Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Frontiers of COVID-19: Scientific and Clinical Aspects of the Novel Coronavirus 2019 ; : 561-574, 2022.
Article in English | Scopus | ID: covidwho-20239082

ABSTRACT

Since the beginning of the COVID-19 pandemic in late 2019, SARS-CoV-2 has started to optimize itself. After crossing the species barrier between bats and humans, it has developed mutations in the viral spike protein, in particular at positions 69/70, 452, 501, 614, and 681, that enhance binding to the ACE-2 receptor and entry into host cells, thereby promoting viral transmissibility and pathogenesis. Mutations at positions 417 and 484 have begun to undermine the effectiveness of convalescent plasma, monoclonal antibodies, and currently available vaccines. The targeted and convergent evolution of SARS-CoV-2, which occurred despite the proofreading activity of the exonuclease, has resulted so far in five variants of concern, which have replaced previous strains. This calls for a worldwide surveillance of viral evolution including animal transmission and the development of vaccines responding to escape variants and inducing mucosal immunity. © The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature Switzerland AG 2022.

2.
Genomics Proteomics Bioinformatics ; 2022 Jun 24.
Article in English | MEDLINE | ID: covidwho-2323660

ABSTRACT

SARS-CoV-2 is a new RNA virus affecting humans and spreads extensively through world populations since its first outbreak in December, 2019. Whether the transmissibility and pathogenicity of SARS-CoV-2 in humans after zoonotic transfer are actively evolving, and driven by adaptation to the new host and environments is still under debate. Understanding the evolutionary mechanism underlying epidemiological and pathological characteristics of COVID-19 is essential for predicting the epidemic trend, and providing guidance for disease control and treatments. Interrogating novel strategies for identifying natural selection using within-species polymorphisms and 3,674,076 SARS-CoV-2 genome sequences of 169 countries as of December 30, 2021, we demonstrate with population genetic evidence that during the course of SARS-CoV-2 pandemic in humans, (i) SARS-CoV-2 genomes are overall conserved under purifying selection, especially for the 14 genes related to viral RNA replication, transcription, and assembly; (ii) Ongoing positive selection is actively driving the evolution of 6 genes (e.g., S, ORF3a, and N) that play critical roles in molecular processes involving pathogen-host interactions, including viral invasion into and egress from host cells, viral inhibition, or evasion of host immune response, possibly leading to high transmissibility and mild symptom in SARS-CoV-2 evolution. According to an established haplotype phylogenetic relationship of 138 viral clusters, a spatial and temporal landscape of 556 critical mutations is constructed based on their divergence among viral haplotype clusters or repeatedly increase in frequency within at least 2 clusters, of which multiple mutations potentially conferring alterations in viral transmissibility, pathogenicity, and virulence of SARS-CoV-2 are highlighted, warranting attentions.

3.
Topics in Antiviral Medicine ; 31(2):117, 2023.
Article in English | EMBASE | ID: covidwho-2314619

ABSTRACT

Background: Individuals living with HIV are at increased risk of morbidity and mortality from COVID-19. Furthermore, SARS-CoV-2 infection in immunocompromised HIV infected individuals poses a risk to prolonged infection and viral shedding and the emergence of new variants of concern (VOCs). Using the SIV macaque model for AIDS, we are investigating the hypothesis that immune dysfunction during HIV infection will prolong SARSCoV- 2 viral infection, promote enhanced COVID-19 disease, and accelerate viral evolution. Here, we report the impact of SIV-CoV-2 co-infection on immune responses and pathogenesis. Method(s): Eight female rhesus macaques (aged 7-15 years, 5.5-9.9kg) were infected with SIVmac251 via low dose intravaginal challenge and then inoculated with 6.5x105 TCID50/mL SARS-CoV-2 (WA-1) at 17-34 weeks post-SIV infection via combined intranasal and intratracheal routes. Blood, bronchoalveolar lavage (BAL), stool, and nasal, oral, and rectal swabs were collected pre-infection through 14 days post-infection (DPI) to measure immune responses and viremia. ELISAs, ELISPOT, qRT-PCR, lung pathology, cytokine multiplex, and virus neutralization assays were performed to measure viral loads, pathogenesis, and immune responses. Result(s): Three days post-SARS-CoV-2 infection, we observed a transient decrease in CD4 counts, but there were no changes in clinical symptoms or plasma SIV viral loads. However, SARS-CoV-2 replication persisted in the upper respiratory tract, but not the lower respiratory tract. In addition, SARS-CoV-2 IgG seroconversion was delayed and antigen-specific T-cell responses were dampened. Notably, viral RNA levels in nasal swabs were significantly higher 7-14 DPI in SIV+ compared to previously published results using the same SARS-CoV-2 challenge virus in SIV- rhesus (PMCID: PMC8462335, PMC8829873). In addition, SIV/CoV-2 co-infected animals exhibited elevated levels of myeloperoxidase (MPO), a marker of neutrophil activation and increased lung inflammation. Conclusion(s): Here we provide evidence for the utility of the rhesus macaque in modeling human HIV-SARS-CoV-2 co-infection. Our results suggest that immunosuppression during SIV infection impairs de novo generation of anti-SARS-CoV-2 immunity, that may contribute to prolonged SARS-CoV-2 viral shedding, increased transmission windows, altered disease pathogenesis, and lower protection against subsequent SARS-CoV-2 exposures. Studies in progress will determine if SARS-CoV-2 viral evolution is accelerated in SIV-infected macaques.

4.
Viruses ; 15(4)2023 03 30.
Article in English | MEDLINE | ID: covidwho-2298421

ABSTRACT

Brazil currently ranks second in absolute deaths by COVID-19, even though most of its population has completed the vaccination protocol. With the introduction of Omicron in late 2021, the number of COVID-19 cases soared once again in the country. We investigated in this work how lineages BA.1 and BA.2 entered and spread in the country by sequencing 2173 new SARS-CoV-2 genomes collected between October 2021 and April 2022 and analyzing them in addition to more than 18,000 publicly available sequences with phylodynamic methods. We registered that Omicron was present in Brazil as early as 16 November 2021 and by January 2022 was already more than 99% of samples. More importantly, we detected that Omicron has been mostly imported through the state of São Paulo, which in turn dispersed the lineages to other states and regions of Brazil. This knowledge can be used to implement more efficient non-pharmaceutical interventions against the introduction of new SARS-CoV variants focused on surveillance of airports and ground transportation.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , Brazil/epidemiology , Transportation , Vaccination
5.
J Proteome Res ; 22(6): 1984-1996, 2023 06 02.
Article in English | MEDLINE | ID: covidwho-2303154

ABSTRACT

SARS-CoV-2 has significantly mutated its genome during the past 3 years, leading to the periodic emergence of several variants. Some of the variants possess enhanced fitness advantage, transmissibility, and pathogenicity and can also reduce vaccine efficacy. Thus, it is important to track the viral evolution to prevent and protect the mankind from SARS-CoV-2 infection. To this end, an interactive web-GUI platform, namely, CoVe-tracker (SARS-CoV-2 evolution tracker), is developed to track its pan proteome evolutionary dynamics (https://project.iith.ac.in/cove-tracker/). CoVe-tracker provides an opportunity for the user to fetch the country-wise and protein-wise amino acid mutations (currently, 44139) of SARS-CoV-2 and their month-wise distribution. It also provides position-wise evolution observed in the SARS-CoV-2 proteome. Importantly, CoVe-tracker provides month- and country-wise distributions of 2065 phylogenetic assignment of named global outbreak (PANGO) lineages and their 177564 variants. It further provides periodic updates on SARS-CoV-2 variant(s) evolution. CoVe-tracker provides the results in a user-friendly interactive fashion by projecting the results onto the world map (for country-wise distribution) and protein 3D structure (for protein-wise mutation). The application of CoVe-tracker in tracking the closest cousin(s) of a variant is demonstrated by considering BA.4 and BA.5 PANGO lineages as test cases. Thus, CoVe-tracker would be useful in the quick surveillance of newly emerging mutations/variants/lineages to facilitate the understanding of viral evolution, transmission, and disease epidemiology.


Subject(s)
COVID-19 , Proteome , Humans , Proteome/genetics , SARS-CoV-2/genetics , COVID-19/epidemiology , Phylogeny , Mutation
6.
Cell ; 185(12): 2086-2102.e22, 2022 06 09.
Article in English | MEDLINE | ID: covidwho-2293192

ABSTRACT

Across biological scales, gene-regulatory networks employ autorepression (negative feedback) to maintain homeostasis and minimize failure from aberrant expression. Here, we present a proof of concept that disrupting transcriptional negative feedback dysregulates viral gene expression to therapeutically inhibit replication and confers a high evolutionary barrier to resistance. We find that nucleic-acid decoys mimicking cis-regulatory sites act as "feedback disruptors," break homeostasis, and increase viral transcription factors to cytotoxic levels (termed "open-loop lethality"). Feedback disruptors against herpesviruses reduced viral replication >2-logs without activating innate immunity, showed sub-nM IC50, synergized with standard-of-care antivirals, and inhibited virus replication in mice. In contrast to approved antivirals where resistance rapidly emerged, no feedback-disruptor escape mutants evolved in long-term cultures. For SARS-CoV-2, disruption of a putative feedback circuit also generated open-loop lethality, reducing viral titers by >1-log. These results demonstrate that generating open-loop lethality, via negative-feedback disruption, may yield a class of antimicrobials with a high genetic barrier to resistance.


Subject(s)
Antiviral Agents , Gene Expression Regulation, Viral/drug effects , Animals , Antiviral Agents/pharmacology , Drug Resistance, Viral , Gene Regulatory Networks/drug effects , Mice , SARS-CoV-2/drug effects , Virus Replication
7.
Uncovering The Science of Covid-19 ; : 14-28, 2022.
Article in English | Scopus | ID: covidwho-2280888

ABSTRACT

On 30 January 2020, the World Health Organization (WHO) characterized the novel severe acute respiratory syndrome Coronavirus 2 (SARSCoV- 2) outbreak as a Public Health Emergency of International Concern. Subsequently, on 11 March 2020, WHO declared the global spread of Coronavirus disease 2019 (COVID-19) as a pandemic triggered by this causative virus. This COVID-19 pandemic has impacted lives and livelihoods worldwide, resulting in unprecedented social disruption and economic losses. In order to design and develop effective diagnostics, vaccines and therapeutic interventions against SARS-CoV-2, it is imperative to understand the molecular and cellular mechanisms underpinning the complex interactions between this virus, its variants, and its infected hosts. This chapter provides an overview on the classification, genomic organization and evolution of SARS-CoV-2 (including the emergence of variants from Alpha to Omicron), and summarizes existing and emerging testing strategies. With unprecedented speed, an array of conventional and new COVID-19 vaccines has been developed, evaluated in clinical trials, and administered to billions worldwide. Current and novel antiviral drugs and immunomodulatory approaches are discussed for the therapeutic and prophylactic management of SARS-CoV-2 infections. Finally, much remains for humanity to discover and learn as the world must continue to adapt and live with endemic COVID-19 and SARSCoV- 2 evolution. © 2023 by World Scientific Publishing Co. Pte. Ltd.

8.
Viruses ; 15(3)2023 03 05.
Article in English | MEDLINE | ID: covidwho-2287153

ABSTRACT

Almost all published rooting and dating studies on SARS-CoV-2 assumed that (1) evolutionary rate does not change over time although different lineages can have different evolutionary rates (uncorrelated relaxed clock), and (2) a zoonotic transmission occurred in Wuhan and the culprit was immediately captured, so that only the SARS-CoV-2 genomes obtained in 2019 and the first few months of 2020 (resulting from the first wave of the global expansion from Wuhan) are sufficient for dating the common ancestor. Empirical data contradict the first assumption. The second assumption is not warranted because mounting evidence suggests the presence of early SARS-CoV-2 lineages cocirculating with the Wuhan strains. Large trees with SARS-CoV-2 genomes beyond the first few months are needed to increase the likelihood of finding SARS-CoV-2 lineages that might have originated at the same time as (or even before) those early Wuhan strains. I extended a previously published rapid rooting method to model evolutionary rate as a linear function instead of a constant. This substantially improves the dating of the common ancestor of sampled SARS-CoV-2 genomes. Based on two large trees with 83,688 and 970,777 high-quality and full-length SARS-CoV-2 genomes that contain complete sample collection dates, the common ancestor was dated to 12 June 2019 and 7 July 2019 with the two trees, respectively. The two data sets would give dramatically different or even absurd estimates if the rate was treated as a constant. The large trees were also crucial for overcoming the high rate-heterogeneity among different viral lineages. The improved method was implemented in the software TRAD.


Subject(s)
COVID-19 , SARS-CoV-2 , SARS-CoV-2/genetics , Trees , Phylogeny , Evolution, Molecular
9.
New Microbiol ; 46(1): 1-8, 2023 Feb.
Article in English | MEDLINE | ID: covidwho-2267794

ABSTRACT

The study of characteristics, prevalence and patterns of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections is significant to monitor and define the status of the pandemic, helping to design and evaluate control strategies. In this setting, the continuous emergence of new variants and their dynamic of replacement underline the importance of implementing genomic epidemiology and phylogenetic methods for the molecular monitoring and surveillance of this new virus. The current profile of the pandemic can change rapidly when new variants emerge and spread, impacting epidemiology and public health in terms of prevention and treatment and making it necessary to develop new molecules and formulate vaccines. In this paper, we reviewed and synthesized the main studies on molecular genomics and phylogeny of SARS-CoV-2 during the pandemic, and highlighted their contributions to our understanding of this new emergent pathogen.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , Phylogeny , Pandemics , COVID-19/epidemiology , Genomics
10.
Elife ; 122023 04 04.
Article in English | MEDLINE | ID: covidwho-2273482

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a generalist virus, infecting and evolving in numerous mammals, including captive and companion animals, free-ranging wildlife, and humans. Transmission among non-human species poses a risk for the establishment of SARS-CoV-2 reservoirs, makes eradication difficult, and provides the virus with opportunities for new evolutionary trajectories, including the selection of adaptive mutations and the emergence of new variant lineages. Here, we use publicly available viral genome sequences and phylogenetic analysis to systematically investigate the transmission of SARS-CoV-2 between human and non-human species and to identify mutations associated with each species. We found the highest frequency of animal-to-human transmission from mink, compared with lower transmission from other sampled species (cat, dog, and deer). Although inferred transmission events could be limited by sampling biases, our results provide a useful baseline for further studies. Using genome-wide association studies, no single nucleotide variants (SNVs) were significantly associated with cats and dogs, potentially due to small sample sizes. However, we identified three SNVs statistically associated with mink and 26 with deer. Of these SNVs, ~⅔ were plausibly introduced into these animal species from local human populations, while the remaining ~⅓ were more likely derived in animal populations and are thus top candidates for experimental studies of species-specific adaptation. Together, our results highlight the importance of studying animal-associated SARS-CoV-2 mutations to assess their potential impact on human and animal health.


Subject(s)
COVID-19 , Deer , Animals , Cats , Dogs , SARS-CoV-2/genetics , COVID-19/genetics , Phylogeny , Mink/genetics , Genome-Wide Association Study , Deer/genetics , Zoonoses , Mutation , Genome, Viral
11.
Wall Street Journal - Online Edition ; : N.PAG-N.PAG, 2023.
Article in English | Academic Search Complete | ID: covidwho-2240706
12.
Microbiol Res ; 270: 127337, 2023 May.
Article in English | MEDLINE | ID: covidwho-2244307

ABSTRACT

The SARS-CoV-2 Hydra with many heads (variants) has been causing the COVID-19 pandemic for 3 years. The appearance of every new head (SARS-CoV-2 variant) causes a new pandemic wave. The last in the series is the XBB.1.5 "Kraken" variant. In the general public (social media) and in the scientific community (scientific journals), during the last several weeks since the variant has appeared, the question was raised of whether the infectivity of the new variant will be greater. This article attempts to provide the answer. Analysis of thermodynamic driving forces of binding and biosynthesis leads to the conclusion that infectivity of the XBB.1.5 variant could be increased to a certain extent. The pathogenicity of the XBB.1.5 variant seems to be unchanged compared to the other Omicron variants.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Pandemics
13.
Viruses ; 15(2)2023 02 20.
Article in English | MEDLINE | ID: covidwho-2240156

ABSTRACT

Brazil is one of the nations most affected by Coronavirus disease 2019 (COVID-19). The introduction and establishment of new virus variants can be related to an increase in cases and fatalities. The emergence of Omicron, the most modified SARS-CoV-2 variant, caused alarm for the public health of Brazil. In this study, we examined the effects of the Omicron introduction in Minas Gerais (MG), the second-most populous state of Brazil. A total of 430 Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) samples from November 2021 to June 2022 from Belo Horizonte (BH) city were sequenced. These newly sequenced genomes comprise 72% of all previously available SARS-CoV-2 genomes for the city. Evolutionary analysis of novel viral genomes reveals that a great diversity of Omicron sublineages have circulated in BH, a pattern in-keeping with observations across Brazil more generally. Bayesian phylogeographic reconstructions indicate that this diversity is a product of a large number of international and national importations. As observed previously, São Paulo state is shown as a significant hub for viral spread throughout the country, contributing to around 70% of all viral Omicron introductions detected in MG.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , Brazil/epidemiology , COVID-19/epidemiology , Bayes Theorem
14.
Combinatorial Chemistry and High Throughput Screening ; 26(1):2023/02/01 00:00:00.000, 2023.
Article in English | EMBASE | ID: covidwho-2232779
15.
Open Forum Infect Dis ; 10(2): ofad001, 2023 Feb.
Article in English | MEDLINE | ID: covidwho-2236756

ABSTRACT

Background: The limited variation observed among severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) consensus sequences makes it difficult to reconstruct transmission linkages in outbreak settings. Previous studies have recovered variation within individual SARS-CoV-2 infections but have not yet measured the informativeness of within-host variation for transmission inference. Methods: We performed tiled amplicon sequencing on 307 SARS-CoV-2 samples, including 130 samples from 32 individuals in 14 households and 47 longitudinally sampled individuals, from 4 prospective studies with household membership data, a proxy for transmission linkage. Results: Consensus sequences from households had limited diversity (mean pairwise distance, 3.06 single-nucleotide polymorphisms [SNPs]; range, 0-40). Most (83.1%, 255 of 307) samples harbored at least 1 intrahost single-nucleotide variant ([iSNV] median, 117; interquartile range [IQR], 17-208), above a minor allele frequency threshold of 0.2%. Pairs in the same household shared significantly more iSNVs (mean, 1.20 iSNVs; 95% confidence interval [CI], 1.02-1.39) than did pairs in different households infected with the same viral clade (mean, 0.31 iSNVs; 95% CI, .28-.34), a signal that decreases with increasingly stringent minor allele frequency thresholds. The number of shared iSNVs was significantly associated with an increased odds of household membership (adjusted odds ratio, 1.35; 95% CI, 1.23-1.49). However, the poor concordance of iSNVs detected across sequencing replicates (24.8% and 35.0% above a 0.2% and 1% threshold) confirms technical concerns that current sequencing and bioinformatic workflows do not consistently recover low-frequency within-host variants. Conclusions: Shared within-host variation may augment the information in consensus sequences for predicting transmission linkages. Improving sensitivity and specificity of within-host variant identification will improve the informativeness of within-host variation.

16.
Infect Genet Evol ; 108: 105405, 2023 03.
Article in English | MEDLINE | ID: covidwho-2236360

ABSTRACT

The COVID-19 pandemic has brought significant challenges for genomic surveillance strategies in public health systems worldwide. During the past thirty-four months, many countries faced several epidemic waves of SARS-CoV-2 infections, driven mainly by the emergence and spread of novel variants. In that line, genomic surveillance has been a crucial toolkit to study the real-time SARS-CoV-2 evolution, for the assessment and optimization of novel diagnostic assays, and to improve the efficacy of existing vaccines. During the pandemic, the identification of emerging lineages carrying lineage-specific mutations (particularly those in the Receptor Binding domain) showed how these mutations might significantly impact viral transmissibility, protection from reinfection and vaccination. So far, an unprecedented number of SARS-CoV-2 viral genomes has been released in public databases (i.e., GISAID, and NCBI), achieving 14 million genome sequences available as of early-November 2022. In the present review, we summarise the global landscape of SARS-CoV-2 during the first thirty-four months of viral circulation and evolution. It demonstrates the urgency and importance of sustained investment in genomic surveillance strategies to timely identify the emergence of any potential viral pathogen or associated variants, which in turn is key to epidemic and pandemic preparedness.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , Pandemics/prevention & control , COVID-19/epidemiology , COVID-19/prevention & control , Genomics , Databases, Factual , Mutation , Genome, Viral
17.
Comput Biol Med ; 152: 106264, 2023 01.
Article in English | MEDLINE | ID: covidwho-2177832

ABSTRACT

The widespread of SARS-CoV-2 presents a significant threat to human society, as well as public health and economic development. Extensive efforts have been undertaken to battle against the pandemic, whereas effective approaches such as vaccination would be weakened by the continuous mutations, leading to considerable attention being attracted to the mutation prediction. However, most previous studies lack attention to phylogenetics. In this paper, we propose a novel and effective model TEMPO for predicting the mutation of SARS-CoV-2 evolution. Specifically, we design a phylogenetic tree-based sampling method to generate sequence evolution data. Then, a transformer-based model is presented for the site mutation prediction after learning the high-level representation of these sequence data. We conduct experiments to verify the effectiveness of TEMPO, leveraging a large-scale SARS-CoV- 2 dataset. Experimental results show that TEMPO is effective for mutation prediction of SARS- CoV-2 evolution and outperforms several state-of-the-art baseline methods. We further perform mutation prediction experiments of other infectious viruses, to explore the feasibility and robustness of TEMPO, and experimental results verify its superiority. The codes and datasets are freely available at https://github.com/ZJUDataIntelligence/TEMPO.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/genetics , Phylogeny , Mutation , Pandemics
18.
Open Forum Infectious Diseases ; 9(Supplement 2):S746, 2022.
Article in English | EMBASE | ID: covidwho-2189907

ABSTRACT

Background. Global genomic surveillance has allowed identification of SARS-CoV-2 circulating variants responsible for the COVID-19 pandemic. Statewide variant characterization can guide local public health mitigations and provide educational opportunities. We characterized statewide evolution of SARS-CoV-2 variants in Rhode Island (RI). Methods. De identified RI SARS-CoV-2 sequences since 2/2020, generated at authors, CDC and commercial laboratories, were extracted from https://www.gisaid. org. Genomic and phylogenetic analyses were conducted with available tools and custom python scripts and, after quality control, sequences were classified as variants of Concern (VOC), variants being monitored (VBM), or non-VOC/ non-VBM, per CDC definitions. Specific mutations that are characteristic of the most recent VOCs (Delta or Omicron) were explored outside of their designated lineages. Results. Of the 1.1 million RI population, 14,933 SARS-CoV-2 sequences were available between 2/2020 and 3/2022. These included 1,542 (11%) sequences from 37 non-VOC/non-VBM lineages until 2/2021, most commonly B.1.2 (21%), B.1.375 (13%), and B.1.517 (6%);2,910 (19%) sequences from 7VBM lineages between 3-6/2021, most commonly Alpha (48%), Iota (34%), and Gamma (10%);and 10,481 (70%) sequences from 2 VOC lineages, including 7,574 (72%) Delta mostly between 6/2021 and 12/2021, and 2,907 (28%) Omicron mostly between 1/2022 and 3/2022. Phylogeny showed expected clustering of local variants within regional and global sequences, and continued viral evolution over time. Further VOC evolution was observed, including 87 Delta sub-lineages, most commonly AY.103 (17%), AY.3 (15%), and AY.44 (12%);and 4 Omicron sub-lineages BA.1 (61%), BA.1.1 (32%), BA.2 (7%), and BA.3 (< 1%). Omicron-associated mutations S:del69/70, S:H655Y, or N: P13L were observed in 219 Delta sequences, and Delta-associated mutations ORF1b: G662S, N:D377Y, or M:I82T were observed in 16 Omicron sequences. Conclusion. Statewide SARS-CoV-2 genomic surveillance allows for continued characterization of locally circulating variants and monitoring of viral evolution. Such data guide public health policies, inform the local health force, and mitigate the impact of SARS-CoV-2 on public health.

19.
Springer Protocols Handbooks ; : 189-198, 2022.
Article in English | EMBASE | ID: covidwho-2173513

ABSTRACT

Canine coronavirus (CCoV) is usually the cause of mild gastroenteritis in dogs and is known to have spread worldwide. In the last decade, as a consequence of the extraordinary large RNA genome, novel recombinant variants of CCoV have been found that are closely related to feline and porcine strains. Moreover highly virulent pantropic CCoV strains were recently identified in dogs. The molecular characterization of the CCoV circulating in canine population is essential for understanding viral evolution. Copyright © Springer Science+Business Media New York 2016.

20.
Front Med (Lausanne) ; 9: 1061142, 2022.
Article in English | MEDLINE | ID: covidwho-2199002

ABSTRACT

Introduction: The current coronavirus pandemic is being combated worldwide by nontherapeutic measures and massive vaccination programs. Nevertheless, therapeutic options such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main-protease (Mpro) inhibitors are essential due to the ongoing evolution toward escape from natural or induced immunity. While antiviral strategies are vulnerable to the effects of viral mutation, the relatively conserved Mpro makes an attractive drug target: Nirmatrelvir, an antiviral targeting its active site, has been authorized for conditional or emergency use in several countries since December 2021, and a number of other inhibitors are under clinical evaluation. We analyzed recent SARS-CoV-2 genomic data, since early detection of potential resistances supports a timely counteraction in drug development and deployment, and discovered accelerated mutational dynamics of Mpro since early December 2021. Methods: We performed a comparative analysis of 10.5 million SARS-CoV-2 genome sequences available by June 2022 at GISAID to the NCBI reference genome sequence NC_045512.2. Amino-acid exchanges within high-quality regions in 69,878 unique Mpro sequences were identified and time- and in-depth sequence analyses including a structural representation of mutational dynamics were performed using in-house software. Results: The analysis showed a significant recent event of mutational dynamics in Mpro. We report a remarkable increase in mutational variability in an eight-residue long consecutive region (R188-G195) near the active site since December 2021. Discussion: The increased mutational variability in close proximity to an antiviral-drug binding site as described herein may suggest the onset of the development of antiviral resistance. This emerging diversity urgently needs to be further monitored and considered in ongoing drug development and lead optimization.

SELECTION OF CITATIONS
SEARCH DETAIL