Saline inhibits SARS-CoV-2 replication in Vero cells; possible interactions involve the viral ACE2-entry mechanism (chloride-dependent ACE2 configuration), furin and 3CLpro (inhibition by NaCl), and the sodium channel ENaC. Saline shifts myeloperoxidase activity in epithelial or phagocytic cells to produce hypochlorous acid. Clinically, nasal or respiratory airway care with saline reduces symptoms of seasonal coronaviruses and other common cold viruses. Its use as aerosol reduces hospitalization rates for bronchiolitis in children. Preliminary data suggest symptom reduction in symptomatic COVID-19 patients if saline is initiated within 48 h of symptom onset. CONCLUSIONS Saline interacts at various levels relevant to nasal or respiratory hygiene (nasal irrigation, gargling or aerosol). If used from the onset of common cold symptoms, it may represent a useful add-on to first-line interventions for COVID-19. Formal evaluation in mild COVID-19 is desirable as to establish efficacy and optimal treatment regimens.
Collapse
Affiliation(s)
| | - Levi Hoste
- Pediatric Pulmonology, Infectious Diseases and Immunology, Ghent University Hospital, Ghent, Belgium
- Primary Immunodeficiency Research Lab, Center for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Center, Ghent University Hospital, Ghent, Belgium
| | - Guido Vanham
- Department of Biomedical Sciences, Institute of Tropical Medicine and University of Antwerp, Antwerp, Belgium
| |
Collapse
|
17
|
Chitranshi N, Kumar A, Sheriff S, Gupta V, Godinez A, Saks D, Sarkar S, Shen T, Mirzaei M, Basavarajappa D, Abyadeh M, Singh SK, Dua K, Zhang KYJ, Graham SL, Gupta V. Identification of Novel Cathepsin B Inhibitors with Implications in Alzheimer's Disease: Computational Refining and Biochemical Evaluation. Cells 2021; 10:cells10081946. [PMID: 34440715 PMCID: PMC8391575 DOI: 10.3390/cells10081946] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
Amyloid precursor protein (APP), upon proteolytic degradation, forms aggregates of amyloid β (Aβ) and plaques in the brain, which are pathological hallmarks of Alzheimer’s disease (AD). Cathepsin B is a cysteine protease enzyme that catalyzes the proteolytic degradation of APP in the brain. Thus, cathepsin B inhibition is a crucial therapeutic aspect for the discovery of new anti-Alzheimer’s drugs. In this study, we have employed mixed-feature ligand-based virtual screening (LBVS) by integrating pharmacophore mapping, docking, and molecular dynamics to detect small, potent molecules that act as cathepsin B inhibitors. The LBVS model was generated by using hydrophobic (HY), hydrogen bond acceptor (HBA), and hydrogen bond donor (HBD) features, using a dataset of 24 known cathepsin B inhibitors of both natural and synthetic origins. A validated eight-feature pharmacophore hypothesis (Hypo III) was utilized to screen the Maybridge chemical database. The docking score, MM-PBSA, and MM-GBSA methodology was applied to prioritize the lead compounds as virtual screening hits. These compounds share a common amide scaffold, and showed important interactions with Gln23, Cys29, His110, His111, Glu122, His199, and Trp221. The identified inhibitors were further evaluated for cathepsin-B-inhibitory activity. Our study suggests that pyridine, acetamide, and benzohydrazide compounds could be used as a starting point for the development of novel therapeutics.
Collapse
Affiliation(s)
- Nitin Chitranshi
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
- Correspondence: (N.C.); (V.G.); Tel.: +61-(02)-9850-2804 (N.C.)
| | - Ashutosh Kumar
- Center for Biosystems Dynamics Research, Laboratory for Structural Bioinformatics, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama 230-0045, Kanagawa, Japan; (A.K.); (K.Y.J.Z.)
| | - Samran Sheriff
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Veer Gupta
- School of Medicine, Faculty of Health, Deakin University, Geelong, VIC 3220, Australia;
| | - Angela Godinez
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Danit Saks
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Soumalya Sarkar
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Ting Shen
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Mehdi Mirzaei
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Devaraj Basavarajappa
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Morteza Abyadeh
- Cell Science Research Center, Department of Molecular Systems Biology, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran;
| | - Sachin K. Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia;
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kam Y. J. Zhang
- Center for Biosystems Dynamics Research, Laboratory for Structural Bioinformatics, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama 230-0045, Kanagawa, Japan; (A.K.); (K.Y.J.Z.)
| | - Stuart L. Graham
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Vivek Gupta
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
- Correspondence: (N.C.); (V.G.); Tel.: +61-(02)-9850-2804 (N.C.)
| |
Collapse
|
18
|
Menezes JCJMDS, Campos VR. Natural biflavonoids as potential therapeutic agents against microbial diseases. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 769:145168. [PMID: 33493916 DOI: 10.1016/j.scitotenv.2021.145168] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 06/12/2023]
Abstract
Microbes broadly constitute several organisms like viruses, protozoa, bacteria, and fungi present in our biosphere. Fast-paced environmental changes have influenced contact of human populations with newly identified microbes resulting in diseases that can spread quickly. These microbes can cause infections like HIV, SARS-CoV2, malaria, nosocomial Escherichia coli, methicillin-resistant Staphylococcus aureus (MRSA), or Candida infection for which there are no available vaccines/drugs or are less efficient to prevent or treat these infections. In the pursuit to find potential safe agents for therapy of microbial infections, natural biflavonoids like amentoflavone, tetrahydroamentoflavone, ginkgetin, bilobetin, morelloflavone, agathisflavone, hinokiflavone, Garcinia biflavones 1 (GB1), Garcinia biflavones 2 (GB2), robustaflavone, strychnobiflavone, ochnaflavone, dulcisbiflavonoid C, tetramethoxy-6,6″-bigenkwanin and other derivatives isolated from several species of plants can provide effective starting points and become a source of future drugs. These biflavonoids show activity against influenza, severe acute respiratory syndrome (SARS), dengue, HIV-AIDS, coxsackieviral, hepatitis, HSV, Epstein-Barr virus (EBV), protozoal (Leishmaniasis, Malaria) infections, bacterial and fungal infections. Some of the biflavonoids can provide antiviral and protozoal activity by inhibition of neuraminidase, chymotrypsin-like protease, DV-NS5 RNA dependant RNA polymerase, reverse transcriptase (RT), fatty acid synthase, DNA polymerase, UL54 gene expression, Epstein-Barr virus early antigen activation, recombinant cysteine protease type 2.8 (r-CPB2.8), Plasmodium falciparum enoyl-acyl carrier protein (ACP) reductase or cause depolarization of parasitic mitochondrial membranes. They may also provide anti-inflammatory therapeutic activity against the infection-induced cytokine storm. Considering the varied bioactivity of these biflavonoids against these organisms, their structure-activity relationships are derived and wherever possible compared with monoflavones. Overall, this review aims to highlight these natural biflavonoids and briefly discuss their sources, reported mechanism of action, pharmacological uses, and comment on resistance mechanism, flavopiridol repurposing and the bioavailability aspects to provide a starting point for anti-microbial research in this area.
Collapse
Affiliation(s)
- José C J M D S Menezes
- Section of Functional Morphology, Faculty of Pharmaceutical Sciences, Nagasaki International University, 2825-7 Huis Ten Bosch, Sasebo, Nagasaki 859-3298, Japan.
| | - Vinícius R Campos
- Department of Organic Chemistry, Institute of Chemistry, Fluminense Federal University, Campus do Valonguinho, 24020-141 Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Eberle RJ, Olivier DS, Amaral MS, Gering I, Willbold D, Arni RK, Coronado MA. The Repurposed Drugs Suramin and Quinacrine Cooperatively Inhibit SARS-CoV-2 3CL pro In Vitro. Viruses 2021; 13:873. [PMID: 34068686 PMCID: PMC8170883 DOI: 10.3390/v13050873] [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: 04/02/2021] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023] Open
Abstract
Since the first report of a new pneumonia disease in December 2019 (Wuhan, China) the WHO reported more than 148 million confirmed cases and 3.1 million losses globally up to now. The causative agent of COVID-19 (SARS-CoV-2) has spread worldwide, resulting in a pandemic of unprecedented magnitude. To date, several clinically safe and efficient vaccines (e.g., Pfizer-BioNTech, Moderna, Johnson & Johnson, and AstraZeneca COVID-19 vaccines) as well as drugs for emergency use have been approved. However, increasing numbers of SARS-Cov-2 variants make it imminent to identify an alternative way to treat SARS-CoV-2 infections. A well-known strategy to identify molecules with inhibitory potential against SARS-CoV-2 proteins is repurposing clinically developed drugs, e.g., antiparasitic drugs. The results described in this study demonstrated the inhibitory potential of quinacrine and suramin against SARS-CoV-2 main protease (3CLpro). Quinacrine and suramin molecules presented a competitive and noncompetitive inhibition mode, respectively, with IC50 values in the low micromolar range. Surface plasmon resonance (SPR) experiments demonstrated that quinacrine and suramin alone possessed a moderate or weak affinity with SARS-CoV-2 3CLpro but suramin binding increased quinacrine interaction by around a factor of eight. Using docking and molecular dynamics simulations, we identified a possible binding mode and the amino acids involved in these interactions. Our results suggested that suramin, in combination with quinacrine, showed promising synergistic efficacy to inhibit SARS-CoV-2 3CLpro. We suppose that the identification of effective, synergistic drug combinations could lead to the design of better treatments for the COVID-19 disease and repurposable drug candidates offer fast therapeutic breakthroughs, mainly in a pandemic moment.
Collapse
Affiliation(s)
- Raphael J. Eberle
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52428 Jülich, Germany; (I.G.); (D.W.)
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße, 40225 Düsseldorf, Germany
| | - Danilo S. Olivier
- Campus Cimba, Federal University of Tocantins, Araguaína, TO 77824-838, Brazil;
| | - Marcos S. Amaral
- Institute of Physics, Federal University of Mato Grosso do Sul, Campo Grande, MS 79070-900, Brazil;
| | - Ian Gering
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52428 Jülich, Germany; (I.G.); (D.W.)
| | - Dieter Willbold
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52428 Jülich, Germany; (I.G.); (D.W.)
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße, 40225 Düsseldorf, Germany
- JuStruct: Jülich Centre for Structural Biology, Forchungszentrum Jülich, 52428 Jülich, Germany
| | - Raghuvir K. Arni
- Multiuser Center for Biomolecular Innovation, IBILCE, Universidade Estadual Paulista (UNESP), São Jose do Rio Preto, SP 15054-000, Brazil;
| | - Monika A. Coronado
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52428 Jülich, Germany; (I.G.); (D.W.)
- Multiuser Center for Biomolecular Innovation, IBILCE, Universidade Estadual Paulista (UNESP), São Jose do Rio Preto, SP 15054-000, Brazil;
| |
Collapse
|
20
|
Ulfah M, Helianti I. Bioinformatic analysis of the whole genome sequences of SARS-CoV-2 from Indonesia. IRANIAN JOURNAL OF MICROBIOLOGY 2021; 13:145-155. [PMID: 34540148 PMCID: PMC8408024 DOI: 10.18502/ijm.v13i2.5973] [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: 12/22/2022]
Abstract
BACKGROUND AND OBJECTIVES In first May 2020, Indonesia has been successfully submitted the first three full-length sequence of SARS-CoV-2 to GISAID database. Until September 10th, 2020, Indonesia had submitted 54 WGS. In this study, we have analyzed and annotated SARS-CoV-2 mutations in spike protein and main proteases. MATERIALS AND METHODS The Whole Genome Sequence (WGS) of Indonesia were obtained from GISAID data base. The 54 data were taken from March to September 10th, 2020. The sequences corresponded to Spike Protein (SP), 3-chymotrypsin like protease (3CLpro), and papain like protease (PLpro) were selected. The Wuhan genome was used as reference. RESULTS In total WGS from Indonesia, we found 5 major clades, which dominated as G clade, where the mutation of D614G was found. This D614G was identified as much as 59%, which mostly reported in late samples submitted. Beside D614G mutation, we report three unique mutations: A352S, S477I, and Q677H. Besides, some mutations were also detected in two domains that were expected to be conserved region, the main viral proteases: PLpro (P77L and V205I), 3CLpro (M49I and L50F). CONCLUSION The analysis of SARS-CoV-2 from WGS Indonesia showed a high genetic variation. The diversity in SARS-CoV-2 may epidemiologically enhance virulence and transmission of this virus. The prevalence of D614G over the time in different locations, indicating that changes in this mutation may related to host infection and the viral transmission. However, some mutations that have been reported in this study were not eligible for the most stable conformation.
Collapse
Affiliation(s)
- Maria Ulfah
- Department of Biocatalyst, Centre for Bioindustrial Technology, Agency for Assessment and Application of Technology (BPPT), Laboratorium of Bioindustrial Technology, LAPTIAB BPPT, Puspiptek-Serpong, Tangerang Selatan, Indonesia
| | - Is Helianti
- Department of Biocatalyst, Centre for Bioindustrial Technology, Agency for Assessment and Application of Technology (BPPT), Laboratorium of Bioindustrial Technology, LAPTIAB BPPT, Puspiptek-Serpong, Tangerang Selatan, Indonesia
| |
Collapse
|
21
|
Das JK, Roy S. A study on non-synonymous mutational patterns in structural proteins of SARS-CoV-2. Genome 2021; 64:665-678. [PMID: 33788636 DOI: 10.1139/gen-2020-0157] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SARS-CoV-2 is mutating and creating divergent variants across the world. An in-depth investigation of the amino acid substitutions in the genomic signature of SARS-CoV-2 proteins is highly essential for understanding its host adaptation and infection biology. A total of 9587 SARS-CoV-2 structural protein sequences collected from 49 different countries are used to characterize protein-wise variants, substitution patterns (type and location), and major substitution changes. The majority of the substitutions are distinct, mostly in a particular location, and lead to a change in an amino acid's biochemical properties. In terms of mutational changes, envelope (E) and membrane (M) proteins are relatively more stable than nucleocapsid (N) and spike (S) proteins. Several co-occurrence substitutions are observed, particularly in S and N proteins. Substitution specific to active sub-domains reveals that heptapeptide repeat, fusion peptides, transmembrane in S protein, and N-terminal and C-terminal domains in the N protein are remarkably mutated. We also observe a few deleterious mutations in the above domains. The overall study on non-synonymous mutation in structural proteins of SARS-CoV-2 at the start of the pandemic indicates a diversity amongst virus sequences.
Collapse
Affiliation(s)
- Jayanta Kumar Das
- Department of Pediatrics, Johns Hopkins University School of Medicine, Maryland, USA
| | - Swarup Roy
- Network Reconstruction & Analysis (NetRA) Lab, Department of Computer Applications, Sikkim University, Gangtok, India
| |
Collapse
|
22
|
Denesyuk AI, Permyakov SE, Johnson MS, Permyakov EA, Uversky VN, Denessiouk K. Structural leitmotif and functional variations of the structural catalytic core in (chymo)trypsin-like serine/cysteine fold proteinases. Int J Biol Macromol 2021; 179:601-609. [PMID: 33713772 DOI: 10.1016/j.ijbiomac.2021.03.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 11/29/2022]
Abstract
Proteinases with the (chymo)trypsin-like serine/cysteine fold comprise a large superfamily performing their function through the Acid - Base - Nucleophile catalytic triad. In our previous work (Denesyuk AI, Johnson MS, Salo-Ahen OMH, Uversky VN, Denessiouk K. Int J Biol Macromol. 2020;153:399-411), we described a universal three-dimensional (3D) structural motif, NBCZone, that contains eleven amino acids: dipeptide 42 T-43 T, pentapeptide 54 T-55 T-56 T-57 T(base)-58 T, tripeptide 195 T(nucleophile)-196 T-197 T and residue 213 T (T - numeration of amino acids in trypsin). The comparison of the NBCZones among the members of the (chymo)trypsin-like protease family suggested the existence of 15 distinct groups. Within each group, the NBCZones incorporate an identical set of conserved interactions and bonds. In the present work, the structural environment of the catalytic acid at the position 102 T and the fourth member of the "catalytic tetrad" at the position 214 T was analyzed in 169 3D structures of proteinases with the (chymo)trypsin-like serine/cysteine fold. We have identified a complete Structural Catalytic Core (SCC) consisting of two classes and four groups. The proteinases belonging to different classes and groups differ from each other by the nature of the interaction between their N- and C-terminal β-barrels. Comparative analysis of the 3CLpro(s) from SARS-CoV-2 and SARS-CoV, used as an example, showed that the amino acids at positions 103 T and 179 T affect the nature of the interaction of the "catalytic acid" core (102 T-Core, N-terminal β-barrel) with the "supplementary" core (S-Core, C-terminal β-barrel), which ultimately results in the modulation of the enzymatic activity. The reported analysis represents an important standalone contribution to the analysis and systematization of the 3D structures of (chymo)trypsin-like serine/cysteine fold proteinases. The use of the developed approach for the comparison of 3D structures will allow, in the event of the appearance of new representatives of a given fold in the PDB, to quickly determine their structural homologues with the identification of possible differences.
Collapse
Affiliation(s)
- Alexander I Denesyuk
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow region 142290, Russia; Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland.
| | - Sergei E Permyakov
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow region 142290, Russia
| | - Mark S Johnson
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Eugene A Permyakov
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow region 142290, Russia
| | - Vladimir N Uversky
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow region 142290, Russia; Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Konstantin Denessiouk
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| |
Collapse
|
23
|
C. T. S, M. D, P. R. R, K. M, E. M. A, Balachandran I. Chemical profiling of selected Ayurveda formulations recommended for COVID-19. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2021; 10:2. [PMID: 33457430 PMCID: PMC7799399 DOI: 10.1186/s43088-020-00089-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/26/2020] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND The novel coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is the global health concern since December 2019. It has become a big challenge for the researchers to find a solution for this newly evolved pandemic. In Ayurveda point of view, COVID-19 is a Janapadodhwamsa vikara (epidemic disease), a situation where the environment-air, water, land, and seasons-is vitiated, causing a simultaneous manifestation of a disease among large populations. The aim of this study is to identify the active compounds of selected Ayurveda medicines recommended for COVID-19. RESULTS The selected preparations are traditionally recommended for the management of various kinds of fever including the infectious ones and to enhance the immunity. HPTLC analysis of the same showed presence of many active molecules like umbelliferone, scopoletin, caffeic acid, ferulic acid, gallic acid, piperine, curcumin, berberine, and palmatine. CONCLUSION The study provided valuable scientific data regarding the active ingredients of the selected medicines with proven therapeutic potentials like anti-viral, immunomodulatory, and anti-inflammatory activities.
Collapse
Affiliation(s)
- Sulaiman C. T.
- Phytochemistry Division, Centre for Medicinal Plants Research, Arya Vaidya Sala, Kottakkal, Malappuram, Kerala 676503 India
| | - Deepak M.
- Phytochemistry Division, Centre for Medicinal Plants Research, Arya Vaidya Sala, Kottakkal, Malappuram, Kerala 676503 India
| | - Ramesh P. R.
- Clinical Research Department, Arya Vaidya Sala, Kottakkal, Kerala India
| | - Mahesh K.
- Clinical Research Department, Arya Vaidya Sala, Kottakkal, Kerala India
| | - Anandan E. M.
- Product Development Department, Arya Vaidya Sala, Kottakkal, Kerala India
| | - Indira Balachandran
- Phytochemistry Division, Centre for Medicinal Plants Research, Arya Vaidya Sala, Kottakkal, Malappuram, Kerala 676503 India
| |
Collapse
|
24
|
Rathi H, Burman V, Datta SK, Rana SV, Mirza AA, Saha S, Kumar R, Naithani M. Review on COVID-19 Etiopathogenesis, Clinical Presentation and Treatment Available with Emphasis on ACE2. Indian J Clin Biochem 2021; 36:3-22. [PMID: 33424145 PMCID: PMC7778574 DOI: 10.1007/s12291-020-00953-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 12/19/2020] [Indexed: 02/07/2023]
Abstract
In December 2019, Wuhan city in the Hubei province of China reported for the first time a cluster of patients infected with a novel coronavirus, since then there has been an outburst of this disease across the globe affecting millions of human inhabitants. Severe acute respiratory syndrome coronavirus type-2 (SARS-CoV-2), is a member of beta coronavirus family which upon exposure caused a highly infectious disease called novel coronavirus disease-2019 (COVID-19). COVID-19, a probably bat originated disease was declared by World Health Organization (WHO) as a global pandemic in March 2020. Since then, despite rigorous global containment and quarantine efforts, the disease has affected nearly 56,261,952 laboratory confirmed human population and caused deaths of over 1,349,506 lives worldwide. Virus passes in majority through respiratory droplets and then enters lung epithelial cells by binding to angiotensin converting enzyme 2 (ACE2) receptor and there it undergoes replication and targeting host cells causing severe pathogenesis. Majority of human population exposed to SARS-CoV-2 having fully functional immune system undergo asymptomatic infection while 5-10% are symptomatic and only 1-2% are critically affected and requires ventilation support. Older people or people with co-morbidities are severely affected by COVID-19. These categories of patients also display cytokine storm due to dysfunctional immune response which brutally destroys the affected organs and may lead to death in some. Real time PCR is still considered as standard method of diagnosis along with other serology, radiological and biochemical investigations. Till date, no specific validated medication is available for the treatment of COVID-19 patients. Thus, this review provides detailed knowledge about the different landscapes of disease incidence, etiopathogenesis, involvement of various organs, diagnostic criteria's and treatment guidelines followed for management of COVID-19 infection since its inception. In conclusion, extensive research to recognize novel pathways and their cross talk to combat this virus in precarious settings is our future positive hope.
Collapse
Affiliation(s)
- Himani Rathi
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, Uttarakhand India
| | - Vishakha Burman
- Department of Biotechnology, SVBP University of Agriculture and Technology, Meerut, Uttar Pradesh India
| | - Sudip Kumar Datta
- Department of Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Satya Vati Rana
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, Uttarakhand India
| | - Anissa Atif Mirza
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, Uttarakhand India
| | - Sarama Saha
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, Uttarakhand India
| | - Raman Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, Uttarakhand India
| | - Manisha Naithani
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, Uttarakhand India
| |
Collapse
|
25
|
Turakhia Y, De Maio N, Thornlow B, Gozashti L, Lanfear R, Walker CR, Hinrichs AS, Fernandes JD, Borges R, Slodkowicz G, Weilguny L, Haussler D, Goldman N, Corbett-Detig R. Stability of SARS-CoV-2 phylogenies. PLoS Genet 2020; 16:e1009175. [PMID: 33206635 PMCID: PMC7721162 DOI: 10.1371/journal.pgen.1009175] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/07/2020] [Accepted: 10/06/2020] [Indexed: 12/23/2022] Open
Abstract
The SARS-CoV-2 pandemic has led to unprecedented, nearly real-time genetic tracing due to the rapid community sequencing response. Researchers immediately leveraged these data to infer the evolutionary relationships among viral samples and to study key biological questions, including whether host viral genome editing and recombination are features of SARS-CoV-2 evolution. This global sequencing effort is inherently decentralized and must rely on data collected by many labs using a wide variety of molecular and bioinformatic techniques. There is thus a strong possibility that systematic errors associated with lab-or protocol-specific practices affect some sequences in the repositories. We find that some recurrent mutations in reported SARS-CoV-2 genome sequences have been observed predominantly or exclusively by single labs, co-localize with commonly used primer binding sites and are more likely to affect the protein-coding sequences than other similarly recurrent mutations. We show that their inclusion can affect phylogenetic inference on scales relevant to local lineage tracing, and make it appear as though there has been an excess of recurrent mutation or recombination among viral lineages. We suggest how samples can be screened and problematic variants removed, and we plan to regularly inform the scientific community with our updated results as more SARS-CoV-2 genome sequences are shared (https://virological.org/t/issues-with-sars-cov-2-sequencing-data/473 and https://virological.org/t/masking-strategies-for-sars-cov-2-alignments/480). We also develop tools for comparing and visualizing differences among very large phylogenies and we show that consistent clade- and tree-based comparisons can be made between phylogenies produced by different groups. These will facilitate evolutionary inferences and comparisons among phylogenies produced for a wide array of purposes. Building on the SARS-CoV-2 Genome Browser at UCSC, we present a toolkit to compare, analyze and combine SARS-CoV-2 phylogenies, find and remove potential sequencing errors and establish a widely shared, stable clade structure for a more accurate scientific inference and discourse.
Collapse
Affiliation(s)
- Yatish Turakhia
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, United States of America
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, United States of America
| | - Nicola De Maio
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, United Kingdom
| | - Bryan Thornlow
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, United States of America
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, United States of America
| | - Landen Gozashti
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, United States of America
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, United States of America
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, United States of America
| | - Robert Lanfear
- Department of Ecology and Evolution, Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Conor R. Walker
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Angie S. Hinrichs
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, United States of America
| | - Jason D. Fernandes
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, United States of America
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, United States of America
- Howard Hughes Medical Institute, University of California, Santa Cruz, CA, United States of America
| | - Rui Borges
- Institut für Populationsgenetik, Vetmeduni Vienna, Wien, Austria
| | - Greg Slodkowicz
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Lukas Weilguny
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, United Kingdom
| | - David Haussler
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, United States of America
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, United States of America
- Howard Hughes Medical Institute, University of California, Santa Cruz, CA, United States of America
| | - Nick Goldman
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, United Kingdom
| | - Russell Corbett-Detig
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, United States of America
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, United States of America
| |
Collapse
|
26
|
Das SK. The Pathophysiology, Diagnosis and Treatment of Corona Virus Disease 2019 (COVID-19). Indian J Clin Biochem 2020; 35:385-396. [PMID: 32837036 PMCID: PMC7424135 DOI: 10.1007/s12291-020-00919-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022]
Abstract
Since the beginning of this century, beta coronaviruses (CoV) have caused three zoonotic outbreaks. However, little is currently known about the biology of the newly emerged SARS-CoV-2 in late 2019. There is a spectrum of clinical features from mild to severe life threatening disease with major complications like severe pneumonia, acute respiratory distress syndrome, acute cardiac injury and septic shock. The genome of SARS-CoV-2 encodes polyproteins, four structural proteins and six accessory proteins. SARS-CoV-2 tends to utilize Angiotensin-converting enzyme 2 (ACE2) of various mammals. The imbalance between ACE/Ang II/AT1R pathway and ACE2/Ang(1-7)/Mas receptor pathway in the renin-angiotensin system leads to multi-system inflammation. The early symptoms of COVID-19 pneumonia are low to midgrade fever, dry cough and fatigue. Vigilant screening is important. The diagnosis of COVID-19 should be based on imaging findings along with epidemiological history and nucleic acid detection. Isolation and quarantine of suspected cases is recommended. Management is primarily supportive, with newer antiviral drugs/vaccines under investigation.
Collapse
Affiliation(s)
- Subir Kumar Das
- Department of Biochemistry, College of Medicine and JNM Hospital, WBUHS, Kalyani, Nadia, West Bengal 741235 India
| |
Collapse
|