Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Intervalo de año de publicación
1.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-22282629

RESUMEN

In many regions of the world, the Alpha, Beta and Gamma SARS-CoV-2 Variants of Concern (VOCs) co-circulated during 2020-21 and fueled waves of infections. During 2021, these variants were almost completely displaced by the Delta variant, causing a third wave of infections worldwide. This phenomenon of global viral lineage displacement was observed again in late 2021, when the Omicron variant disseminated globally. In this study, we use phylogenetic and phylogeographic methods to reconstruct the dispersal patterns of SARS-CoV-2 VOCs worldwide. We find that the source-sink dynamics of SARS-CoV-2 varied substantially by VOC, and identify countries that acted as global hubs of variant dissemination, while other countries became regional contributors to the export of specific variants. We demonstrate a declining role of presumed origin countries of VOCs to their global dispersal: we estimate that India contributed <15% of all global exports of Delta to other countries and South Africa <1-2% of all global Omicron exports globally. We further estimate that >80 countries had received introductions of Omicron BA.1 100 days after its inferred date of emergence, compared to just over 25 countries for the Alpha variant. This increased speed of global dissemination was associated with a rebound in air travel volume prior to Omicron emergence in addition to the higher transmissibility of Omicron relative to Alpha. Our study highlights the importance of global and regional hubs in VOC dispersal, and the speed at which highly transmissible variants disseminate through these hubs, even before their detection and characterization through genomic surveillance. HighlightsO_LIGlobal phylogenetic analysis reveals relationship between air travel and speed of dispersal of SARS-CoV-2 variants of concern (VOCs) C_LIO_LIOmicron VOC spread to 5x more countries within 100 days of its emergence compared to all other VOCs C_LIO_LIOnward transmission and dissemination of VOCs Delta and Omicron was primarily from secondary hubs rather than initial country of detection during a time of increased global air travel C_LIO_LIAnalysis highlights highly connected countries identified as major global and regional exporters of VOCs C_LI

2.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21267606

RESUMEN

The Delta variant of concern of SARS-CoV-2 has spread globally causing large outbreaks and resurgences of COVID-19 cases1-3. The emergence of Delta in the UK occurred on the background of a heterogeneous landscape of immunity and relaxation of non-pharmaceutical interventions4,5. Here we analyse 52,992 Delta genomes from England in combination with 93,649 global genomes to reconstruct the emergence of Delta, and quantify its introduction to and regional dissemination across England, in the context of changing travel and social restrictions. Through analysis of human movement, contact tracing, and virus genomic data, we find that the focus of geographic expansion of Delta shifted from India to a more global pattern in early May 2021. In England, Delta lineages were introduced >1,000 times and spread nationally as non-pharmaceutical interventions were relaxed. We find that hotel quarantine for travellers from India reduced onward transmission from importations; however the transmission chains that later dominated the Delta wave in England had been already seeded before restrictions were introduced. In England, increasing inter-regional travel drove Deltas nationwide dissemination, with some cities receiving >2,000 observable lineage introductions from other regions. Subsequently, increased levels of local population mixing, not the number of importations, was associated with faster relative growth of Delta. Among US states, we find that regions that previously experienced large waves also had faster Delta growth rates, and a model including interactions between immunity and human behaviour could accurately predict the rise of Delta there. Deltas invasion dynamics depended on fine scale spatial heterogeneity in immunity and contact patterns and our findings will inform optimal spatial interventions to reduce transmission of current and future VOCs such as Omicron.

3.
Raquel Viana; Sikhulile Moyo; Daniel Gyamfi Amoako; Houriiyah Tegally; Cathrine Scheepers; Richard J Lessells; Jennifer Giandhari; Nicole Wolter; Josie Everatt; Andrew Rambaut; Christian Althaus; Eduan Wilkinson; Adriano Mendes; Amy Strydom; Michaela Davids; Simnikiwe Mayaphi; Simani Gaseitsiwe; Wonderful T Choga; Dorcas Maruapula; Boitumelo Zuze; Botshelo Radibe; Legodile Koopile; Roger Shapiro; Shahin Lockman; Mpaphi B. Mbulawa; Thongbotho Mphoyakgosi; Pamela Smith-Lawrence; Mosepele Mosepele; Mogomotsi Matshaba; Kereng Masupu; Mohammed Chand; Charity Joseph; Lesego Kuate-Lere; Onalethatha Lesetedi-Mafoko; Kgomotso Moruisi; Lesley Scott; Wendy Stevens; Constantinos Kurt Wibmer; Anele Mnguni; Arshad Ismail; Boitshoko Mahlangu; Darren P. Martin; Verity Hill; Rachel Colquhoun; Modisa S. Motswaledi; James Emmanuel San; Noxolo Ntuli; Gerald Motsatsi; Sureshnee Pillay; Thabo Mohale; Upasana Ramphal; Yeshnee Naidoo; Naume Tebeila; Marta Giovanetti; Koleka Mlisana; Carolyn Williamson; Nei-yuan Hsiao; Nokukhanya Msomi; Kamela Mahlakwane; Susan Engelbrecht; Tongai Maponga; Wolfgang Preiser; Zinhle Makatini; Oluwakemi Laguda-Akingba; Lavanya Singh; Ugochukwu J. Anyaneji; Monika Moir; Stephanie van Wyk; Derek Tshiabuila; Yajna Ramphal; Arisha Maharaj; Sergei Pond; Alexander G Lucaci; Steven Weaver; Maciej F Boni; Koen Deforche; Kathleen Subramoney; Diana Hardie; Gert Marais; Deelan Doolabh; Rageema Joseph; Nokuzola Mbhele; Luicer Olubayo; Arash Iranzadeh; Alexander E Zarebski; Joseph Tsui; Moritz UG Kraemer; Oliver G Pybus; Dominique Goedhals; Phillip Armand Bester; Martin M Nyaga; Peter N Mwangi; Allison Glass; Florette Treurnicht; Marietjie Venter; Jinal N. Bhiman; Anne von Gottberg; Tulio de Oliveira.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21268028

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) epidemic in southern Africa has been characterised by three distinct waves. The first was associated with a mix of SARS-CoV-2 lineages, whilst the second and third waves were driven by the Beta and Delta variants respectively1-3. In November 2021, genomic surveillance teams in South Africa and Botswana detected a new SARS-CoV-2 variant associated with a rapid resurgence of infections in Gauteng Province, South Africa. Within three days of the first genome being uploaded, it was designated a variant of concern (Omicron) by the World Health Organization and, within three weeks, had been identified in 87 countries. The Omicron variant is exceptional for carrying over 30 mutations in the spike glycoprotein, predicted to influence antibody neutralization and spike function4. Here, we describe the genomic profile and early transmission dynamics of Omicron, highlighting the rapid spread in regions with high levels of population immunity.

4.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21260122

RESUMEN

We report the first local transmission of the SARS-CoV-2 Delta variant in mainland China. All 167 infections could be traced back to the first index case. Daily sequential PCR testing of the quarantined subjects indicated that the viral loads of Delta infections, when they first become PCR+, were on average [~]1000 times greater compared to A/B lineage infections during initial epidemic wave in China in early 2020, suggesting potentially faster viral replication and greater infectiousness of Delta during early infection. We performed high-quality sequencing on samples from 126 individuals. Reliable epidemiological data meant that, for 111 transmission events, the donor and recipient cases were known. The estimated transmission bottleneck size was 1-3 virions with most minor intra-host single nucleotide variants (iSNVs) failing to transmit to the recipients. However, transmission heterogeneity of SARS-CoV-2 was also observed. The transmission of minor iSNVs resulted in at least 4 of the 30 substitutions identified in the outbreak, highlighting the contribution of intra-host variants to population level viral diversity during rapid spread. Disease control activities, such as the frequency of population testing, quarantine during pre-symptomatic infection, and level of virus genomic surveillance should be adjusted in order to account for the increasing prevalence of the Delta variant worldwide.

5.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21258229

RESUMEN

Vaccines provide powerful tools to mitigate the enormous public health and economic costs that the ongoing SARS-CoV-2 pandemic continues to exert globally, yet vaccine distribution remains unequal between countries. To examine the potential epidemiological and evolutionary impacts of vaccine nationalism, we extend previous models to include simple scenarios of stockpiling. In general, we find that stockpiling vaccines by countries with high availability leads to large increases in infections in countries with low vaccine availability, the magnitude of which depends on the strength and duration of natural and vaccinal immunity. Additionally, a number of subtleties arise when the populations and transmission rates in each country differ depending on evolutionary assumptions and vaccine availability. Furthermore, the movement of infected individuals between countries combined with the possibility of increases in viral transmissibility may greatly magnify local and combined infection numbers, suggesting that countries with high vaccine availability must invest in surveillance strategies to prevent case importation. Dose-sharing is likely a high-return strategy because equitable allocation brings non-linear benefits and also alleviates costs of surveillance (e.g. border testing, genomic surveillance) in settings where doses are sufficient to maintain cases at low numbers. Across a range of immunological scenarios, we find that vaccine sharing is also a powerful tool to decrease the potential for antigenic evolution, especially if infections after the waning of natural immunity contribute most to evolutionary potential. Overall, our results stress the importance of equitable global vaccine distribution.

6.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-445313

RESUMEN

Comparison of evolution among related viruses can provide insights into shared adaptive processes, for example following host switching to a mutual host species. Whilst phylogenetic methods can help identify mutations that may be important for evolutionary processes such as adaptation to a new host, these can be enhanced by positioning candidate mutations to known functional sites on protein structures. Over the past two decades, three zoonotic betacoronaviruses have significantly impacted human public health: SARS-CoV-1, MERS-CoV and SARS-CoV-2, whilst two other betacoronaviruses, HKU1 and OC43, have circulated endemically in the human population for over 100 years. In this study, we use a comparative approach to prospectively search for potentially evolutionarily-relevant mutations within the Orf1ab and S genes across betacoronavirus species that have demonstrated sustained human-to-human transmission (HKU1, OC43, SARS-CoV-1 and SARS-CoV-2). We used a combination of molecular evolution methods to identify 30 sites that display evidence of homoplasy and/or stepwise evolution, that may be suggestive of adaptation across emerging and endemic betacoronaviruses. Of these, seven sites also display evidence of being selectively relevant. Drawing upon known protein structure data, we find that four of the identified mutations [18121 (exonuclease/27), 21623 (spike/21), 21635 (spike/25) and 23948 (spike/796), in SARS-CoV-2 genome coordinates] are proximal to regions of known functionality. Our results provide a molecular-level context for common evolutionary pathways that betacoronaviruses may undergo during adaptation to the human host.

7.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21254685

RESUMEN

Characterisation of SARS-CoV-2 genetic diversity through space and time can reveal trends in virus importation and domestic circulation, and permit the exploration of questions regarding the early transmission dynamics. Here we present a detailed description of SARS-CoV-2 genomic epidemiology in Ecuador, one of the hardest hit countries during the early stages of the COVID-19 pandemic. We generate and analyse 160 whole genome sequences sampled from all provinces of Ecuador in 2020. Molecular clock and phylgeographic analysis of these sequences in the context of global SARS-CoV-2 diversity enable us to identify and characterise individual transmission lineages within Ecuador, explore their spatiotemporal distributions, and consider their introduction and domestic circulation. Our results reveal a pattern of multiple international importations across the country, with apparent differences between key provinces. Transmission lineages were mostly introduced before the implementation of non-pharmaceutical interventions (NPIs), with differential degrees of persistence and national dissemination.

8.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21252554

RESUMEN

Cases of SARS-CoV-2 infection in Manaus, Brazil, resurged in late 2020, despite high levels of previous infection there. Through genome sequencing of viruses sampled in Manaus between November 2020 and January 2021, we identified the emergence and circulation of a novel SARS-CoV-2 variant of concern, lineage P.1, that acquired 17 mutations, including a trio in the spike protein (K417T, E484K and N501Y) associated with increased binding to the human ACE2 receptor. Molecular clock analysis shows that P.1 emergence occurred around early November 2020 and was preceded by a period of faster molecular evolution. Using a two-category dynamical model that integrates genomic and mortality data, we estimate that P.1 may be 1.4-2.2 times more transmissible and 25-61% more likely to evade protective immunity elicited by previous infection with non-P.1 lineages. Enhanced global genomic surveillance of variants of concern, which may exhibit increased transmissibility and/or immune evasion, is critical to accelerate pandemic responsiveness. One-Sentence SummaryWe report the evolution and emergence of a SARS-CoV-2 lineage of concern associated with rapid transmission in Manaus.

9.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-20194787

RESUMEN

The herd immunity threshold is the proportion of a population that must be immune to an infectious disease, either by natural infection or vaccination such that, in the absence of additional preventative measures, new cases decline and the effective reproduction number falls below unity. This fundamental epidemiological parameter is still unknown for the recently-emerged COVID-19, and mathematical models have predicted very divergent results. Population studies using antibody testing to infer total cumulative infections can provide empirical evidence of the level of population immunity in severely affected areas. Here we show that the transmission of SARS-CoV-2 in Manaus, located in the Brazilian Amazon, increased quickly during March and April and declined more slowly from May to September. In June, one month following the epidemic peak, 44% of the population was seropositive for SARS-CoV-2, equating to a cumulative incidence of 52%, after correcting for the false-negative rate of the antibody test. The seroprevalence fell in July and August due to antibody waning. After correcting for this, we estimate a final epidemic size of 66%. Although non-pharmaceutical interventions, plus a change in population behavior, may have helped to limit SARS-CoV-2 transmission in Manaus, the unusually high infection rate suggests that herd immunity played a significant role in determining the size of the epidemic.

10.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-20127043

RESUMEN

Using 65 transmission pairs of SARS-CoV-2 reported to the Brazilian Ministry of Health we estimate the mean and standard deviation for the serial interval to be 2.97 and 3.29 days respectively. We also present a model for the serial interval probability distribution using only two parameters.

11.
Darlan da Silva Candido; Ingra Morales Claro; Jaqueline Goes de Jesus; William Marciel de Souza; Filipe Romero Rebello Moreira; Simon Dellicour; Thomas A. Mellan; Louis du Plessis; Rafael Henrique Moraes Pereira; Flavia Cristina da Silva Sales; Erika Regina Manuli; Julien Theze; Luis Almeida; Mariane Talon de Menezes; Carolina Moreira Voloch; Marcilio Jorge Fumagalli; Thais de Moura Coletti; Camila Alves Maia Silva; Mariana Severo Ramundo; Mariene Ribeiro Amorim; Henrique Hoeltgebaum; Swapnil Mishra; Mandev Gill; Luiz Max Carvalho; Lewis Fletcher Buss; Carlos Augusto Prete Jr.; Jordan Ashworth; Helder Nakaya; Pedro da Silva Peixoto; Oliver J Brady; Samuel M. Nicholls; Amilcar Tanuri; Atila Duque Rossi; Carlos Kaue Vieira Braga; Alexandra Lehmkuhl Gerber; Ana Paula Guimaraes; Nelson Gaburo Jr.; Cecilia Salete Alencar; Alessandro Clayton de Souza Ferreira; Cristiano Xavier Lima; Jose Eduardo Levi; Celso Granato; Giula Magalhaes Ferreira; Ronaldo da Silva Francisco Jr.; Fabiana Granja; Marcia Teixeira Garcia; Maria Luiza Moretti; Mauricio Wesley Perroud Jr.; Terezinha Marta Pereira Pinto Castineiras; Carolina Dos Santos Lazari; Sarah C Hill; Andreza Aruska de Souza Santos; Camila Lopes Simeoni; Julia Forato; Andrei Carvalho Sposito; Angelica Zaninelli Schreiber; Magnun Nueldo Nunes Santos; Camila Zolini Sa; Renan Pedra Souza; Luciana Cunha Resende Moreira; Mauro Martins Teixeira; Josy Hubner; Patricia Asfora Falabella Leme; Rennan Garcias Moreira; Mauricio Lacerda Nogueira; - CADDE-Genomic-Network; Neil Ferguson; Silvia Figueiredo Costa; Jose Luiz Proenca-Modena; Ana Tereza Vasconcelos; Samir Bhatt; Philippe Lemey; Chieh-Hsi Wu; Andrew Rambaut; Nick J Loman; Renato Santana Aguiar; Oliver G Pybus; Ester Cerdeira Sabino; Nuno Rodrigues Faria.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-20128249

RESUMEN

Brazil currently has one of the fastest growing SARS-CoV-2 epidemics in the world. Due to limited available data, assessments of the impact of non-pharmaceutical interventions (NPIs) on virus transmission and epidemic spread remain challenging. We investigate the impact of NPIs in Brazil using epidemiological, mobility and genomic data. Mobility-driven transmission models for Sao Paulo and Rio de Janeiro cities show that the reproduction number (Rt) reached below 1 following NPIs but slowly increased to values between 1 to 1.3 (1.0-1.6). Genome sequencing of 427 new genomes and analysis of a geographically representative genomic dataset from 21 of the 27 Brazilian states identified >100 international introductions of SARS-CoV-2 in Brazil. We estimate that three clades introduced from Europe emerged between 22 and 27 February 2020, and were already well-established before the implementation of NPIs and travel bans. During this first phase of the epidemic establishment of SARS-CoV-2 in Brazil, we find that the virus spread mostly locally and within-state borders. Despite sharp decreases in national air travel during this period, we detected a 25% increase in the average distance travelled by air passengers during this time period. This coincided with the spread of SARS-CoV-2 from large urban centers to the rest of the country. In conclusion, our results shed light on the role of large and highly connected populated centres in the rapid ignition and establishment of SARS-CoV-2, and provide evidence that current interventions remain insufficient to keep virus transmission under control in Brazil. One Sentence SummaryJoint analysis of genomic, mobility and epidemiological novel data provide unique insight into the spread and transmission of the rapidly evolving epidemic of SARS-CoV-2 in Brazil.

12.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-015941

RESUMEN

Two notable features have been identified in the SARS-CoV-2 genome: (1) the receptor binding domain of SARS-CoV-2; (2) a unique insertion of twelve nucleotide or four amino acids (PRRA) at the S1 and S2 boundary. For the first feature, the similar RBD identified in SARs-like virus from pangolin suggests the RBD in SARS-CoV-2 may already exist in animal host(s) before it transmitted into human. The left puzzle is the history and function of the insertion at S1/S2 boundary, which is uniquely identified in SARS-CoV-2. In this study, we identified two variants from the first Guangdong SARS-CoV-2 cell strain, with deletion mutations on polybasic cleavage site (PRRAR) and its flank sites. More extensive screening indicates the deletion at the flank sites of PRRAR could be detected in 3 of 68 clinical samples and half of 22 in vitro isolated viral strains. These data indicate (1) the deletion of QTQTN, at the flank of polybasic cleavage site, is likely benefit the SARS-CoV-2 replication or infection in vitro but under strong purification selection in vivo since it is rarely identified in clinical samples; (2) there could be a very efficient mechanism for deleting this region from viral genome as the variants losing 23585-23599 is commonly detected after two rounds of cell passage. The mechanistic explanation for this in vitro adaptation and in vivo purification processes (or reverse) that led to such genomic changes in SARS-CoV-2 requires further work. Nonetheless, this study has provided valuable clues to aid further investigation of spike protein function and virus evolution. The deletion mutation identified in vitro isolation should be also noted for current vaccine development.

13.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-20036392

RESUMEN

HighlightThe global outbreak caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been declared a pandemic by the WHO. As the number of imported SARS-CoV-2 cases is on the rise in Brazil, we use incidence and historical air travel data to estimate the most important routes of importation into the country.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...