101
|
He J, Mei Q, Peng Y, Xie J, Li W, Ding C, Jiang C, Chen Q, Wang J, Zhang Y, Ni S, Yu J, Liu T, Yang W, Gong L, Zhang X, Yuan Y, Zhang Z, He L, He H, Sun Y, Wu J, Liu Z, Gao Y. Are the original SARS-CoV-2 novel mutants from in vitro culture able to escape the immune response? J Med Virol 2023; 95:e28931. [PMID: 37448226 DOI: 10.1002/jmv.28931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023]
Abstract
Monitoring variations in the virus genome to understand the SARS-CoV-2 evolution and spread of the virus is extremely important. Seven early SARS-CoV-2 isolates in China were cultured in vitro and were analyzed for their viral infectivity through viral growth assay, tissue culture infectious dose (TCID50 ) assay, spike protein quantification, and next generation sequencing analysis, and the resultant mutations in spike protein were used to generate the corresponding pseudoviruses for analysis of immune escape from vaccination and postinfection immunity. The results revealed that in vitro cultured SARS-CoV-2 virus had much higher mutation frequency (up to ~20 times) than that in infected patients, suggesting that SARS-CoV-2 diversify under favorable conditions. Monitoring viral mutations is not only helpful for better understanding of virus evolution and virulence change, but also the key to prevent virus transmission and disease progression. Compared with the D614G reference strain, a pseudovirus strain of SARS-CoV-2 was constructed with a high mutation rate site on the spike protein. We found some novel spike mutations during in vitro culture, such as E868Q, conferred further immune escape ability.
Collapse
Affiliation(s)
- Jun He
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Qingmin Mei
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Yousong Peng
- Hunan Provincial Key Laboratory of Medical Virology, Bioinformatics Center, College of Biology, Hunan University, Changsha, China
| | - Jiajia Xie
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Weiwei Li
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Chengchao Ding
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Chengcheng Jiang
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Qingqing Chen
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Jing Wang
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Yuqing Zhang
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Shuangshuang Ni
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Junling Yu
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Tao Liu
- Annoroad Gene Technology (Beijing) Co., Beijing, China
| | - Weifei Yang
- Annoroad Gene Technology (Beijing) Co., Beijing, China
| | - Lei Gong
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Xiangyu Zhang
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Yuan Yuan
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Zhuhui Zhang
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Lan He
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Hongliang He
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Yong Sun
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Jiabing Wu
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Zhirong Liu
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Public Health Research Institute of Anhui Province, Hefei, Anhui, China
| | - Yong Gao
- United Laboratory of The First Affiliated Hospital of USTC and Anhui Provincial Center for Disease Control and Prevention, Hefei, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
102
|
Wu CY, Yang YH, Lin YS, Shu LH, Cheng YC, Liu HT, Lin YY, Lee IY, Shih WT, Yang PR, Tsai YY, Chang GH, Hsu CM, Yeh RA, Wu YH, Wu YH, Shen RC, Tsai MS. The anti-SARS-CoV-2 effect and mechanism of Chiehyuan herbal oral protection solution. Heliyon 2023; 9:e17701. [PMID: 37483781 PMCID: PMC10359827 DOI: 10.1016/j.heliyon.2023.e17701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/07/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
The Chiehyuan herbal oral protection solution (GB-2) is a herbal mixture commonly utilized in Taiwan for combating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as per traditional Chinese medicine practices. This study assessed the clinical impact of GB-2 through prospective clinical trials. With twice-daily use for a week, GB-2 was shown to diminish the expression of angiotensin-converting enzyme 2 (ACE2) in oral mucosal cells. Moreover, after two weeks of use, it could reduce transmembrane protease, serine 2 (TMRPSS2) expression in these cells. Additionally, in vitro experiments demonstrated that GB-2 lessened the entry efficiency of the Omicron, L452R-D614G, T478K-D614G, and L452R-T478K-D614G variants of the SARS-CoV-2 pseudotyped lentivirus. It also impeded the interaction between ACE2 and the receptor-binding domain (RBD) presenting N501Y-K417N-E484A-G339D-Q493R-G496S-Q498R and L452R-T478K mutations. Glycyrrhizic acid, a major compound in GB-2, also hindered the entry of the Omicron variant (BA.1) of the SARS-CoV-2 pseudotyped lentivirus by obstructing the binding between ACE2 and the RBD presenting the N501Y-K417N-E484A-G339D-Q493R-G496S-Q498R mutation. To sum up, these findings suggest that GB-2 can decrease ACE2 and TMPRSS2 expression in oral mucosal cells. Both glycyrrhizic acid and GB-2 were found to reduce the entry efficiency of the Omicron variant (BA.1) of the SARS-CoV-2 pseudotyped lentivirus and block the binding between ACE2 and the RBD with the N501Y-K417N-E484A-G339D-Q493R-G496S-Q498R mutation. This evidence implies that GB-2 might be a potential candidate for further study as a preventative measure against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ching-Yuan Wu
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Yao-Hsu Yang
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Shih Lin
- Department of Pharmacy, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Li-Hsin Shu
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yu-Ching Cheng
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- Department of Otolaryngology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Hung-Te Liu
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yin-Yin Lin
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - I-Yun Lee
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Wei-Tai Shih
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Pei-Rung Yang
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ying-Ying Tsai
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Geng-He Chang
- Department of Otolaryngology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- Faculty of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Health Information and Epidemiology Laboratory, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Cheng-Ming Hsu
- Department of Otolaryngology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- Faculty of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Reming-Albert Yeh
- Department of Otolaryngology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yu-Huei Wu
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Heng Wu
- Department of Electrical Engineering, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Rou-Chen Shen
- Department of Otolaryngology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ming-Shao Tsai
- Department of Otolaryngology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- Faculty of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
103
|
de Vries EM, Cogan NOI, Gubala AJ, Rodoni BC, Lynch SE. Fine-scale genomic tracking of Ross River virus using nanopore sequencing. Parasit Vectors 2023; 16:186. [PMID: 37280650 PMCID: PMC10243270 DOI: 10.1186/s13071-023-05734-z] [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: 11/29/2022] [Accepted: 03/11/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Ross River virus (RRV) is Australia's most common and widespread mosquito-transmitted arbovirus and is of significant public health concern. With increasing anthropogenic impacts on wildlife and mosquito populations, it is important that we understand how RRV circulates in its endemic hotspots to determine where public health efforts should be directed. Current surveillance methods are effective in locating the virus but do not provide data on the circulation of the virus and its strains within the environment. This study examined the ability to identify single nucleotide polymorphisms (SNPs) within the variable E2/E3 region by generating full-length haplotypes from a range of mosquito trap-derived samples. METHODS A novel tiled primer amplification workflow for amplifying RRV was developed with analysis using Oxford Nanopore Technology's MinION and a custom ARTIC/InterARTIC bioinformatic protocol. By creating a range of amplicons across the whole genome, fine-scale SNP analysis was enabled by specifically targeting the variable region that was amplified as a single fragment and established haplotypes that informed spatial-temporal variation of RRV in the study site in Victoria. RESULTS A bioinformatic and laboratory pipeline was successfully designed and implemented on mosquito whole trap homogenates. Resulting data showed that genotyping could be conducted in real time and that whole trap consensus of the viruses (with major SNPs) could be determined in a timely manner. Minor variants were successfully detected from the variable E2/E3 region of RRV, which allowed haplotype determination within complex mosquito homogenate samples. CONCLUSIONS The novel bioinformatic and wet laboratory methods developed here will enable fast detection and characterisation of RRV isolates. The concepts presented in this body of work are transferable to other viruses that exist as quasispecies in samples. The ability to detect minor SNPs, and thus haplotype strains, is critically important for understanding the epidemiology of viruses their natural environment.
Collapse
Affiliation(s)
- Ellen M. de Vries
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC 3083 Australia
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC 3083 Australia
| | - Noel O. I. Cogan
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC 3083 Australia
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC 3083 Australia
| | - Aneta J. Gubala
- Sensors and Effectors Division, Defence Science & Technology Group, Fishermans Bend, VIC 3207 Australia
| | - Brendan C. Rodoni
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC 3083 Australia
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC 3083 Australia
| | - Stacey E. Lynch
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC 3083 Australia
| |
Collapse
|
104
|
Jalal D, Samir O, Elzayat MG, El-Shqanqery HE, Diab AA, ElKaialy L, Mohammed AM, Hamdy D, Matar IK, Amer K, Elnakib M, Hassan W, Mansour T, Soliman S, Hassan R, Al-Toukhy GM, Hammad M, Abdo I, Sayed AA. Genomic characterization of SARS-CoV-2 in Egypt: insights into spike protein thermodynamic stability. Front Microbiol 2023; 14:1190133. [PMID: 37333655 PMCID: PMC10273679 DOI: 10.3389/fmicb.2023.1190133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/09/2023] [Indexed: 06/20/2023] Open
Abstract
The overall pattern of the SARS-CoV-2 pandemic so far has been a series of waves; surges in new cases followed by declines. The appearance of novel mutations and variants underlie the rises in infections, making surveillance of SARS-CoV-2 mutations and prediction of variant evolution of utmost importance. In this study, we sequenced 320 SARS-CoV-2 viral genomes isolated from patients from the outpatient COVID-19 clinic in the Children's Cancer Hospital Egypt 57357 (CCHE 57357) and the Egypt Center for Research and Regenerative Medicine (ECRRM). The samples were collected between March and December 2021, covering the third and fourth waves of the pandemic. The third wave was found to be dominated by Nextclade 20D in our samples, with a small number of alpha variants. The delta variant was found to dominate the fourth wave samples, with the appearance of omicron variants late in 2021. Phylogenetic analysis reveals that the omicron variants are closest genetically to early pandemic variants. Mutation analysis shows SNPs, stop codon mutation gain, and deletion/insertion mutations, with distinct patterns of mutations governed by Nextclade or WHO variant. Finally, we observed a large number of highly correlated mutations, and some negatively correlated mutations, and identified a general inclination toward mutations that lead to enhanced thermodynamic stability of the spike protein. Overall, this study contributes genetic and phylogenetic data, as well as provides insights into SARS-CoV-2 viral evolution that may eventually help in the prediction of evolving mutations for better vaccine development and drug targets.
Collapse
Affiliation(s)
- Deena Jalal
- Department of Basic Research, Genomics and Epigenomics Program, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Omar Samir
- Department of Basic Research, Genomics and Epigenomics Program, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Mariam G. Elzayat
- Department of Basic Research, Genomics and Epigenomics Program, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Hend E. El-Shqanqery
- Department of Basic Research, Genomics and Epigenomics Program, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Aya A. Diab
- Department of Basic Research, Genomics and Epigenomics Program, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Lamiaa ElKaialy
- Department of Basic Research, Genomics and Epigenomics Program, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Aya M. Mohammed
- Department of Basic Research, Genomics and Epigenomics Program, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Donia Hamdy
- Department of Basic Research, Genomics and Epigenomics Program, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Islam K. Matar
- Department of Basic Research, Genomics and Epigenomics Program, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
- Department of Chemistry, Saint Mary’s University, Halifax, NS, Canada
| | - Khaled Amer
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Mostafa Elnakib
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Wael Hassan
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Tarek Mansour
- Department of Virology and Immunology, National Cancer Institute, Cairo University, Cairo, Egypt
- Department of Clinical Pathology, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Sonia Soliman
- Department of Clinical Pathology, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Reem Hassan
- Department of Clinical and Chemical Pathology, Kasr Al-Aini School of Medicine, Cairo University, Cairo, Egypt
- Molecular Microbiology Unit, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Ghada M. Al-Toukhy
- Department of Virology and Immunology, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Mahmoud Hammad
- Department of Pediatric Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
- Department of Pediatric Oncology, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Ibrahim Abdo
- Department of Clinical Pharmacy, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Ahmed A. Sayed
- Department of Basic Research, Genomics and Epigenomics Program, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
- Faculty of Science, Department of Biochemistry, Ain Shams University, Cairo, Egypt
| |
Collapse
|
105
|
Singh RS, Singh A, Masih GD, Batra G, Sharma AR, Joshi R, Prakash A, Suroy B, Sarma P, Prajapat M, Kaur H, Bhattacharyya A, Upadhyay S, Medhi B. A comprehensive insight on the challenges for COVID-19 vaccine: A lesson learnt from other viral vaccines. Heliyon 2023; 9:e16813. [PMID: 37303517 PMCID: PMC10245239 DOI: 10.1016/j.heliyon.2023.e16813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/15/2023] [Accepted: 05/29/2023] [Indexed: 06/13/2023] Open
Abstract
The aim of this study is to comprehensively analyze previous viral vaccine programs and identify potential challenges and effective measures for the COVID-19 vaccine program. Previous viral vaccine programs, such as those for HIV, Zika, Influenza, Ebola, Dengue, SARS, and MERS, were evaluated. Paramount challenges were identified, including quasi-species, cross-reactivity, duration of immunity, revaccination, mutation, immunosenescence, and adverse events related to viral vaccines. Although a large population has been vaccinated, mutations in SARS-CoV-2 and adverse events related to vaccines pose significant challenges. Previous vaccine programs have taught us that predicting the final outcome of the current vaccine program for COVID-19 cannot be determined at a given state. Long-term follow-up studies are essential. Validated preclinical studies, long-term follow-up studies, alternative therapeutic approaches, and alternative vaccines are necessary.
Collapse
Affiliation(s)
- Rahul Soloman Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Ashutosh Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Gladson David Masih
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Gitika Batra
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Amit Raj Sharma
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Rupa Joshi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Ajay Prakash
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Benjamin Suroy
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Phulen Sarma
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Manisha Prajapat
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Hardeep Kaur
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Anusuya Bhattacharyya
- Department of Ophthalmology, Government Medical College & Hospital, Sector-32, Chandigarh, 160030, India
| | - Sujata Upadhyay
- Department of Physiology, Dr. Harvansh Singh Judge Institute of Dental Sciences & Hospital, Panjab University, Chandigarh, 160014, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| |
Collapse
|
106
|
Nicoletto G, Richter SN, Frasson I. Presence, Location and Conservation of Putative G-Quadruplex Forming Sequences in Arboviruses Infecting Humans. Int J Mol Sci 2023; 24:ijms24119523. [PMID: 37298474 DOI: 10.3390/ijms24119523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023] Open
Abstract
Guanine quadruplexes (G4s) are non-canonical nucleic acid structures formed by guanine (G)-rich tracts that assemble into a core of stacked planar tetrads. G4s are found in the human genome and in the genomes of human pathogens, where they are involved in the regulation of gene expression and genome replication. G4s have been proposed as novel pharmacological targets in humans and their exploitation for antiviral therapy is an emerging research topic. Here, we report on the presence, conservation and localization of putative G4-forming sequences (PQSs) in human arboviruses. The prediction of PQSs was performed on more than twelve thousand viral genomes, belonging to forty different arboviruses that infect humans, and revealed that the abundance of PQSs in arboviruses is not related to the genomic GC content, but depends on the type of nucleic acid that constitutes the viral genome. Positive-strand ssRNA arboviruses, especially Flaviviruses, are significantly enriched in highly conserved PQSs, located in coding sequences (CDSs) or untranslated regions (UTRs). In contrast, negative-strand ssRNA and dsRNA arboviruses contain few conserved PQSs. Our analyses also revealed the presence of bulged PQSs, accounting for 17-26% of the total predicted PQSs. The data presented highlight the presence of highly conserved PQS in human arboviruses and present non-canonical nucleic acid-structures as promising therapeutic targets in arbovirus infections.
Collapse
Affiliation(s)
- Giulia Nicoletto
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy
| | - Sara N Richter
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy
| | - Ilaria Frasson
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy
| |
Collapse
|
107
|
Caspi I, Meir M, Ben Nun N, Abu Rass R, Yakhini U, Stern A, Ram Y. Mutation rate, selection, and epistasis inferred from RNA virus haplotypes via neural posterior estimation. Virus Evol 2023; 9:vead033. [PMID: 37305706 PMCID: PMC10256221 DOI: 10.1093/ve/vead033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/30/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
RNA viruses are particularly notorious for their high levels of genetic diversity, which is generated through the forces of mutation and natural selection. However, disentangling these two forces is a considerable challenge, and this may lead to widely divergent estimates of viral mutation rates, as well as difficulties in inferring the fitness effects of mutations. Here, we develop, test, and apply an approach aimed at inferring the mutation rate and key parameters that govern natural selection, from haplotype sequences covering full-length genomes of an evolving virus population. Our approach employs neural posterior estimation, a computational technique that applies simulation-based inference with neural networks to jointly infer multiple model parameters. We first tested our approach on synthetic data simulated using different mutation rates and selection parameters while accounting for sequencing errors. Reassuringly, the inferred parameter estimates were accurate and unbiased. We then applied our approach to haplotype sequencing data from a serial passaging experiment with the MS2 bacteriophage, a virus that parasites Escherichia coli. We estimated that the mutation rate of this phage is around 0.2 mutations per genome per replication cycle (95% highest density interval: 0.051-0.56). We validated this finding with two different approaches based on single-locus models that gave similar estimates but with much broader posterior distributions. Furthermore, we found evidence for reciprocal sign epistasis between four strongly beneficial mutations that all reside in an RNA stem loop that controls the expression of the viral lysis protein, responsible for lysing host cells and viral egress. We surmise that there is a fine balance between over- and underexpression of lysis that leads to this pattern of epistasis. To recap, we have developed an approach for joint inference of the mutation rate and selection parameters from full haplotype data with sequencing errors and used it to reveal features governing MS2 evolution.
Collapse
Affiliation(s)
- Itamar Caspi
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, P.O. Box 39040, Tel Aviv 6997801, Israel
| | - Moran Meir
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, P.O. Box 39040, Tel Aviv 6997801, Israel
| | - Nadav Ben Nun
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, P.O. Box 39040, Tel Aviv 6997801, Israel
- School of Zoology, Faculty of Life Sciences, Tel Aviv University, P.O. Box 39040, Tel Aviv 6997801, Israel
| | | | - Uri Yakhini
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, P.O. Box 39040, Tel Aviv 6997801, Israel
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, P.O. Box 39040, Tel Aviv 6997801, Israel
| | | | - Yoav Ram
- *Corresponding author: E-mail: ;
| |
Collapse
|
108
|
Panja A, Roy J, Mazumder A, Choudhury SM. Divergent mutations of Delta and Omicron variants: key players behind differential viral attributes across the COVID-19 waves. Virusdisease 2023:1-14. [PMID: 37363365 PMCID: PMC10171727 DOI: 10.1007/s13337-023-00823-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/17/2023] [Indexed: 06/28/2023] Open
Abstract
The third SARS-CoV-2 pandemic wave causing Omicron variant has comparatively higher replication rate and transmissibility than the second wave-causing Delta variant. The exact mechanism behind the differential properties of Delta and Omicron in respect to infectivity and virulence is not properly understood yet. This study reports the analysis of different mutations within the receptor binding domain (RBD) of spike glycoprotein and non-structural protein (nsp) of Delta and Omicron strains. We have used computational studies to evaluate the properties of Delta and Omicron variants in this work. Q498R, Q493R and S375F mutations of RBD showed better docking scores for Omicron compared to Delta variant of SARS-CoV-2, whereas nsp3_L1266I with PARP15 (7OUX), nsp3_L1266I with PARP15 (7OUX), and nsp6_G107 with ISG15 (1Z2M) showed significantly higher docking score. The findings of the present study might be helpful to reveal the probable cause of relatively milder form of COVID-19 disease manifested by Omicron in comparison to Delta variant of SARS-CoV-2 virus. Supplementary Information The online version contains supplementary material available at 10.1007/s13337-023-00823-0.
Collapse
Affiliation(s)
- Amrita Panja
- Biochemistry, Molecular Endocrinology, and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Paschim Medinipore, Midnapore, West Bengal 721102 India
| | - Jayita Roy
- National Institute of Biomedical Genomics (NIBMG), Nadia, Kalyani, West Bengal 741251 India
| | - Anup Mazumder
- National Institute of Biomedical Genomics (NIBMG), Nadia, Kalyani, West Bengal 741251 India
| | - Sujata Maiti Choudhury
- Biochemistry, Molecular Endocrinology, and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Paschim Medinipore, Midnapore, West Bengal 721102 India
| |
Collapse
|
109
|
Ren R, Zheng L, Han J, Perdoncini Carvalho C, Miyashita S, Zhang D, Qu F. Intracellular bottlenecking permits no more than three tomato yellow leaf curl virus genomes to initiate replication in a single cell. PLoS Pathog 2023; 19:e1011365. [PMID: 37126519 PMCID: PMC10174518 DOI: 10.1371/journal.ppat.1011365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 05/11/2023] [Accepted: 04/17/2023] [Indexed: 05/02/2023] Open
Abstract
Viruses are constantly subject to natural selection to enrich beneficial mutations and weed out deleterious ones. However, it remains unresolved as to how the phenotypic gains or losses brought about by these mutations cause the viral genomes carrying the very mutations to become more or less numerous. Previous investigations by us and others suggest that viruses with plus strand (+) RNA genomes may compel such selection by bottlenecking the replicating genome copies in each cell to low single digits. Nevertheless, it is unclear if similarly stringent reproductive bottlenecks also occur in cells invaded by DNA viruses. Here we investigated whether tomato yellow leaf curl virus (TYLCV), a small virus with a single-stranded DNA genome, underwent population bottlenecking in cells of its host plants. We engineered a TYLCV genome to produce two replicons that express green fluorescent protein and mCherry, respectively, in a replication-dependent manner. We found that among the cells entered by both replicons, less than 65% replicated both, whereas at least 35% replicated either of them alone. Further probability computation concluded that replication in an average cell was unlikely to have been initiated with more than three replicon genome copies. Furthermore, sequential inoculations unveiled strong mutual exclusions of these two replicons at the intracellular level. In conclusion, the intracellular population of the small DNA virus TYLCV is actively bottlenecked, and such bottlenecking may be a virus-encoded, evolutionarily conserved trait that assures timely selection of new mutations emerging through error-prone replication.
Collapse
Affiliation(s)
- Ruifan Ren
- Longping Branch, College of Biology, Hunan University, Changsha, China
- Department of Plant Pathology, The Ohio State University, Wooster, Ohio, United States of America
- Hunan Plant Protection Institute, Changsha, China
| | - Limin Zheng
- Department of Plant Pathology, The Ohio State University, Wooster, Ohio, United States of America
| | - Junping Han
- Department of Plant Pathology, The Ohio State University, Wooster, Ohio, United States of America
| | | | - Shuhei Miyashita
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Deyong Zhang
- Longping Branch, College of Biology, Hunan University, Changsha, China
- Hunan Plant Protection Institute, Changsha, China
| | - Feng Qu
- Department of Plant Pathology, The Ohio State University, Wooster, Ohio, United States of America
| |
Collapse
|
110
|
De A, Bhattacharya S, Debroy B, Bhattacharya A, Pal K. Exploring the pharmacological aspects of natural phytochemicals against SARS-CoV-2 Nsp14 through an in silico approach. In Silico Pharmacol 2023; 11:12. [PMID: 37131867 PMCID: PMC10141836 DOI: 10.1007/s40203-023-00143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 03/13/2023] [Indexed: 05/04/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), possesses an important bifunctional nonstructural protein (nsp14) with a C-terminal N7-methyltransferase (N7-MTase) domain and an N-terminal domain with exoribonuclease (ExoN) activity that is required for maintaining high-fidelity viral replication. Viruses use the error-prone replication mechanism, which results in high mutation rates, to adapt quickly to stressful situations. The efficiency with which nsp14 removes mismatched nucleotides due to the presence of ExoN activity protects viruses from mutagenesis. We investigated the pharmacological role of the phytochemicals (Baicalein, Bavachinin, Emodin, Kazinol F, Lycorine, Sinigrin, Procyanidin A2, Tanshinone IIA, Tanshinone IIB, Tomentin A, and Tomentin E) against the highly conserved nsp14 protein using docking-based computational analyses in search of new potential natural drug targets. The selected eleven phytochemicals failed to bind the active site of N7-Mtase in the global docking study, while the local docking study identified the top five phytochemicals with high binding energy scores ranging from - 9.0 to - 6.4 kcal/mol. Procyanidin A2 and Tomentin A showed the highest docking score of - 9.0 and - 8.1 kcal/mol, respectively. Local docking of isoform variants was also conducted, yielding the top five phytochemicals, with Procyanidin A1 having the highest binding energy value of - 9.1 kcal/mol. The phytochemicals were later tested for pharmacokinetics and pharmacodynamics analysis for Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) which resulted in choosing Tomentin A as a potential candidate. The molecular dynamics simulations studies of nsp14 revealed significant conformational changes upon complex formation with the identified compound, implying that these phytochemicals could be used as safe nutraceuticals which will impart long-term immunological competence in the human population against CoVs. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s40203-023-00143-7.
Collapse
Affiliation(s)
- Arkajit De
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal 700126 India
| | - Somdatta Bhattacharya
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal 700126 India
- Cancer Biology Laboratory, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal 700126 India
| | - Bishal Debroy
- Department of Biological Sciences, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal 700126 India
| | - Arijit Bhattacharya
- Department of Biological Sciences, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal 700126 India
- Anti-Microbial Resistance Laboratory, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal 700126 India
| | - Kuntal Pal
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal 700126 India
- Cancer Biology Laboratory, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal 700126 India
| |
Collapse
|
111
|
Krishnan K, Katneni VK, Prabhudas SK, Kaikkolante N, Jangam AK, Katneni UK, Hauton C, Peruzza L, Mudagandur SS, Koyadan VK, Poochirian JK, Jena J. MRF: a tool to overcome the barrier of inconsistent genome annotations and perform comparative genomics studies for the largest animal DNA virus. Virol J 2023; 20:72. [PMID: 37072853 PMCID: PMC10111743 DOI: 10.1186/s12985-023-02035-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/09/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND The genome of the largest known animal virus, the white spot syndrome virus (WSSV) responsible for huge economic losses and loss of employment in aquaculture, suffers from inconsistent annotation nomenclature. Novel genome sequence, circular genome and variable genome length led to nomenclature inconsistencies. Since vast knowledge has already accumulated in the past two decades with inconsistent nomenclature, the insights gained on a genome could not be easily extendable to other genomes. Therefore, the present study aims to perform comparative genomics studies in WSSV on uniform nomenclature. METHODS We have combined the standard mummer tool with custom scripts to develop missing regions finder (MRF) that documents the missing genome regions and coding sequences in virus genomes in comparison to a reference genome and in its annotation nomenclature. The procedure was implemented as web tool and in command-line interface. Using MRF, we have documented the missing coding sequences in WSSV and explored their role in virulence through application of phylogenomics, machine learning models and homologous genes. RESULTS We have tabulated and depicted the missing genome regions, missing coding sequences and deletion hotspots in WSSV on a common annotation nomenclature and attempted to link them to virus virulence. It was observed that the ubiquitination, transcription regulation and nucleotide metabolism might be essentially required for WSSV pathogenesis; and the structural proteins, VP19, VP26 and VP28 are essential for virus assembly. Few minor structural proteins in WSSV would act as envelope glycoproteins. We have also demonstrated the advantage of MRF in providing detailed graphic/tabular output in less time and also in handling of low-complexity, repeat-rich and highly similar regions of the genomes using other virus cases. CONCLUSIONS Pathogenic virus research benefits from tools that could directly indicate the missing genomic regions and coding sequences between isolates/strains. In virus research, the analyses performed in this study provides an advancement to find the differences between genomes and to quickly identify the important coding sequences/genomes that require early attention from researchers. To conclude, the approach implemented in MRF complements similarity-based tools in comparative genomics involving large, highly-similar, length-varying and/or inconsistently annotated viral genomes.
Collapse
Affiliation(s)
- Karthic Krishnan
- Centre for Bioinformatics, Nutrition Genetics and Biotechnology Division, ICAR - Central Institute of Brackishwater Aquaculture, 75, Santhome High Road, MRC Nagar, RA Puram, Chennai, Tamil Nadu, 600028, India
| | - Vinaya Kumar Katneni
- Centre for Bioinformatics, Nutrition Genetics and Biotechnology Division, ICAR - Central Institute of Brackishwater Aquaculture, 75, Santhome High Road, MRC Nagar, RA Puram, Chennai, Tamil Nadu, 600028, India.
| | - Sudheesh K Prabhudas
- Centre for Bioinformatics, Nutrition Genetics and Biotechnology Division, ICAR - Central Institute of Brackishwater Aquaculture, 75, Santhome High Road, MRC Nagar, RA Puram, Chennai, Tamil Nadu, 600028, India
| | - Nimisha Kaikkolante
- Centre for Bioinformatics, Nutrition Genetics and Biotechnology Division, ICAR - Central Institute of Brackishwater Aquaculture, 75, Santhome High Road, MRC Nagar, RA Puram, Chennai, Tamil Nadu, 600028, India
| | - Ashok Kumar Jangam
- Centre for Bioinformatics, Nutrition Genetics and Biotechnology Division, ICAR - Central Institute of Brackishwater Aquaculture, 75, Santhome High Road, MRC Nagar, RA Puram, Chennai, Tamil Nadu, 600028, India
| | - Upendra Kumar Katneni
- The Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, Maryland, USA
| | - Chris Hauton
- Ocean and Earth Science, National Oceanography Centre Southampton, University of Southampton Waterfront Campus, Southampton, UK
| | - Luca Peruzza
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, Padua, Italy
| | - Shashi Shekhar Mudagandur
- Centre for Bioinformatics, Nutrition Genetics and Biotechnology Division, ICAR - Central Institute of Brackishwater Aquaculture, 75, Santhome High Road, MRC Nagar, RA Puram, Chennai, Tamil Nadu, 600028, India
| | - Vijayan K Koyadan
- Centre for Bioinformatics, Nutrition Genetics and Biotechnology Division, ICAR - Central Institute of Brackishwater Aquaculture, 75, Santhome High Road, MRC Nagar, RA Puram, Chennai, Tamil Nadu, 600028, India
| | - Jithendran Karingalakkandy Poochirian
- Aquatic Animal Health and Environment Division, ICAR - Central Institute of Brackishwater Aquaculture, 75, Santhome High Road, MRC Nagar, RA Puram, Chennai, Tamil Nadu, 600028, India
| | | |
Collapse
|
112
|
Bass AL, Bateman AW, Kaukinen KH, Li S, Ming T, Patterson DA, Hinch SG, Miller KM. The spatial distribution of infectious agents in wild Pacific salmon along the British Columbia coast. Sci Rep 2023; 13:5473. [PMID: 37016008 PMCID: PMC10071257 DOI: 10.1038/s41598-023-32583-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 03/29/2023] [Indexed: 04/06/2023] Open
Abstract
Although infectious agents can act as strong population regulators, knowledge of their spatial distributions in wild Pacific salmon is limited, especially in the marine environment. Characterizing pathogen distributions during early marine residence, a period considered a survival bottleneck for Pacific salmon, may reveal where salmon populations are exposed to potentially detrimental pathogens. Using high-throughput qPCR, we determined the prevalence of 56 infectious agents in 5719 Chinook, 2032 Coho and 4062 Sockeye salmon, sampled between 2008 and 2018, in their first year of marine residence along coastal Western Canada. We identified high prevalence clusters, which often shifted geographically with season, for most of the 41 detected agents. A high density of infection clusters was found in the Salish Sea along the east coast of Vancouver Island, an important migration route and residence area for many salmon populations, some experiencing chronically poor marine survival. Maps for each infectious agent taxa showing clusters across all host species are provided. Our novel documentation of salmon pathogen distributions in the marine environment contributes to the ecological knowledge regarding some lesser known pathogens, identifies salmon populations potentially impacted by specific pathogens, and pinpoints priority locations for future research and remediation.
Collapse
Affiliation(s)
- Arthur L Bass
- Forest and Conservation Sciences, University of British Columbia, Vancouver, V6T 1Z4, Canada.
| | - Andrew W Bateman
- Pacific Salmon Foundation, Vancouver, V6J 4S6, Canada
- Ecology and Evolutionary Biology, University of Toronto, Toronto, M5S 1A1, Canada
| | - Karia H Kaukinen
- Fisheries and Oceans Canada, Pacific Biological Station, Nanaimo, V9T 6N7, Canada
| | - Shaorong Li
- Fisheries and Oceans Canada, Pacific Biological Station, Nanaimo, V9T 6N7, Canada
| | - Tobi Ming
- Fisheries and Oceans Canada, Pacific Biological Station, Nanaimo, V9T 6N7, Canada
| | - David A Patterson
- Fisheries and Oceans Canada, Science Branch, Pacific Region, School of Resource and Environmental Management, Simon Fraser University, Burnaby, V5A 1S6, Canada
| | - Scott G Hinch
- Forest and Conservation Sciences, University of British Columbia, Vancouver, V6T 1Z4, Canada
| | - Kristina M Miller
- Forest and Conservation Sciences, University of British Columbia, Vancouver, V6T 1Z4, Canada
- Fisheries and Oceans Canada, Pacific Biological Station, Nanaimo, V9T 6N7, Canada
| |
Collapse
|
113
|
Alizadehmohajer N, Zahedifar S, Sohrabi E, Shaddel Basir S, Nourigheimasi S, Falak R, Nedaeinia R, A Ferns G, Emami Nejad A, Manian M. Using In Silico Bioinformatics Algorithms for the Accurate Prediction of the Impact of Spike Protein Mutations on the Pathogenicity, Stability, and Functionality of the SARS-CoV-2 Virus and Analysis of Potential Therapeutic Targets. Biochem Genet 2023; 61:778-808. [PMID: 36173498 PMCID: PMC9521556 DOI: 10.1007/s10528-022-10282-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 09/01/2022] [Indexed: 11/02/2022]
Abstract
Coronavirus disease 2019 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We have used bioinformatics to investigate seventeen mutations in the spike protein of SARS-CoV-2, as this mediates infection of human cells and is the target of most vaccine strategies and antibody-based therapies. Two mutations, H146Y and S221W, were identified as being most pathogenic. Mutations at positions D614G, A829T, and P1263L might also have deleterious effects on protein function. We hypothesized that candidate small molecules may be repurposed to combat viral infection. We investigated changes in binding energies of the ligands and the mutant proteins by assessing molecular docking. For an understanding of cellular function and organization, protein-protein interactions are also critical. Protein-protein docking for naïve and mutated structures of SARS-CoV-2 S protein was evaluated for their binding energy with the angiotensin-converting enzyme 2 (ACE2). These interactions might limit the binding of the SARS-CoV-2 spike protein to the ACE2 receptor or may have a deleterious effect on protein function that may limit infection. These results may have important implications for the transmission of SARS-CoV-2, its pathogenesis, and the potential for drug repurposing and immune therapies.
Collapse
Affiliation(s)
- Negin Alizadehmohajer
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133, Milan, Italy
| | - Shahrzad Zahedifar
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ehsan Sohrabi
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sedighe Shaddel Basir
- Department of Microbiology, Faculty of New Sciences and Technologies Branch, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Falmer, Brighton, BN1 9PH, Sussex, UK
| | - Asieh Emami Nejad
- Department of Biology, Payame Noor University (PNU), P.O.Box 19395-3697, Tehran, Iran.
| | - Mostafa Manian
- Department of Medical Laboratory Science, Faculty of Medical Science, Kermanshah Branch, Imam Khomeini Campus, Islamic Azad University, Farhikhtegan Bld., Shahid J'afari St., 6718997551, Kermanshah, Iran.
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
114
|
Popovic M. Never ending story? Evolution of SARS-CoV-2 monitored through Gibbs energies of biosynthesis and antigen-receptor binding of Omicron BQ.1, BQ.1.1, XBB and XBB.1 variants. MICROBIAL RISK ANALYSIS 2023; 23:100250. [PMID: 36777740 PMCID: PMC9896887 DOI: 10.1016/j.mran.2023.100250] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 06/01/2023]
Abstract
RNA viruses exhibit a great tendency to mutate. Mutations occur in the parts of the genome that encode the spike glycoprotein and less often in the rest of the genome. This is why Gibbs energy of binding changes more than that of biosynthesis. Starting from 2019, the wild type that was labeled Hu-1 has during the last 3 years evolved to produce several dozen new variants, as a consequence of mutations. Mutations cause changes in empirical formulas of new virus strains, which lead to change in thermodynamic properties of biosynthesis and binding. These changes cause changes in the rate of reactions of binding of virus antigen to the host cell receptor and the rate of virus multiplication in the host cell. Changes in thermodynamic and kinetic parameters lead to changes in biological parameters of infectivity and pathogenicity. Since the beginning of the COVID-19 pandemic, SARS-CoV-2 has been evolving towards increase in infectivity and maintaining constant pathogenicity, or for some variants a slight decrease in pathogenicity. In the case of Omicron BQ.1, BQ.1.1, XBB and XBB.1 variants pathogenicity is identical as in the Omicron BA.2.75 variant. On the other hand, infectivity of the Omicron BQ.1, BQ.1.1, XBB and XBB.1 variants is greater than those of previous variants. This will most likely result in the phenomenon of asymmetric coinfection, that is circulation of several variants in the population, some being dominant.
Collapse
Affiliation(s)
- Marko Popovic
- School of Life Sciences, Technical University of Munich, Freising, Germany
| |
Collapse
|
115
|
Koopman G, Amacker M, Stegmann T, Verschoor EJ, Verstrepen BE, Bhoelan F, Bemelman D, Böszörményi KP, Fagrouch Z, Kiemenyi-Kayere G, Mortier D, Verel DE, Niphuis H, Acar RF, Kondova I, Kap YS, Bogers WMJM, Mooij P, Fleury S. A low dose of RBD and TLR7/8 agonist displayed on influenza virosome particles protects rhesus macaque against SARS-CoV-2 challenge. Sci Rep 2023; 13:5074. [PMID: 36977691 PMCID: PMC10044094 DOI: 10.1038/s41598-023-31818-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Influenza virosomes serve as antigen delivery vehicles and pre-existing immunity toward influenza improves the immune responses toward antigens. Here, vaccine efficacy was evaluated in non-human primates with a COVID-19 virosome-based vaccine containing a low dose of RBD protein (15 µg) and the adjuvant 3M-052 (1 µg), displayed together on virosomes. Vaccinated animals (n = 6) received two intramuscular administrations at week 0 and 4 and challenged with SARS-CoV-2 at week 8, together with unvaccinated control animals (n = 4). The vaccine was safe and well tolerated and serum RBD IgG antibodies were induced in all animals and in the nasal washes and bronchoalveolar lavages in the three youngest animals. All control animals became strongly sgRNA positive in BAL, while all vaccinated animals were protected, although the oldest vaccinated animal (V1) was transiently weakly positive. The three youngest animals had also no detectable sgRNA in nasal wash and throat. Cross-strain serum neutralizing antibodies toward Wuhan-like, Alpha, Beta, and Delta viruses were observed in animals with the highest serum titers. Pro-inflammatory cytokines IL-8, CXCL-10 and IL-6 were increased in BALs of infected control animals but not in vaccinated animals. Virosomes-RBD/3M-052 prevented severe SARS-CoV-2, as shown by a lower total lung inflammatory pathology score than control animals.
Collapse
Grants
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
Collapse
Affiliation(s)
- Gerrit Koopman
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands.
| | - Mario Amacker
- Mymetics SA, 4 Route de La Corniche, 1066, Epalinges, Switzerland
- Department for BioMedical Research DBMR, Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, 3008, Bern, Switzerland
| | - Toon Stegmann
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Ernst J Verschoor
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Babs E Verstrepen
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Farien Bhoelan
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Denzel Bemelman
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Kinga P Böszörményi
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Zahra Fagrouch
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | | | - Daniella Mortier
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Dagmar E Verel
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Henk Niphuis
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Roja Fidel Acar
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Ivanela Kondova
- Animal Science Department, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Yolanda S Kap
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Willy M J M Bogers
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Petra Mooij
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Sylvain Fleury
- Mymetics SA, 4 Route de La Corniche, 1066, Epalinges, Switzerland.
| |
Collapse
|
116
|
Haseeb M, Amir A, Ikram A. In Silico Analysis of SARS-CoV-2 Spike Proteins of Different Field Variants. Vaccines (Basel) 2023; 11:vaccines11040736. [PMID: 37112648 PMCID: PMC10145761 DOI: 10.3390/vaccines11040736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Coronaviruses belong to the group of RNA family of viruses that trigger diseases in birds, humans, and mammals, which can cause respiratory tract infections. The COVID-19 pandemic has badly affected every part of the world. Our study aimed to explore the genome of SARS-CoV-2, followed by in silico analysis of its proteins. Different nucleotide and protein variants of SARS-CoV-2 were retrieved from NCBI. Contigs and consensus sequences were developed to identify these variants using SnapGene. Data of the variants that significantly differed from each other was run through Predict Protein software to understand the changes produced in the protein structure. The SOPMA web server was used to predict the secondary structure of the proteins. Tertiary structure details of the selected proteins were analyzed using the web server SWISS-MODEL. Sequencing results showed numerous single nucleotide polymorphisms in the surface glycoprotein, nucleocapsid, ORF1a, and ORF1ab polyprotein while the envelope, membrane, ORF3a, ORF6, ORF7a, ORF8, and ORF10 genes had no or few SNPs. Contigs were used to identify variations in the Alpha and Delta variants of SARS-CoV-2 with the reference strain (Wuhan). Some of the secondary structures of the SARS-CoV-2 proteins were predicted by using Sopma software and were further compared with reference strains of SARS-CoV-2 (Wuhan) proteins. The tertiary structure details of only spike proteins were analyzed through the SWISS-MODEL and Ramachandran plots. Through the Swiss-model, a comparison of the tertiary structure model of the SARS-CoV-2 spike protein of the Alpha and Delta variants was made with the reference strain (Wuhan). Alpha and Delta variants of the SARS-CoV-2 isolates submitted in GISAID from Pakistan with changes in structural and nonstructural proteins were compared with the reference strain, and 3D structure mapping of the spike glycoprotein and mutations in the amino acids were seen. The surprisingly increased rate of SARS-CoV-2 transmission has forced numerous countries to impose a total lockdown due to an unusual occurrence. In this research, we employed in silico computational tools to analyze the SARS-CoV-2 genomes worldwide to detect vital variations in structural proteins and dynamic changes in all SARS-CoV-2 proteins, mainly spike proteins, produced due to many mutations. Our analysis revealed substantial differences in the functionality, immunological, physicochemical, and structural variations in the SARS-CoV-2 isolates. However, the real impact of these SNPs can only be determined further by experiments. Our results can aid in vivo and in vitro experiments in the future.
Collapse
|
117
|
Farlow A, Torreele E, Gray G, Ruxrungtham K, Rees H, Prasad S, Gomez C, Sall A, Magalhães J, Olliaro P, Terblanche P. The Future of Epidemic and Pandemic Vaccines to Serve Global Public Health Needs. Vaccines (Basel) 2023; 11:vaccines11030690. [PMID: 36992275 DOI: 10.3390/vaccines11030690] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/31/2023] Open
Abstract
This Review initiates a wide-ranging discussion over 2023 by selecting and exploring core themes to be investigated more deeply in papers submitted to the Vaccines Special Issue on the "Future of Epidemic and Pandemic Vaccines to Serve Global Public Health Needs". To tackle the SARS-CoV-2 pandemic, an acceleration of vaccine development across different technology platforms resulted in the emergency use authorization of multiple vaccines in less than a year. Despite this record speed, many limitations surfaced including unequal access to products and technologies, regulatory hurdles, restrictions on the flow of intellectual property needed to develop and manufacture vaccines, clinical trials challenges, development of vaccines that did not curtail or prevent transmission, unsustainable strategies for dealing with variants, and the distorted allocation of funding to favour dominant companies in affluent countries. Key to future epidemic and pandemic responses will be sustainable, global-public-health-driven vaccine development and manufacturing based on equitable access to platform technologies, decentralised and localised innovation, and multiple developers and manufacturers, especially in low- and middle-income countries (LMICs). There is talk of flexible, modular pandemic preparedness, of technology access pools based on non-exclusive global licensing agreements in exchange for fair compensation, of WHO-supported vaccine technology transfer hubs and spokes, and of the creation of vaccine prototypes ready for phase I/II trials, etc. However, all these concepts face extraordinary challenges shaped by current commercial incentives, the unwillingness of pharmaceutical companies and governments to share intellectual property and know-how, the precariousness of building capacity based solely on COVID-19 vaccines, the focus on large-scale manufacturing capacity rather than small-scale rapid-response innovation to stop outbreaks when and where they occur, and the inability of many resource-limited countries to afford next-generation vaccines for their national vaccine programmes. Once the current high subsidies are gone and interest has waned, sustaining vaccine innovation and manufacturing capability in interpandemic periods will require equitable access to vaccine innovation and manufacturing capabilities in all regions of the world based on many vaccines, not just "pandemic vaccines". Public and philanthropic investments will need to leverage enforceable commitments to share vaccines and critical technology so that countries everywhere can establish and scale up vaccine development and manufacturing capability. This will only happen if we question all prior assumptions and learn the lessons offered by the current pandemic. We invite submissions to the special issue, which we hope will help guide the world towards a global vaccine research, development, and manufacturing ecosystem that better balances and integrates scientific, clinical trial, regulatory, and commercial interests and puts global public health needs first.
Collapse
Affiliation(s)
- Andrew Farlow
- Nuffield Department of Medicine, University of Oxford, Broad St., Oxford OX1 3BD, UK
- Oxford Martin School, University of Oxford, Broad St., Oxford OX1 3BD, UK
| | - Els Torreele
- Independent Consultant and Institute for Innovation & Public Purpose (IIPP), University College London, London WC1E 6BT, UK
| | - Glenda Gray
- Office of the President, South African Medical Research Council (SAMRC), Tygerberg 7050, South Africa
| | - Kiat Ruxrungtham
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Bangkok 10330, Thailand
- School of Global Health (SGH), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Helen Rees
- Wits RHI, University of Witwatersrand, Johannesburg 2050, South Africa
| | - Sai Prasad
- Bharat Biotech International Limited, Genome Valley, Shameerpet, Hyderabad 500 078, India
| | - Carolina Gomez
- Facultad de Derecho, Universidad Nacional de Colombia, Cra 45, Bogotá 111321, Colombia
| | - Amadou Sall
- Virology Department, Institut Pasteur de Dakar, 36, Avenue Pasteur, Dakar 10200, Senegal
| | - Jorge Magalhães
- Centre for Technological Innovation, Institute of Drugs Technology-Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro 21041-210, Brazil
| | - Piero Olliaro
- ISARIC Global Support Centre International Severe Acute Respiratory and Emerging Infection Consortium, Pandemic Sciences Institute, University of Oxford, Oxford OX1 3BD, UK
| | | |
Collapse
|
118
|
Roa-Linares VC, Escudero-Flórez M, Vicente-Manzanares M, Gallego-Gómez JC. Host Cell Targets for Unconventional Antivirals against RNA Viruses. Viruses 2023; 15:v15030776. [PMID: 36992484 PMCID: PMC10058429 DOI: 10.3390/v15030776] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/12/2023] [Accepted: 02/28/2023] [Indexed: 03/31/2023] Open
Abstract
The recent COVID-19 crisis has highlighted the importance of RNA-based viruses. The most prominent members of this group are SARS-CoV-2 (coronavirus), HIV (human immunodeficiency virus), EBOV (Ebola virus), DENV (dengue virus), HCV (hepatitis C virus), ZIKV (Zika virus), CHIKV (chikungunya virus), and influenza A virus. With the exception of retroviruses which produce reverse transcriptase, the majority of RNA viruses encode RNA-dependent RNA polymerases which do not include molecular proofreading tools, underlying the high mutation capacity of these viruses as they multiply in the host cells. Together with their ability to manipulate the immune system of the host in different ways, their high mutation frequency poses a challenge to develop effective and durable vaccination and/or treatments. Consequently, the use of antiviral targeting agents, while an important part of the therapeutic strategy against infection, may lead to the selection of drug-resistant variants. The crucial role of the host cell replicative and processing machinery is essential for the replicative cycle of the viruses and has driven attention to the potential use of drugs directed to the host machinery as therapeutic alternatives to treat viral infections. In this review, we discuss small molecules with antiviral effects that target cellular factors in different steps of the infectious cycle of many RNA viruses. We emphasize the repurposing of FDA-approved drugs with broad-spectrum antiviral activity. Finally, we postulate that the ferruginol analog (18-(phthalimide-2-yl) ferruginol) is a potential host-targeted antiviral.
Collapse
Affiliation(s)
- Vicky C Roa-Linares
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Manuela Escudero-Flórez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain
| | - Juan C Gallego-Gómez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| |
Collapse
|
119
|
Mayer ADM, Gröhs Ferrareze PA, de Oliveira LFV, Gregianini TS, Neves CLAM, Caldana GD, Kmetzsch L, Thompson CE. Genomic characterization and molecular evolution of SARS-CoV-2 in Rio Grande do Sul State, Brazil. Virology 2023; 582:1-11. [PMID: 36989935 PMCID: PMC10018445 DOI: 10.1016/j.virol.2023.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023]
Abstract
SARS-CoV-2 is the virus responsible for the COVID-19 and has afflicted the world since the end of 2019. Different lineages have been discovered and the Gamma lineage, which started the second wave of infections, was first described in Brazil, one of the most affected countries by pandemic. Therefore, this study analyzed SARS-CoV-2 sequenced genomes from Esteio city in Rio Grande do Sul, Southern Brazil. We also comparatively analyzed genomes of the two first years of the pandemic from Rio Grande do Sul state for understanding their genomic and evolutionary patterns. The phylogenomic analysis showed monophyletic groups for Alpha, Gamma, Delta and Omicron, as well as for other circulating lineages in the state. Molecular evolutionary analysis identified several sites under adaptive selection in membrane and nucleocapsid proteins which could be related to a prevalent stabilizing effect on membrane protein structure, as well as majoritarily destabilizing effects on C-terminal nucleocapsid domain.
Collapse
Affiliation(s)
- Amanda de Menezes Mayer
- Center of Biotechnology, Graduate Program in Cell and Molecular Biology (PPGBCM), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Patrícia Aline Gröhs Ferrareze
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | | | - Tatiana Schäffer Gregianini
- Laboratório Central de Saúde Pública do Centro Estadual de Vigilância em Saúde da Secretaria de Saúde do Estado do Rio Grande do Sul (LACEN/CEVS/SES-RS), Porto Alegre, RS, Brazil
| | | | - Gabriel Dickin Caldana
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Lívia Kmetzsch
- Center of Biotechnology, Graduate Program in Cell and Molecular Biology (PPGBCM), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Claudia Elizabeth Thompson
- Center of Biotechnology, Graduate Program in Cell and Molecular Biology (PPGBCM), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Department of Pharmacosciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil.
| |
Collapse
|
120
|
Gattinger P, Ohradanova-Repic A, Valenta R. Importance, Applications and Features of Assays Measuring SARS-CoV-2 Neutralizing Antibodies. Int J Mol Sci 2023; 24:ijms24065352. [PMID: 36982424 PMCID: PMC10048970 DOI: 10.3390/ijms24065352] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/17/2023] Open
Abstract
More than three years ago, the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) caused the unforeseen COVID-19 pandemic with millions of deaths. In the meantime, SARS-CoV-2 has become endemic and is now part of the repertoire of viruses causing seasonal severe respiratory infections. Due to several factors, among them the development of SARS-CoV-2 immunity through natural infection, vaccination and the current dominance of seemingly less pathogenic strains belonging to the omicron lineage, the COVID-19 situation has stabilized. However, several challenges remain and the possible new occurrence of highly pathogenic variants remains a threat. Here we review the development, features and importance of assays measuring SARS-CoV-2 neutralizing antibodies (NAbs). In particular we focus on in vitro infection assays and molecular interaction assays studying the binding of the receptor binding domain (RBD) with its cognate cellular receptor ACE2. These assays, but not the measurement of SARS-CoV-2-specific antibodies per se, can inform us of whether antibodies produced by convalescent or vaccinated subjects may protect against the infection and thus have the potential to predict the risk of becoming newly infected. This information is extremely important given the fact that a considerable number of subjects, in particular vulnerable persons, respond poorly to the vaccination with the production of neutralizing antibodies. Furthermore, these assays allow to determine and evaluate the virus-neutralizing capacity of antibodies induced by vaccines and administration of plasma-, immunoglobulin preparations, monoclonal antibodies, ACE2 variants or synthetic compounds to be used for therapy of COVID-19 and assist in the preclinical evaluation of vaccines. Both types of assays can be relatively quickly adapted to newly emerging virus variants to inform us about the magnitude of cross-neutralization, which may even allow us to estimate the risk of becoming infected by newly appearing virus variants. Given the paramount importance of the infection and interaction assays we discuss their specific features, possible advantages and disadvantages, technical aspects and not yet fully resolved issues, such as cut-off levels predicting the degree of in vivo protection.
Collapse
Affiliation(s)
- Pia Gattinger
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Karl Landsteiner University, 3500 Krems an der Donau, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
- NRC Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
- Correspondence:
| |
Collapse
|
121
|
Sharma A, Krishna S, Sowdhamini R. Bioinformatics Analysis of Mutations Sheds Light on the Evolution of Dengue NS1 Protein With Implications in the Identification of Potential Functional and Druggable Sites. Mol Biol Evol 2023; 40:7043264. [PMID: 36795614 PMCID: PMC9989740 DOI: 10.1093/molbev/msad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/26/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Non-structural protein (NS1) is a 350 amino acid long conserved protein in the dengue virus. Conservation of NS1 is expected due to its importance in dengue pathogenesis. The protein is known to exist in dimeric and hexameric states. The dimeric state is involved in its interaction with host proteins and viral replication, and the hexameric state is involved in viral invasion. In this work, we performed extensive structure and sequence analysis of NS1 protein, and uncovered the role of NS1 quaternary states in its evolution. A three-dimensional modeling of unresolved loop regions in NS1 structure is performed. "Conserved" and "Variable" regions within NS1 protein were identified from sequences obtained from patient samples and the role of compensatory mutations in selecting destabilizing mutations were identified. Molecular dynamics (MD) simulations were performed to extensively study the effect of a few mutations on NS1 structure stability and compensatory mutations. Virtual saturation mutagenesis, predicting the effect of every individual amino acid substitution on NS1 stability sequentially, revealed virtual-conserved and variable sites. The increase in number of observed and virtual-conserved regions across NS1 quaternary states suggest the role of higher order structure formation in its evolutionary conservation. Our sequence and structure analysis could enable in identifying possible protein-protein interfaces and druggable sites. Virtual screening of nearly 10,000 small molecules, including FDA-approved drugs, permitted us to recognize six drug-like molecules targeting the dimeric sites. These molecules could be promising due to their stable interactions with NS1 throughout the simulation.
Collapse
Affiliation(s)
- Abhishek Sharma
- National Centre for Biological Science, TIFR, Bangalore, India
| | - Sudhir Krishna
- National Centre for Biological Science, TIFR, Bangalore, India.,Department of School of Interdisciplinary Life Sciences, Indian Institute of Technology Goa, Farmagudi, Pond-403401, Goa, India
| | - Ramanathan Sowdhamini
- National Centre for Biological Science, TIFR, Bangalore, India.,Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India.,Computational Biology, Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
| |
Collapse
|
122
|
Awan T, Khan KB, Mannan A. A compact CNN model for automated detection of COVID-19 using thorax x-ray images. JOURNAL OF INTELLIGENT & FUZZY SYSTEMS 2023. [DOI: 10.3233/jifs-223704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
COVID-19 is an epidemic, causing an enormous death toll. The mutational changing of an RNA virus is causing diagnostic complexities. RT-PCR and Rapid Tests are used for the diagnosis, but unfortunately, these methods are ineffective in diagnosing all strains of COVID-19. There is an utmost need to develop a diagnostic procedure for timely identification. In the proposed work, we come up with a lightweight algorithm based on deep learning to develop a rapid detection system for COVID-19 with thorax chest x-ray (CXR) images. This research aims to develop a fine-tuned convolutional neural network (CNN) model using improved EfficientNetB5. Design is based on compound scaling and trained on the best possible feature extraction algorithm. The low convergence rate of the proposed work can be easily deployed into limited computational resources. It will be helpful for the rapid triaging of victims. 2-fold cross-validation further improves the performance. The algorithm proposed is trained, validated, and testing is performed in the form of internal and external validation on a self-collected and compiled a real-time dataset of CXR. The training dataset is relatively extensive compared to the existing ones. The performance of the proposed technique is measured, validated, and compared with other state-of-the-art pre-trained models. The proposed methodology gives remarkable accuracy (99.5%) and recall (99.5%) for biclassification. The external validation using two different test dataset also give exceptional predictions. The visual depiction of predictions is represented by Grad-CAM maps, presenting the extracted features of the predicted results.
Collapse
Affiliation(s)
- Tehreem Awan
- Department of Electrical Engineering, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
- Department of Electrical Engineering, NFC Institute of Engineering & Technology Multan, Multan, Pakistan
| | - Khan Bahadar Khan
- Department of Electrical Engineering, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Abdul Mannan
- Department of Electrical Engineering, NFC Institute of Engineering & Technology Multan, Multan, Pakistan
| |
Collapse
|
123
|
Hosaka Y, Yan Y, Naito T, Oyama R, Tsuchiya K, Yamamoto N, Nojiri S, Hori S, Takahashi K, Tabe Y. SARS-CoV-2 evolution among patients with immunosuppression in a nosocomial cluster of a Japanese medical center during the Delta (AY.29 sublineage) surge. Front Microbiol 2023; 14:944369. [PMID: 36846745 PMCID: PMC9947280 DOI: 10.3389/fmicb.2023.944369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 01/11/2023] [Indexed: 02/11/2023] Open
Abstract
Background Previous studies have shown that patients with immunosuppression tend to have longer-lasting SARS-CoV-2 infections and a number of mutations were observed during the infection period. However, these studies were, in general, conducted longitudinally. Mutation evolution among groups of patients with immunosuppression have not been well studied, especially among Asian populations. Methods Our study targeted a nosocomial cluster of SARS-CoV-2 infection in a Japanese medical center during Delta surge (AY.29 sublineage), involving ward nurses and inpatients. Whole-genome sequencing analyses were performed to examine mutation changes. Haplotype and minor variant analyses were furtherly performed to detect the mutations on the viral genomes in detail. In addition, sequences of the first wild-type strain hCoV-19/Wuhan/WIV04/2019 and AY.29 wild-type strain hCoV-19/Japan/TKYK15779/2021 were used as references to assess the phylogenetical development of this cluster. Results A total of 6 nurses and 14 inpatients were identified as a nosocomial cluster from September 14 through 28, 2021. All were Delta variant (AY.29 sublineage) positive. 92.9% of infected patients (13 out of 14) were either cancer patients and/or receiving immunosuppressive or steroid treatments. Compared to AY.29 wild type, a total of 12 mutations were found in the 20 cases. Haplotype analysis found one index group of eight cases with F274F (N) mutation and 10 other haplotypes with one to three additional mutations. Furthermore, we found that cases with more than three minor variants were all cancer patients under immunosuppressive treatments. The phylogenetical tree analysis, including 20 nosocomial cluster-associated viral genomes, the first wild-type strain and the AY.29 wild-type strain as references, indicated the mutation development of the AY.29 virus in this cluster. Conclusion Our study of a nosocomial SARS-CoV-2 cluster highlights mutation acquisition during transmission. More importantly, it provided new evidence emphasizing the need to further improve infection control measures to prevent nosocomial infection among immunosuppressed patients.
Collapse
Affiliation(s)
- Yoshie Hosaka
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yan Yan
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Toshio Naito
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Research Support Utilizing Bioresource Bank, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Rieko Oyama
- Department of Research Support Utilizing Bioresource Bank, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Koji Tsuchiya
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Norio Yamamoto
- Department of Microbiology, Tokai University School of Medicine, Hiratsuka, Kanagawa, Japan
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Satoshi Hori
- Infection Control Unit, Juntendo University Hospital, Tokyo, Japan
| | - Kazuhiko Takahashi
- Department of Research Support Utilizing Bioresource Bank, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Research Support Utilizing Bioresource Bank, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
124
|
D'Addiego J, Elaldi N, Wand N, Osman K, Bagci BK, Kennedy E, Pektas AN, Hart E, Slack G, Hewson R. Investigating the effect of ribavirin treatment on genetic mutations in Crimean-Congo haemorrhagic fever virus (CCHFV) through next-generation sequencing. J Med Virol 2023; 95:e28548. [PMID: 36734067 DOI: 10.1002/jmv.28548] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/12/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Crimean-Congo haemorrhagic fever (CCHF) is the most widespread tick-borne viral haemorrhagic fever affecting humans, and yet a licensed drug against the virus (CCHFV) is still not available. While several studies have suggested the efficacy of ribavirin against CCHFV, current literature remains inconclusive. In this study, we have utilised next-generation sequencing to investigate the mutagenic effect of ribavirin on the CCHFV genome during clinical disease. Samples collected from CCHF patients receiving ribavirin treatment or supportive care only at Sivas Cumhuriyet University Hospital, Turkey, were analysed. By comparing the frequency of mutations in each group, we found little evidence of an overall mutagenic effect. This suggests that ribavirin, administered at the acute stages of CCHFV infection (at the World Health Organization-recommended dose) is unable to induce lethal mutagenesis that would cause an extinction event in the CCHFV population and reduce viremia.
Collapse
Affiliation(s)
- Jake D'Addiego
- UK Health Security Agency, Salisbury, UK
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Nazif Elaldi
- Department of Infectious Diseases and Clinical Microbiology, Sivas Cumhuriyet University Faculty of Medicine, Sivas, Turkey
| | | | | | - Binnur Koksal Bagci
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Sivas Cumhuriyet University, Sivas, Turkey
| | | | - Ayse Nur Pektas
- Cumhuriyet University Advanced Technology Application and Research Center (CUTAM), Sivas Cumhuriyet University, Sivas, Turkey
| | | | | | - Roger Hewson
- UK Health Security Agency, Salisbury, UK
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
125
|
Sequence analysis of SARS-CoV-2 Delta variant isolated from Makassar, South Sulawesi, Indonesia. Heliyon 2023; 9:e13382. [PMID: 36744069 PMCID: PMC9886429 DOI: 10.1016/j.heliyon.2023.e13382] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 01/20/2023] [Accepted: 01/29/2023] [Indexed: 02/01/2023] Open
Abstract
Introduction This study aimed to perform mutation and phylogenetic analyses of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Delta variants and analyze the characteristic signs and symptoms of patients infected with SARS-CoV-2 Delta variant originated from Makassar during the Delta outbreak.Methods: We collected samples from patients who were infected with coronavirus disease 2019 (COVID-19) between June and October 2021. We selected the Quantitative Reverse Transcription-Polymerase Chain Reaction (PCR)-positive samples with a cycle threshold value of <30 for whole genome sequencing. Total viral ribonucleic acid (RNA) was isolated from 34 PCR-positive nasopharyngeal swab samples, and whole genome sequencing was performed using the Oxford Nanopore GridlON sequencer. Phylogenetic and maximum clade credibility analyses were performed using the Bayesian Markov chain Monte Carlo method. Results It was found that 33 patients were infected with the SARS-CoV-2 Delta variant in this cohort study, among whom 63.6% (21) patients were female. According to the clinical data, 24 (72.7%), 7 (21.2%), and 2 (6.1%) patients had mild, moderate, and severe COVID-19 infections. Phylogenetic analysis based on the spike and RNA-dependent RNA polymerase (RdRp) genes showed that the collected samples were clustered in the main lineage of B.1.617.2 (Delta variant). The Delta variants had a high frequency of distinct mutations in the spike protein region, including T19R (94.12%), L452R (88.23%), T478K (91.17%), D614G (97%), P681R (97%), and D950 N (97%). Other unique mutations found in a smaller frequency in our samples were present in the N-terminal domain, including A27T (2.94%) and A222V (14.70%), and in the receptor-binding domain, including Q414K (5.88%), G446V (2.94%), and T470 N (2.94%). Conclusion This study revealed the unique mutations in the S protein region of Delta variants. T19R, L452R, T478K/T478R, D614G, P681R, and D950 N were the most common substitutions in Makassar's Delta variant.
Collapse
|
126
|
Shiraz R, Tripathi S. Enhanced recombination among Omicron subvariants of SARS-CoV-2 contributes to viral immune escape. J Med Virol 2023; 95:e28519. [PMID: 36691935 DOI: 10.1002/jmv.28519] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/14/2022] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Abstract
Genetic recombination is an important driver of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evolution, which requires the coinfection of a single host cell with different SARS-CoV-2 strains. To understand the emergence and prevalence of recombinant SARS-CoV-2 lineages through time and space, we analyzed SARS-CoV-2 genome sequences collected from November 2019 to July 2022. We observed an extraordinary increase in the emergence of SARS-CoV-2 recombinant lineages during the Omicron wave, particularly in Northern America and Europe. This phenomenon was independent of the sequencing frequency or genetic diversity of circulating SARS-CoV-2 strains. The recombination breakpoints were more prevalent in the 3'-untranslated region of the viral genome. Importantly, we noted the enrichment of certain amino acids in the Spike protein of recombinant lineages, which have been reported to confer immune escape from neutralizing antibodies and increase angiotensin-converting enzyme 2 receptor binding in some cases. We also observed I42V amino acid change genetically fixated in the NSP14 of the Omicron lineage, which needs further characterization for its potential role in enhanced recombination. Overall, we report the important and timely observation of accelerated recombination in the currently circulating SARS-CoV-2 Omicron variants and explore their potential contribution to viral fitness, particularly immune escape.
Collapse
Affiliation(s)
- Rishad Shiraz
- Microbiology and Cell Biology Department, Indian Institute of Science, Bengaluru, India.,Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
| | - Shashank Tripathi
- Microbiology and Cell Biology Department, Indian Institute of Science, Bengaluru, India.,Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
127
|
Mei R, Heng X, Liu X, Chen G. Glycopolymers for Antibacterial and Antiviral Applications. Molecules 2023; 28:molecules28030985. [PMID: 36770653 PMCID: PMC9919862 DOI: 10.3390/molecules28030985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 01/21/2023] Open
Abstract
Diseases induced by bacterial and viral infections are common occurrences in our daily life, and the main prevention and treatment strategies are vaccination and taking antibacterial/antiviral drugs. However, vaccines can only be used for specific viral infections, and the abuse of antibacterial/antiviral drugs will create multi-drug-resistant bacteria and viruses. Therefore, it is necessary to develop more targeted prevention and treatment methods against bacteria and viruses. Proteins on the surface of bacteria and viruses can specifically bind to sugar, so glycopolymers can be used as potential antibacterial and antiviral drugs. In this review, the research of glycopolymers for bacterial/viral detection/inhibition and antibacterial/antiviral applications in recent years are summarized.
Collapse
Affiliation(s)
- Ruoyao Mei
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| | - Xingyu Heng
- Key Laboratory of Polymeric Material Design and Synthesis for Biomedical Function, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren−Ai Road, Suzhou 215123, China
| | - Xiaoli Liu
- Key Laboratory of Polymeric Material Design and Synthesis for Biomedical Function, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren−Ai Road, Suzhou 215123, China
- Correspondence: (X.L.); (G.C.)
| | - Gaojian Chen
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
- Key Laboratory of Polymeric Material Design and Synthesis for Biomedical Function, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren−Ai Road, Suzhou 215123, China
- Correspondence: (X.L.); (G.C.)
| |
Collapse
|
128
|
Park JH, Lee Y, Choi M, Park E. The Role of Some Vitamins in Respiratory-related Viral Infections: A Narrative Review. Clin Nutr Res 2023; 12:77-89. [PMID: 36793782 PMCID: PMC9900078 DOI: 10.7762/cnr.2023.12.1.77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
This study aimed to find out the effect of vitamins on respiratory-related viral infections, including coronavirus disease 2019 (COVID-19), through the literature reviews. From January 2000 to June 2021, the studies (cohort studies, cross-sectional studies, case-control studies, randomized control trials) related to vitamins (vitamin A, D, E, C, B6, folate, and B12) and COVID-19/severe acute respiratory syndrome/Middle East respiratory syndrome/cold/influenza were selected from the PubMed, Embase, and Cochrane libraries and analyzed. The relationship between vitamins and virus-related respiratory diseases was identified. Through the review, 39 studies were selected on vitamin D, one study on vitamin E, 11 studies on vitamin C, and 3 studies on folate. Regarding COVID-19, 18 studies on vitamin D, 4 studies on vitamin C, and 2 studies on folate showed significant effects of the intake of these nutrients in preventing COVID-19. Regarding colds and influenza, 3 studies on vitamin D, 1 study on vitamin E, 3 studies on vitamin C, and 1 study on folate demonstrated that the intake of these nutrients significantly prevents these diseases. Therefore, this review suggested the intake of vitamins D, E, C, and folate is important for preventing respiratory diseases related to viruses, such as COVID-19, colds, and influenza. The relationship between these nutrients and virus-related respiratory diseases should be continuously monitored in the future.
Collapse
Affiliation(s)
- Jae-Hee Park
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Korea
| | - Yunjung Lee
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Korea
| | - Mijoo Choi
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Korea
| | - Eunju Park
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Korea
| |
Collapse
|
129
|
Naidu SAG, Mustafa G, Clemens RA, Naidu AS. Plant-Derived Natural Non-Nucleoside Analog Inhibitors (NNAIs) against RNA-Dependent RNA Polymerase Complex (nsp7/nsp8/nsp12) of SARS-CoV-2. J Diet Suppl 2023; 20:254-283. [PMID: 34850656 DOI: 10.1080/19390211.2021.2006387] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The emergence of fast-spreading SARS-CoV-2 mutants has sparked a new phase of COVID-19 pandemic. There is a dire necessity for antivirals targeting highly conserved genomic domains on SARS-CoV-2 that are less prone to mutation. The nsp12, also known as the RNA-dependent RNA-polymerase (RdRp), the core component of 'SARS-CoV-2 replication-transcription complex', is a potential well-conserved druggable antiviral target. Several FDA-approved RdRp 'nucleotide analog inhibitors (NAIs)' such as remdesivir, have been repurposed to treat COVID-19 infections. The NAIs target RdRp protein translation and competitively block the nucleotide insertion into the RNA chain, resulting in the inhibition of viral replication. However, the replication proofreading function of nsp14-ExoN could provide resistance to SARS-CoV-2 against many NAIs. Conversely, the 'non-nucleoside analog inhibitors (NNAIs)' bind to allosteric sites on viral polymerase surface, change the redox state; thereby, exert antiviral activity by altering interactions between the enzyme substrate and active core catalytic site of the RdRp. NNAIs neither require metabolic activation (unlike NAIs) nor compete with intracellular pool of nucleotide triphosphates (NTPs) for anti-RdRp activity. The NNAIs from phytonutrient origin are potential antiviral candidates compared to their synthetic counterparts. Several in-silico studies reported the antiviral spectrum of natural phytonutrient-NNAIs such as Suramin, Silibinin (flavonolignan), Theaflavin (tea polyphenol), Baicalein (5,6,7-trihydroxyflavone), Corilagin (gallotannin), Hesperidin (citrus bioflavonoid), Lycorine (pyrrolidine alkaloid), with superior redox characteristics (free binding energy, hydrogen-bonds, etc.) than antiviral drugs (i.e. remdesivir, favipiravir). These phytonutrient-NNAIs also exert anti-inflammatory, antioxidant, immunomodulatory and cardioprotective functions, with multifunctional therapeutic benefits in the clinical management of COVID-19.
Collapse
Affiliation(s)
| | - Ghulam Mustafa
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | - Roger A Clemens
- Department of International Regulatory Science, University of Southern California School of Pharmacy, Los Angeles, CA, USA
| | | |
Collapse
|
130
|
Hussen BM, Sabir DK, Karim Y, Karim KK, Hidayat HJ. RETRACTED ARTICLE: Genome sequence analysis of SARS-COV-2 isolated from a COVID-19 patient in Erbil, Iraq. APPLIED NANOSCIENCE 2023; 13:3147. [PMID: 35155057 PMCID: PMC8818371 DOI: 10.1007/s13204-021-02300-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/27/2021] [Indexed: 01/07/2023]
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Iraq
| | - Dana Khdr Sabir
- Department of Medical Laboratory Sciences, University Charmo, Kurdistan Region, Iraq
| | - Yasin Karim
- Medical Research Center, Hawler Medical University, Kurdistan Region, Iraq
| | | | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University, Kurdistan Region, Iraq
| |
Collapse
|
131
|
Pseudotyped Viruses for Marburgvirus and Ebolavirus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:105-132. [PMID: 36920694 DOI: 10.1007/978-981-99-0113-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Marburg virus (MARV) and Ebola virus (EBOV) of the Filoviridae family are the most lethal viruses in terms of mortality rate. However, the development of antiviral treatment is hampered by the requirement for biosafety level-4 (BSL-4) containment. The establishment of BSL-2 pseudotyped viruses can provide important tools for the study of filoviruses. This chapter summarizes general information on the filoviruses and then focuses on the construction of replication-deficient pseudotyped MARV and EBOV (e.g., lentivirus system and vesicular stomatitis virus system). It also details the potential applications of the pseudotyped viruses, including neutralization antibody detection, the study of infection mechanisms, the evaluation of antibody-dependent enhancement, virus entry inhibitor screening, and glycoprotein mutation analysis.
Collapse
|
132
|
Kulabhusan PK, Pishva P, Çapkın E, Tambe P, Yüce M. Aptamer-based Emerging Tools for Viral Biomarker Detection: A Focus on SARS-CoV-2. Curr Med Chem 2023; 30:910-934. [PMID: 35156569 DOI: 10.2174/1568009622666220214101059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/11/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022]
Abstract
Viral infections can cause fatal illnesses to humans as well as animals. Early detection of viruses is therefore crucial to provide effective treatment to patients. Recently, the Covid-19 pandemic has undoubtedly given an alarming call to develop rapid and sensitive detection platforms. The viral diagnostic tools need to be fast, affordable, and easy to operate with high sensitivity and specificity equivalent or superior to the currently used diagnostic methods. The present detection methods include direct detection of viral antigens or measuring the response of antibodies to viral infections. However, the sensitivity and quantification of the virus are still a significant challenge. Detection tools employing synthetic binding molecules like aptamers may provide several advantages over the conventional methods that use antibodies in the assay format. Aptamers are highly stable and tailorable molecules and are therefore ideal for detection and chemical sensing applications. This review article discusses various advances made in aptamer-based viral detection platforms, including electrochemical, optical, and colorimetric methods to detect viruses, specifically SARS-Cov-2. Considering the several advantages, aptamers could be game-changing in designing high-throughput biosensors for viruses and other biomedical applications in the future.
Collapse
Affiliation(s)
- Prabir Kumar Kulabhusan
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK
| | - Parsa Pishva
- Sabanci University, Faculty of Engineering and Natural Sciences, Istanbul, 34956, Turkey
| | - Eda Çapkın
- Sabanci University, Faculty of Engineering and Natural Sciences, Istanbul, 34956, Turkey
| | - Prajakta Tambe
- Wellcome-- Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Meral Yüce
- Sabanci University, SUNUM Nanotechnology Research, and Application Centre, Istanbul, 34956, Turkey
| |
Collapse
|
133
|
Gao J, Zhang Z, Xue L, Li Y, Cheng T, Meng L, Li Y, Cai W, Hong X, Zhang J, Wang J, Chen M, Ye Q, Ding Y, Wu Q. GII.17[P17] and GII.8[P8] noroviruses showed different RdRp activities associated with their epidemic characteristics. J Med Virol 2023; 95:e28216. [PMID: 36254681 DOI: 10.1002/jmv.28216] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/23/2022] [Accepted: 10/07/2022] [Indexed: 01/11/2023]
Abstract
Norovirus is the primary foodborne pathogenic agent causing viral acute gastroenteritis. It possesses broad genetic diversity and the prevalence of different genotypes varies substantially. However, the differences in RNA-dependent RNA polymerase (RdRp) activity among different genotypes of noroviruses remain unclear. In this study, the molecular mechanism of RdRp activity difference between the epidemic strain GII.17[P17] and the non-epidemic strain GII.8[P8] was characterized. By evaluating the evolutionary history of RdRp sequences with Markov Chain Monte Carlo method, the evolution rate of GII.17[P17] variants was higher than that of GII.8[P8] variants (1.22 × 10-3 nucleotide substitutions/site/year to 9.31 × 10-4 nucleotide substitutions/site/year, respectively). The enzyme catalytic reaction demonstrated that the Vmax value of GII.17[P17] RdRp was 2.5 times than that of GII.8[P8] RdRp. And the Km of GII.17[P17] and GII.8[P8] RdRp were 0.01 and 0.15 mmol/L, respectively. Then, GII.8[P8] RdRp fragment mutants (A-F) were designed, among which GII.8[P8]-A/B containing the conserved motif G/F were found to have significant effects on improving RdRp activity. The Km values of GII.8[P8]-A/B reached 0.07 and 0.06 mmol/L, respectively. And their Vmax values were 1.34 times than that of GII.8[P8] RdRp. In summary, our results suggested that RdRp activities were correlated with their epidemic characteristics. These findings will ultimately provide a better understanding in replication mechanism of noroviruses and development of antiviral drugs.
Collapse
Affiliation(s)
- Junshan Gao
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Zilei Zhang
- Inspection and Quarantine Technology Communication Department, Shanghai Customs College, Shanghai, China
| | - Liang Xue
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Tong Cheng
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Luobing Meng
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Yijing Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Weicheng Cai
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Xiaojing Hong
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Moutong Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Qinghua Ye
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Yu Ding
- Department of Food Science & Technology, Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| |
Collapse
|
134
|
Yadav P, Devasurmutt Y, Tatu U. Phylogenomic and Structural Analysis of the Monkeypox Virus Shows Evolution towards Increased Stability. Viruses 2022; 15:127. [PMID: 36680170 PMCID: PMC9864997 DOI: 10.3390/v15010127] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023] Open
Abstract
Monkeypox is an infectious zoonotic disease caused by an Orthopoxvirus and results in symptoms similar to smallpox. In a recent outbreak, monkeypox virus (MPXV) cases have been reported globally since May 2022, and the numbers are increasing. Monkeypox was first diagnosed in humans in the Democratic Republic of Congo and has now spread to throughout Europe, the USA, and Africa. In this study, we analyzed the whole genome sequences of MPXV sequences from recent outbreaks in various countries and performed phylogenomic analysis. Our analysis of the available human MPXV strains showed the highest mutations per sample in 2022 with the average number of mutations per sample being the highest in South America and the European continents in 2022. We analyzed specific mutations in 11 Indian MPXV strains occurring in the variable end regions of the MPXV genome, where the mutation number was as high as 10 mutations per gene. Among these, envelope glycoproteins, the B2R protein, the Ankyrin repeat protein, DNA polymerase, and the INF alpha receptor-like secreted glycoprotein were seen to have a relatively high number of mutations. We discussed the stabilizing effects of the mutations in some of the highly mutating proteins. Our results showed that the proteins involved in binding to the host receptors were mutating at a faster rate, which empowered the virus for active selection towards increased disease transmissibility and severity.
Collapse
Affiliation(s)
| | | | - Utpal Tatu
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
135
|
Pundir H, Joshi T, Pant M, Bhat S, Pandey J, Chandra S, Tamta S. Identification of SARS-CoV-2 RNA dependent RNA polymerase inhibitors using pharmacophore modelling, molecular docking and molecular dynamics simulation approaches. J Biomol Struct Dyn 2022; 40:13366-13377. [PMID: 34637693 DOI: 10.1080/07391102.2021.1987329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The RNA-dependent RNA polymerase (RdRp) is one of the crucial enzymes in severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) catalysing the replication of RNA, therefore acts as a potential target for antiviral drug design. The fixation of a ligand in the active site of RdRp may alter the SARS-CoV-2 life cycle. Present work aimed at identifying novel inhibitors of the SARS-CoV-2 RdRp enzyme by performing pharmacophore-based virtual screening, molecular docking and molecular dynamics simulation (MDS). Initially, the pharmacophore model of SARS-CoV-2 RdRp was constructed and the resulting model was used to screen compounds from ChEMBL, ZINC and PubChem databases. During the investigation, 180 compounds were screened using the above model and subjected to molecular docking with RdRp. Two compounds viz. ChEMBL1276156 and PubChem135548348 showed a strong binding affinity with RdRp than its standard inhibitor, Remdesivir. Toxicity prediction of these two compounds reveals their non-toxic nature. These compounds were further subjected to MDS for 100 ns to check their stability after binding with RdRp. The MDS of RdRp-ChEMBL1276156 and RdRp-PubChem135548348 complexes show enhanced stability in comparison to the RdRp-Remdesivir complex. The average interaction energy calculated after 100 ns of MDS was -146.56 and -172.68 KJ mol-1 for RdRp-CHEMBL1276156 complex and RdRp-PubChem135548348 complex, respectively, while -59.90 KJ mol-1 for RdRp-Remdesivir complex. The current investigation reveals that these two compounds are potent inhibitors of SARS-CoV-2 RdRp and they could be tested in the experimental condition to evaluate their efficacy against SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Hemlata Pundir
- Department of Botany, D.S.B Campus, Kumaun University, Nainital, Uttarakhand, India
| | - Tanuja Joshi
- Computational Biology & Biotechnology Laboratory, Department of Botany, Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Manish Pant
- Department of Post-Harvest Process and Food Engineering, Govind Ballabh Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, India
| | - Sunaullah Bhat
- Insect Biosystematics & Insect-Pest Management Laboratory, Department of Zoology, Kumaun University-SSJ Campus, Almora, Uttarakhand, India
| | - Jyoti Pandey
- Computational Biology & Biotechnology Laboratory, Department of Botany, Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Subhash Chandra
- Computational Biology & Biotechnology Laboratory, Department of Botany, Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Sushma Tamta
- Department of Botany, D.S.B Campus, Kumaun University, Nainital, Uttarakhand, India
| |
Collapse
|
136
|
A Monovalent Mt10-CVB3 Vaccine Prevents CVB4-Accelerated Type 1 Diabetes in NOD Mice. Vaccines (Basel) 2022; 11:vaccines11010076. [PMID: 36679922 PMCID: PMC9864234 DOI: 10.3390/vaccines11010076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 12/30/2022] Open
Abstract
Enteroviruses, which include Coxsackieviruses, are a common cause of virus infections in humans, and multiple serotypes of the group B Coxsackievirus (CVB) can induce similar diseases. No vaccines are currently available to prevent CVB infections because developing serotype-specific vaccines is not practical. Thus, developing a vaccine that induces protective immune responses for multiple serotypes is desired. In that direction, we created a live-attenuated CVB3 vaccine virus, designated mutant (Mt)10, that offers protection against myocarditis and pancreatitis induced by CVB3 and CVB4 in disease-susceptible A/J mice. Here, we report that the Mt10 vaccine protected against CVB4-triggered type 1 diabetes (T1D) in non-obese diabetic (NOD) mice but the expected subsequent development of spontaneous T1D in these genetically predisposed NOD mice was not altered. We noted that Mt10 vaccine induced significant amounts of neutralizing antibodies, predominantly of the IgG2c isotype, and the virus was not detected in vaccine-challenged animals. Furthermore, monitoring blood glucose levels-and to a lesser extent, insulin antibodies-was found to be helpful in predicting vaccine responses. Taken together, our data suggest that the monovalent Mt10 vaccine has the potential to prevent infections caused by multiple CVB serotypes, as we have demonstrated in various pre-clinical models.
Collapse
|
137
|
D'Aoust PM, Tian X, Towhid ST, Xiao A, Mercier E, Hegazy N, Jia JJ, Wan S, Kabir MP, Fang W, Fuzzen M, Hasing M, Yang MI, Sun J, Plaza-Diaz J, Zhang Z, Cowan A, Eid W, Stephenson S, Servos MR, Wade MJ, MacKenzie AE, Peng H, Edwards EA, Pang XL, Alm EJ, Graber TE, Delatolla R. Wastewater to clinical case (WC) ratio of COVID-19 identifies insufficient clinical testing, onset of new variants of concern and population immunity in urban communities. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 853:158547. [PMID: 36067855 PMCID: PMC9444156 DOI: 10.1016/j.scitotenv.2022.158547] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/10/2022] [Accepted: 09/01/2022] [Indexed: 05/14/2023]
Abstract
Clinical testing has been the cornerstone of public health monitoring and infection control efforts in communities throughout the COVID-19 pandemic. With the anticipated reduction of clinical testing as the disease moves into an endemic state, SARS-CoV-2 wastewater surveillance (WWS) will have greater value as an important diagnostic tool. An in-depth analysis and understanding of the metrics derived from WWS is required to interpret and utilize WWS-acquired data effectively (McClary-Gutierrez et al., 2021; O'Keeffe, 2021). In this study, the SARS-CoV-2 wastewater signal to clinical cases (WC) ratio was investigated across seven cities in Canada over periods ranging from 8 to 21 months. This work demonstrates that significant increases in the WC ratio occurred when clinical testing eligibility was modified to appointment-only testing, identifying a period of insufficient clinical testing (resulting in a reduction to testing access and a reduction in the number of daily tests) in these communities, despite increases in the wastewater signal. Furthermore, the WC ratio decreased significantly in 6 of the 7 studied locations, serving as a potential signal of the emergence of the Alpha variant of concern (VOC) in a relatively non-immunized community (40-60 % allelic proportion), while a more muted decrease in the WC ratio signaled the emergence of the Delta VOC in a relatively well-immunized community (40-60 % allelic proportion). Finally, a significant decrease in the WC ratio signaled the emergence of the Omicron VOC, likely because of the variant's greater effectiveness at evading immunity, leading to a significant number of new reported clinical cases, even when community immunity was high. The WC ratio, used as an additional monitoring metric, could complement clinical case counts and wastewater signals as individual metrics in its potential ability to identify important epidemiological occurrences, adding value to WWS as a diagnostic technology during the COVID-19 pandemic and likely for future pandemics.
Collapse
Affiliation(s)
- Patrick M D'Aoust
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada
| | - Xin Tian
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada
| | | | - Amy Xiao
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Elisabeth Mercier
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada
| | - Nada Hegazy
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada
| | - Jian-Jun Jia
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada
| | - Shen Wan
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada
| | - Md Pervez Kabir
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada
| | - Wanting Fang
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada
| | - Meghan Fuzzen
- Department of Biology, University of Waterloo, Waterloo, Canada
| | - Maria Hasing
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Minqing Ivy Yang
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
| | - Jianxian Sun
- Department of Chemistry, University of Toronto, Toronto, Canada
| | - Julio Plaza-Diaz
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Zhihao Zhang
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada
| | - Aaron Cowan
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada
| | - Walaa Eid
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Sean Stephenson
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Mark R Servos
- Department of Biology, University of Waterloo, Waterloo, Canada
| | - Matthew J Wade
- Data, Analytics and Surveillance Group, UK Health Security Agency, London, United Kingdom
| | - Alex E MacKenzie
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Hui Peng
- Department of Chemistry, University of Toronto, Toronto, Canada
| | - Elizabeth A Edwards
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
| | - Xiao-Li Pang
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Eric J Alm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Tyson E Graber
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Robert Delatolla
- Department of Civil Engineering, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
138
|
Costa LC, Atha B, Hu X, Lamour K, Yang Y, O’Connell M, McFarland C, Foster JA, Hurtado-Gonzales OP. High-throughput detection of a large set of viruses and viroids of pome and stone fruit trees by multiplex PCR-based amplicon sequencing. FRONTIERS IN PLANT SCIENCE 2022; 13:1072768. [PMID: 36578329 PMCID: PMC9791224 DOI: 10.3389/fpls.2022.1072768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/21/2022] [Indexed: 06/17/2023]
Abstract
A comprehensive diagnostic method of known plant viruses and viroids is necessary to provide an accurate phytosanitary status of fruit trees. However, most widely used detection methods have a small limit on either the number of targeted viruses/viroids or the number of samples to be evaluated at a time, hampering the ability to rapidly scale up the test capacity. Here we report that by combining the power of high multiplexing PCR (499 primer pairs) of small amplicons (120-135bp), targeting 27 viruses and 7 viroids of fruit trees, followed by a single high-throughput sequencing (HTS) run, we accurately diagnosed the viruses and viroids on as many as 123 pome and stone fruit tree samples. We compared the accuracy, sensitivity, and reproducibility of this approach and contrast it with other detection methods including HTS of total RNA (RNA-Seq) and individual RT-qPCR for every fruit tree virus or viroid under the study. We argue that this robust and high-throughput cost-effective diagnostic tool will enhance the viral/viroid knowledge of fruit trees while increasing the capacity for large scale diagnostics. This approach can also be adopted for the detection of multiple viruses and viroids in other crops.
Collapse
Affiliation(s)
- Larissa Carvalho Costa
- Plant Germplasm Quarantine Program, Animal and Plant Health Inspection Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Benjamin Atha
- Plant Germplasm Quarantine Program, Animal and Plant Health Inspection Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Xiaojun Hu
- Plant Germplasm Quarantine Program, Animal and Plant Health Inspection Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Kurt Lamour
- Department of Entomology and Plant Pathology, University of Tennessee, Knoxville, TN, United States
| | - Yu Yang
- Plant Germplasm Quarantine Program, Animal and Plant Health Inspection Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Mary O’Connell
- Plant Germplasm Quarantine Program, Animal and Plant Health Inspection Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Clint McFarland
- Plant Protection and Quarantine - Field Operations, Animal and Plant Health Inspection Service, United States Department of Agriculture, Raleigh, NC, United States
| | - Joseph A. Foster
- Plant Germplasm Quarantine Program, Animal and Plant Health Inspection Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Oscar P. Hurtado-Gonzales
- Plant Germplasm Quarantine Program, Animal and Plant Health Inspection Service, United States Department of Agriculture, Beltsville, MD, United States
| |
Collapse
|
139
|
Biothermodynamics of Viruses from Absolute Zero (1950) to Virothermodynamics (2022). Vaccines (Basel) 2022; 10:vaccines10122112. [PMID: 36560522 PMCID: PMC9784531 DOI: 10.3390/vaccines10122112] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Biothermodynamics of viruses is among the youngest but most rapidly developing scientific disciplines. During the COVID-19 pandemic, it closely followed the results published by molecular biologists. Empirical formulas were published for 50 viruses and thermodynamic properties for multiple viruses and virus variants, including all variants of concern of SARS-CoV-2, SARS-CoV, MERS-CoV, Ebola virus, Vaccinia and Monkeypox virus. A review of the development of biothermodynamics of viruses during the last several decades and intense development during the last 3 years is described in this paper.
Collapse
|
140
|
Sun Y, Wang M, Lin W, Dong W, Xu J. "Mutation blacklist" and "mutation whitelist" of SARS-CoV-2. JOURNAL OF BIOSAFETY AND BIOSECURITY 2022; 4:114-120. [PMID: 35845149 PMCID: PMC9273572 DOI: 10.1016/j.jobb.2022.06.006] [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: 05/31/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 01/26/2023] Open
Abstract
Over the past two years, scientists throughout the world have completed more than 6 million SARS-CoV-2 genome sequences. Today, the number of SARS-CoV-2 genomes exceeds the total number of all other viral genomes. These genomes are a record of the evolution of SARS-CoV-2 in the human host, and provide information on the emergence of mutations. In this study, analysis of these sequenced genomes identified 296,728 de novo mutations (DNMs), and found that six types of base substitutions reached saturation in the sequenced genome population. Based on this analysis, a "mutation blacklist" of SARS-CoV-2 was compiled. The loci on the "mutation blacklist" are highly conserved, and these mutations likely have detrimental effects on virus survival, replication, and transmission. This information is valuable for SARS-CoV-2 research on gene function, vaccine design, and drug development. Through association analysis of DNMs and viral transmission rates, we identified 185 DNMs that positively correlated with the SARS-CoV-2 transmission rate, and these DNMs where classified as the "mutation whitelist" of SARS-CoV-2. The mutations on the "mutation whitelist" are beneficial for SARS-CoV-2 transmission and could therefore be used to evaluate the transmissibility of new variants. The occurrence of mutations and the evolution of viruses are dynamic processes. To more effectively monitor the mutations and variants of SARS-CoV-2, we built a SARS-CoV-2 mutation and variant monitoring and pre-warning system (MVMPS), which can monitor the occurrence and development of mutations and variants of SARS-CoV-2, as well as provide pre-warning for the prevention and control of SARS-CoV-2 (https://www.omicx.cn/). Additionally, this system could be used in real-time to update the "mutation whitelist" and "mutation blacklist" of SARS-CoV-2.
Collapse
Affiliation(s)
- Yamin Sun
- Research Institute of Public Health, Nankai University, Tianjin, PR China
- Research Center for Functional Genomics and Biochip, Tianjin, PR China
| | - Min Wang
- Research Center for Functional Genomics and Biochip, Tianjin, PR China
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, PR China
| | - Wenchao Lin
- Research Center for Functional Genomics and Biochip, Tianjin, PR China
| | - Wei Dong
- Research Center for Functional Genomics and Biochip, Tianjin, PR China
| | - Jianguo Xu
- Research Institute of Public Health, Nankai University, Tianjin, PR China
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 202206, PR China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| |
Collapse
|
141
|
Thakor JC, Dinesh M, Manikandan R, Bindu S, Sahoo M, Sahoo D, Dhawan M, Pandey MK, Tiwari R, Emran TB, Dhama K, Chaicumpa W. Swine coronaviruses (SCoVs) and their emerging threats to swine population, inter-species transmission, exploring the susceptibility of pigs for SARS-CoV-2 and zoonotic concerns. Vet Q 2022; 42:125-147. [PMID: 35584308 PMCID: PMC9225692 DOI: 10.1080/01652176.2022.2079756] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 03/23/2022] [Accepted: 05/16/2022] [Indexed: 12/23/2022] Open
Abstract
Swine coronaviruses (SCoVs) are one of the most devastating pathogens affecting the livelihoods of farmers and swine industry across the world. These include transmissible gastroenteritis virus (TGEV), porcine epidemic diarrhea virus (PEDV), porcine respiratory coronavirus (PRCV), porcine hemagglutinating encephalomyelitis virus (PHEV), swine acute diarrhea syndrome coronavirus (SADS-CoV), and porcine delta coronavirus (PDCoV). Coronaviruses infect a wide variety of animal species and humans because these are having single stranded-RNA that accounts for high mutation rates and thus could break the species barrier. The gastrointestinal, cardiovascular, and nervous systems are the primary organ systems affected by SCoVs. Infection is very common in piglets compared to adult swine causing high mortality in the former. Bat is implicated to be the origin of all CoVs affecting animals and humans. Since pig is the only domestic animal in which CoVs cause a wide range of diseases; new coronaviruses with high zoonotic potential could likely emerge in the future as observed in the past. The recently emerged severe acute respiratory syndrome coronavirus virus-2 (SARS-CoV-2), causing COVID-19 pandemic in humans, has been implicated to have animal origin, also reported from few animal species, though its zoonotic concerns are still under investigation. This review discusses SCoVs and their epidemiology, virology, evolution, pathology, wildlife reservoirs, interspecies transmission, spill-over events and highlighting their emerging threats to swine population. The role of pigs amid ongoing SARS-CoV-2 pandemic will also be discussed. A thorough investigation should be conducted to rule out zoonotic potential of SCoVs and to design appropriate strategies for their prevention and control.
Collapse
Affiliation(s)
- Jigarji C. Thakor
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Murali Dinesh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Rajendran Manikandan
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Suresh Bindu
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Monalisa Sahoo
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Diptimayee Sahoo
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, India
- The Trafford Group of Colleges, Manchester, United Kingdom
| | - Megha Katare Pandey
- Department of Translational Medicine Center, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
142
|
Oronsky B, Larson C, Caroen S, Reid TR. Could nucleocapsid be a next-generation COVID-19 vaccine candidate - author's reply. Int J Infect Dis 2022; 125:227. [PMID: 36347457 PMCID: PMC9636028 DOI: 10.1016/j.ijid.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/08/2022] Open
|
143
|
Scott AD, King DM, Ordway SW, Bahar S. Phase transitions in evolutionary dynamics. CHAOS (WOODBURY, N.Y.) 2022; 32:122101. [PMID: 36587338 DOI: 10.1063/5.0124274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Sharp changes in state, such as transitions from survival to extinction, are hallmarks of evolutionary dynamics in biological systems. These transitions can be explored using the techniques of statistical physics and the physics of nonlinear and complex systems. For example, a survival-to-extinction transition can be characterized as a non-equilibrium phase transition to an absorbing state. Here, we review the literature on phase transitions in evolutionary dynamics. We discuss directed percolation transitions in cellular automata and evolutionary models, and models that diverge from the directed percolation universality class. We explore in detail an example of an absorbing phase transition in an agent-based model of evolutionary dynamics, including previously unpublished data demonstrating similarity to, but also divergence from, directed percolation, as well as evidence for phase transition behavior at multiple levels of the model system's evolutionary structure. We discuss phase transition models of the error catastrophe in RNA virus dynamics and phase transition models for transition from chemistry to biochemistry, i.e., the origin of life. We conclude with a review of phase transition dynamics in models of natural selection, discuss the possible role of phase transitions in unraveling fundamental unresolved questions regarding multilevel selection and the major evolutionary transitions, and assess the future outlook for phase transitions in the investigation of evolutionary dynamics.
Collapse
Affiliation(s)
- Adam D Scott
- Department of Physics and Astronomy and Center for Neurodynamics, University of Missouri at St. Louis, One University Blvd., St. Louis, Missouri 63121, USA
| | - Dawn M King
- Department of Physics and Astronomy and Center for Neurodynamics, University of Missouri at St. Louis, One University Blvd., St. Louis, Missouri 63121, USA
| | - Stephen W Ordway
- Department of Physics and Astronomy and Center for Neurodynamics, University of Missouri at St. Louis, One University Blvd., St. Louis, Missouri 63121, USA
| | - Sonya Bahar
- Department of Physics and Astronomy and Center for Neurodynamics, University of Missouri at St. Louis, One University Blvd., St. Louis, Missouri 63121, USA
| |
Collapse
|
144
|
Khan MZI, Nazli A, Al-furas H, Asad MI, Ajmal I, Khan D, Shah J, Farooq MA, Jiang W. An overview of viral mutagenesis and the impact on pathogenesis of SARS-CoV-2 variants. Front Immunol 2022; 13:1034444. [PMID: 36518757 PMCID: PMC9742215 DOI: 10.3389/fimmu.2022.1034444] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/04/2022] [Indexed: 11/29/2022] Open
Abstract
Viruses are submicroscopic, obligate intracellular parasites that carry either DNA or RNA as their genome, protected by a capsid. Viruses are genetic entities that propagate by using the metabolic and biosynthetic machinery of their hosts and many of them cause sickness in the host. The ability of viruses to adapt to different hosts and settings mainly relies on their ability to create de novo variety in a short interval of time. The size and chemical composition of the viral genome have been recognized as important factors affecting the rate of mutations. Coronavirus disease 2019 (Covid-19) is a novel viral disease that has quickly become one of the world's leading causes of mortality, making it one of the most serious public health problems in recent decades. The discovery of new medications to cope with Covid-19 is a difficult and time-consuming procedure, as new mutations represent a serious threat to the efficacy of recently developed vaccines. The current article discusses viral mutations and their impact on the pathogenicity of newly developed variants with a special emphasis on Covid-19. The biology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), its mutations, pathogenesis, and treatment strategies are discussed in detail along with the statistical data.
Collapse
Affiliation(s)
| | - Adila Nazli
- Faculty of Biological Sciences, Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hawaa Al-furas
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MOE) of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou, China
| | - Muhammad Imran Asad
- Faculty of Biological Sciences, Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Iqra Ajmal
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, Shanghai, China
| | - Dildar Khan
- Faculty of Biological Sciences, Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jaffer Shah
- Department of Health, New York, NY, United States,*Correspondence: Jaffer Shah, ; Muhammad Asad Farooq, ; Wenzheng Jiang,
| | - Muhammad Asad Farooq
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, Shanghai, China,*Correspondence: Jaffer Shah, ; Muhammad Asad Farooq, ; Wenzheng Jiang,
| | - Wenzheng Jiang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, Shanghai, China,*Correspondence: Jaffer Shah, ; Muhammad Asad Farooq, ; Wenzheng Jiang,
| |
Collapse
|
145
|
Bhoi TK, Samal I, Majhi PK, Komal J, Mahanta DK, Pradhan AK, Saini V, Nikhil Raj M, Ahmad MA, Behera PP, Ashwini M. Insight into aphid mediated Potato Virus Y transmission: A molecular to bioinformatics prospective. Front Microbiol 2022; 13:1001454. [PMID: 36504828 PMCID: PMC9729956 DOI: 10.3389/fmicb.2022.1001454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Potato, the world's most popular crop is reported to provide a food source for nearly a billion people. It is prone to a number of biotic stressors that affect yield and quality, out of which Potato Virus Y (PVY) occupies the top position. PVY can be transmitted mechanically and by sap-feeding aphid vectors. The application of insecticide causes an increase in the resistant vector population along with detrimental effects on the environment; genetic resistance and vector-virus control are the two core components for controlling the deadly PVY. Using transcriptomic tools together with differential gene expression and gene discovery, several loci and genes associated with PVY resistance have been widely identified. To combat this virus we must increase our understanding on the molecular response of the PVY-potato plant-aphid interaction and knowledge of genome organization, as well as the function of PVY encoded proteins, genetic diversity, the molecular aspects of PVY transmission by aphids, and transcriptome profiling of PVY infected potato cultivars. Techniques such as molecular and bioinformatics tools can identify and monitor virus transmission. Several studies have been conducted to understand the molecular basis of PVY resistance/susceptibility interactions and their impact on PVY epidemiology by studying the interrelationship between the virus, its vector, and the host plant. This review presents current knowledge of PVY transmission, epidemiology, genome organization, molecular to bioinformatics responses, and its effective management.
Collapse
Affiliation(s)
- Tanmaya Kumar Bhoi
- Forest Protection Division, ICFRE-Arid Forest Research Institute (AFRI), Jodhpur, Rajasthan, India
| | - Ipsita Samal
- Department of Entomology, Sri Sri University, Cuttack, Odisha, India
| | - Prasanta Kumar Majhi
- Department of Plant Breeding and Genetics, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - J. Komal
- Department of Entomology, Navsari Agricultural University, Navsari, Gujarat, India,J. Komal
| | - Deepak Kumar Mahanta
- Department of Entomology, Dr. Rajendra Prasad Central Agricultural University, Samastipur, India,*Correspondence: Deepak Kumar Mahanta
| | - Asit Kumar Pradhan
- Social Science Division, ICAR-National Rice Research Institute (NRRI), Cuttack, Odisha, India
| | - Varun Saini
- Division of Entomology, ICAR-Indian Agricultural Research Institute (IARI), New Delhi, India
| | - M. Nikhil Raj
- Division of Entomology, ICAR-Indian Agricultural Research Institute (IARI), New Delhi, India
| | - Mohammad Abbas Ahmad
- Department of Entomology, Dr. Rajendra Prasad Central Agricultural University, Samastipur, India
| | | | - Mangali Ashwini
- Department of Entomology, Navsari Agricultural University, Navsari, Gujarat, India
| |
Collapse
|
146
|
Omicron BA.2.75 Sublineage (Centaurus) Follows the Expectations of the Evolution Theory: Less Negative Gibbs Energy of Biosynthesis Indicates Decreased Pathogenicity. MICROBIOLOGY RESEARCH 2022. [DOI: 10.3390/microbiolres13040066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
SARS-CoV-2 belongs to the group of RNA viruses with a pronounced tendency to mutate. Omicron BA.2.75 is a subvariant believed to be able to suppress the currently dominant BA.5 and cause a new winter wave of the COVID-19 pandemic. Omicron BA.2.75 is characterized by a greater infectivity compared to earlier Omicron variants. However, the Gibbs energy of the biosynthesis of virus particles is slightly less negative compared to those of other variants. Thus, the multiplication rate of Omicron BA.2.75 is lower than that of other SARS-CoV-2 variants. This leads to slower accumulation of newly formed virions and less damage to host cells, indicating evolution of SARS-CoV-2 toward decreasing pathogenicity.
Collapse
|
147
|
Perlinska AP, Stasiulewicz A, Nguyen ML, Swiderska K, Zmudzinski M, Maksymiuk AW, Drag M, Sulkowska JI. Amino acid variants of SARS-CoV-2 papain-like protease have impact on drug binding. PLoS Comput Biol 2022; 18:e1010667. [PMID: 36409737 PMCID: PMC9721480 DOI: 10.1371/journal.pcbi.1010667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 12/05/2022] [Accepted: 10/19/2022] [Indexed: 11/22/2022] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused both a health and economic crisis around the world. Its papain-like protease (PLpro) is one of the protein targets utilized in designing new drugs that would aid vaccines in the fight against the virus. Although there are already several potential candidates for a good inhibitor of this protein, the degree of variability of the protein itself is not taken into account. As an RNA virus, SARS-CoV-2 can mutate to a high degree, but PLpro variability has not been studied to date. Based on sequence data available in databases, we analyzed the mutational potential of this protein. We focused on the effect of observed mutations on inhibitors' binding mode and their efficacy as well as protein's activity. Our analysis identifies five mutations that should be monitored and included in the drug design process: P247S, E263D-Y264H and T265A-Y268C.
Collapse
Affiliation(s)
| | - Adam Stasiulewicz
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
- Department of Drug Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Mai Lan Nguyen
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
| | - Karolina Swiderska
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Mikolaj Zmudzinski
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Alicja W. Maksymiuk
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Marcin Drag
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | | |
Collapse
|
148
|
Bhadane R, Salo-Ahen OMH. High-Throughput Molecular Dynamics-Based Alchemical Free Energy Calculations for Predicting the Binding Free Energy Change Associated with the Selected Omicron Mutations in the Spike Receptor-Binding Domain of SARS-CoV-2. Biomedicines 2022; 10:2779. [PMID: 36359299 PMCID: PMC9687918 DOI: 10.3390/biomedicines10112779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 11/10/2023] Open
Abstract
The ongoing pandemic caused by SARS-CoV-2 has gone through various phases. Since the initial outbreak, the virus has mutated several times, with some lineages showing even stronger infectivity and faster spread than the original virus. Among all the variants, omicron is currently classified as a variant of concern (VOC) by the World Health Organization, as the previously circulating variants have been replaced by it. In this work, we have focused on the mutations observed in omicron sub lineages BA.1, BA.2, BA.4 and BA.5, particularly at the receptor-binding domain (RBD) of the spike protein that is responsible for the interactions with the host ACE2 receptor and binding of antibodies. Studying such mutations is particularly important for understanding the viral infectivity, spread of the disease and for tracking the escape routes of this virus from antibodies. Molecular dynamics (MD) based alchemical free energy calculations have been shown to be very accurate in predicting the free energy change, due to a mutation that could have a deleterious or a stabilizing effect on either the protein itself or its binding affinity to another protein. Here, we investigated the significance of five spike RBD mutations on the stability of the spike protein binding to ACE2 by free energy calculations using high throughput MD simulations. For comparison, we also used conventional MD simulations combined with a Molecular Mechanics-Generalized Born Surface Area (MM-GBSA) based approach, and compared our results with the available experimental data. Overall, the alchemical free energy calculations performed far better than the MM-GBSA approach in predicting the individual impact of the mutations. When considering the experimental variation, the alchemical free energy method was able to produce a relatively accurate prediction for N501Y, the mutant that has previously been reported to increase the binding affinity to hACE2. On the other hand, the other individual mutations seem not to have a significant effect on the spike RBD binding affinity towards hACE2.
Collapse
Affiliation(s)
- Rajendra Bhadane
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, FI-20520 Turku, Finland
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Pharmacy, Åbo Akademi University, FI-20520 Turku, Finland
| | - Outi M. H. Salo-Ahen
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, FI-20520 Turku, Finland
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Pharmacy, Åbo Akademi University, FI-20520 Turku, Finland
| |
Collapse
|
149
|
Hardin LT, Xiao N. miRNAs: The Key Regulator of COVID-19 Disease. Int J Cell Biol 2022; 2022:1645366. [PMID: 36345541 PMCID: PMC9637033 DOI: 10.1155/2022/1645366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/30/2022] [Indexed: 01/12/2024] Open
Abstract
As many parts of the world continue to fight the innumerable waves of COVID-19 infection, SARS-CoV-2 continues to sculpt its antigenic determinants to enhance its virulence and evolvability. Several vaccines were developed and used around the world, and oral antiviral medications are being developed against SARS-CoV-2. However, studies showed that the virus is mutating in line with the antibody's neutralization escape; thus, new therapeutic alternatives are solicited. We hereby review the key role that miRNAs can play as epigenetic mediators of the cross-talk between SARS-CoV-2 and the host cells. The limitations resulting from the "virus intelligence" to escape and antagonize the host miRNAs as well as the possible mechanisms that could be used in the viral evasion strategies are discussed. Lastly, we suggest new therapeutic approaches based on viral miRNAs.
Collapse
Affiliation(s)
- Leyla Tahrani Hardin
- Department of Biomedical Sciences at the Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, 94103 CA, USA
| | - Nan Xiao
- Department of Biomedical Sciences at the Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, 94103 CA, USA
| |
Collapse
|
150
|
A New HIV-1 K 28E 32-Reverse Transcriptase Variant Associated with the Rapid Expansion of CRF07_BC among Men Who Have Sex with Men. Microbiol Spectr 2022; 10:e0254522. [PMID: 36214682 PMCID: PMC9604004 DOI: 10.1128/spectrum.02545-22] [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] [Indexed: 01/04/2023] Open
Abstract
HIV-1 CRF07_BC originated among injection drug users (IDUs) in China. After diffusing into men who have sex with men (MSM), CRF07_BC has shown a rapid expansion in this group; however, the mechanism remains unclear. Here, we identified a new K28E32 variant of CRF07_BC that was characterized by five specific mutations (E28K, K32E, E248V, K249Q, and T338S) in reverse transcriptase. This variant was mainly prevalent among MSM, and was overrepresented in transmission clusters, suggesting that it could have driven the rapid expansion of CRF07_BC in MSM, though founder effects cannot be ruled out. It was descended from an evolutionary intermediate accumulating four specific mutations and formed an independent phylogenetic node with an estimated origin time in 2003. The K28E32 variant was demonstrated to have significantly higher in vitro HIV-1 replication ability than the wild type. Mutations E28K and K32E play a critical role in the improvement of in vitro HIV-1 replication ability, reflected by improved reverse transcription activity. The results could allow public health officials to use this marker (especially E28K and K32E mutations in the reverse transcriptase (RT) coding region) to target prevention measures prioritizing MSM population and persons infected with this variant for test and treat initiatives. IMPORTANCE HIV-1 has very high mutation rate that is correlated with the survival and adaption of the virus. The variants with higher transmissibility may be more selective advantage than the strains with higher virulence. Several HIV-1 variants were previously demonstrated to be correlated with higher viral load and lower CD4 T cell count. Here, we first identified a new variant (the K28E32 variant) of HIV-1 CRF07_BC, described its origin and evolutionary dynamics, and demonstrated its higher in vitro HIV-1 replication ability than the wild type. We demonstrated that five RT mutations (especially E28K and K32E) significantly improve in vitro HIV-1 replication ability. The appearance of the new K28E32 variant was associated with the rapidly increasing prevalence of CRF07_BC among MSM.
Collapse
|