Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.649
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 187(17): 4546-4548, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39111311

RESUMO

H5N1 is an avian influenza virus that causes respiratory disease in birds and several land and sea mammals. The recent outbreak in the United States, including infection of dairy workers, has increased the concern around potential transmission and spread. We asked virologists, epidemiologists, and public health experts what the most urgent questions and action points are at this stage of the outbreak.


Assuntos
Surtos de Doenças , Virus da Influenza A Subtipo H5N1 , Influenza Aviária , Influenza Humana , Humanos , Influenza Humana/epidemiologia , Influenza Humana/transmissão , Influenza Humana/virologia , Animais , Virus da Influenza A Subtipo H5N1/patogenicidade , Virus da Influenza A Subtipo H5N1/genética , Estados Unidos/epidemiologia , Influenza Aviária/virologia , Influenza Aviária/transmissão , Influenza Aviária/epidemiologia , Aves/virologia
2.
Cell ; 184(8): 1960-1961, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33831378

RESUMO

The events of the past year have underscored the serious and rapid threat that emerging viruses pose to global health. However, much of the rapid progress in understanding and combating SARS-CoV-2 was made possible because of the decades of important groundwork laid from researchers studying other emergent infectious diseases. The 2021 John Dirks Canada Gairdner Global Health award recognizes the contributions of Joseph Sriyal Malik Peiris and Yi Guan toward understanding the origins and options for control of newly emerging infectious disease outbreaks in Asia, notably zoonotic influenza and severe acute respiratory syndrome (SARS). Cell's Nicole Neuman corresponded with Yi Guan about his path to becoming a viral infection sleuth and the challenges of understanding emerging pathogens and their origins. Excerpts of their exchange are included here.


Assuntos
COVID-19 , Doenças Transmissíveis Emergentes , Surtos de Doenças , Influenza Humana , Zoonoses , Animais , Ásia , COVID-19/epidemiologia , COVID-19/transmissão , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/história , Doenças Transmissíveis Emergentes/transmissão , Surtos de Doenças/história , Saúde Global , História do Século XXI , Humanos , Influenza Humana/epidemiologia , Influenza Humana/história , Influenza Humana/transmissão , Zoonoses/epidemiologia , Zoonoses/transmissão
3.
Nature ; 633(8029): 426-432, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38977017

RESUMO

Highly pathogenic H5N1 avian influenza (HPAI H5N1) viruses occasionally infect, but typically do not transmit, in mammals. In the spring of 2024, an unprecedented outbreak of HPAI H5N1 in bovine herds occurred in the USA, with virus spread within and between herds, infections in poultry and cats, and spillover into humans, collectively indicating an increased public health risk1-4. Here we characterize an HPAI H5N1 virus isolated from infected cow milk in mice and ferrets. Like other HPAI H5N1 viruses, the bovine H5N1 virus spread systemically, including to the mammary glands of both species, however, this tropism was also observed for an older HPAI H5N1 virus isolate. Bovine HPAI H5N1 virus bound to sialic acids expressed in human upper airways and inefficiently transmitted to exposed ferrets (one of four exposed ferrets seroconverted without virus detection). Bovine HPAI H5N1 virus thus possesses features that may facilitate infection and transmission in mammals.


Assuntos
Doenças dos Bovinos , Virus da Influenza A Subtipo H5N1 , Infecções por Orthomyxoviridae , Virulência , Animais , Bovinos , Feminino , Humanos , Camundongos , Furões/virologia , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Virus da Influenza A Subtipo H5N1/patogenicidade , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Humana/transmissão , Influenza Humana/virologia , Influenza Humana/epidemiologia , Glândulas Mamárias Animais/virologia , Camundongos Endogâmicos BALB C , Leite/virologia , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/transmissão , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/virologia , Ácidos Siálicos/metabolismo , Tropismo Viral , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/transmissão , Doenças dos Bovinos/virologia , Estados Unidos/epidemiologia , Zoonoses Virais , Soroconversão , Máscaras Laríngeas/virologia
4.
Cell ; 157(2): 329-339, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24725402

RESUMO

Recently, A/H5N1 influenza viruses were shown to acquire airborne transmissibility between ferrets upon targeted mutagenesis and virus passage. The critical genetic changes in airborne A/Indonesia/5/05 were not yet identified. Here, five substitutions proved to be sufficient to determine this airborne transmission phenotype. Substitutions in PB1 and PB2 collectively caused enhanced transcription and virus replication. One substitution increased HA thermostability and lowered the pH of membrane fusion. Two substitutions independently changed HA binding preference from α2,3-linked to α2,6-linked sialic acid receptors. The loss of a glycosylation site in HA enhanced overall binding to receptors. The acquired substitutions emerged early during ferret passage as minor variants and became dominant rapidly. Identification of substitutions that are essential for airborne transmission of avian influenza viruses between ferrets and their associated phenotypes advances our fundamental understanding of virus transmission and will increase the value of future surveillance programs and public health risk assessments.


Assuntos
Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Humana/transmissão , Influenza Humana/virologia , Substituição de Aminoácidos , Animais , Furões , Genoma Viral , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Virus da Influenza A Subtipo H5N1/genética , Mutação , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Receptores Virais/metabolismo , Seleção Genética
5.
Nature ; 619(7969): 338-347, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37380775

RESUMO

Spillover events of avian influenza A viruses (IAVs) to humans could represent the first step in a future pandemic1. Several factors that limit the transmission and replication of avian IAVs in mammals have been identified. There are several gaps in our understanding to predict which virus lineages are more likely to cross the species barrier and cause disease in humans1. Here, we identified human BTN3A3 (butyrophilin subfamily 3 member A3)2 as a potent inhibitor of avian IAVs but not human IAVs. We determined that BTN3A3 is expressed in human airways and its antiviral activity evolved in primates. We show that BTN3A3 restriction acts primarily at the early stages of the virus life cycle by inhibiting avian IAV RNA replication. We identified residue 313 in the viral nucleoprotein (NP) as the genetic determinant of BTN3A3 sensitivity (313F or, rarely, 313L in avian viruses) or evasion (313Y or 313V in human viruses). However, avian IAV serotypes, such as H7 and H9, that spilled over into humans also evade BTN3A3 restriction. In these cases, BTN3A3 evasion is due to substitutions (N, H or Q) in NP residue 52 that is adjacent to residue 313 in the NP structure3. Thus, sensitivity or resistance to BTN3A3 is another factor to consider in the risk assessment of the zoonotic potential of avian influenza viruses.


Assuntos
Aves , Interações entre Hospedeiro e Microrganismos , Vírus da Influenza A , Influenza Aviária , Influenza Humana , Zoonoses Virais , Animais , Humanos , Aves/virologia , Vírus da Influenza A/classificação , Vírus da Influenza A/genética , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/isolamento & purificação , Influenza Aviária/transmissão , Influenza Aviária/virologia , Influenza Humana/prevenção & controle , Influenza Humana/transmissão , Influenza Humana/virologia , Primatas , Sistema Respiratório/metabolismo , Sistema Respiratório/virologia , Medição de Risco , Zoonoses Virais/prevenção & controle , Zoonoses Virais/transmissão , Zoonoses Virais/virologia , Replicação Viral
6.
Cell ; 153(7): 1475-85, 2013 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-23746829

RESUMO

Of the factors governing human-to-human transmission of the highly pathogenic avian-adapted H5N1 virus, the most critical is the acquisition of mutations on the viral hemagglutinin (HA) to "quantitatively switch" its binding from avian to human glycan receptors. Here, we describe a structural framework that outlines a necessary set of H5 HA receptor-binding site (RBS) features required for the H5 HA to quantitatively switch its preference to human receptors. We show here that the same RBS HA mutations that lead to aerosol transmission of A/Vietnam/1203/04 and A/Indonesia/5/05 viruses, when introduced in currently circulating H5N1, do not lead to a quantitative switch in receptor preference. We demonstrate that HAs from circulating clades require as few as a single base pair mutation to quantitatively switch their binding to human receptors. The mutations identified by this study can be used to monitor the emergence of strains having human-to-human transmission potential.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Virus da Influenza A Subtipo H5N1/química , Influenza Aviária/virologia , Influenza Humana/transmissão , Influenza Humana/virologia , Sequência de Aminoácidos , Animais , Aves , Evolução Molecular , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Especificidade de Hospedeiro , Humanos , Virus da Influenza A Subtipo H5N1/patogenicidade , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Humana/epidemiologia , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ácido N-Acetilneuramínico/metabolismo , Filogenia , Receptores Virais/química , Receptores Virais/metabolismo , Alinhamento de Sequência
7.
Proc Natl Acad Sci U S A ; 121(33): e2322660121, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39361828

RESUMO

Sustained community spread of influenza viruses relies on efficient person-to-person transmission. Current experimental transmission systems do not mimic environmental conditions (e.g., air exchange rates, flow patterns), host behaviors, or exposure durations relevant to real-world settings. Therefore, results from these traditional systems may not be representative of influenza virus transmission in humans. To address this pitfall, we developed a close-range transmission setup that implements a play-based scenario and used it to investigate the impact of ventilation rates on transmission. In this setup, four immunologically naive recipient ferrets were exposed to a donor ferret infected with a genetically barcoded 2009 H1N1 virus (H1N1pdm09) for 4 h. The ferrets interacted in a shared space that included toys, similar to a childcare setting. Transmission efficiency was assessed under low and high ventilation, with air exchange rates of ~1.3 h-1 and 23 h-1, respectively. Transmission efficiencies observed in three independent replicate studies were similar between ventilation conditions. The presence of infectious virus or viral RNA on surfaces and in air throughout the exposure area was also not impacted by the ventilation rate. While high viral genetic diversity in donor ferret nasal washes was maintained during infection, recipient ferret nasal washes displayed low diversity, revealing a narrow transmission bottleneck regardless of ventilation rate. Examining the frequency and duration of ferret physical touches revealed no link between these interactions and a successful transmission event. Our findings indicate that exposures characterized by frequent, close-range interactions and the presence of fomites can overcome the benefits of increased ventilation.


Assuntos
Furões , Vírus da Influenza A Subtipo H1N1 , Infecções por Orthomyxoviridae , Ventilação , Animais , Furões/virologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H1N1/genética , Infecções por Orthomyxoviridae/transmissão , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/veterinária , Masculino , Influenza Humana/transmissão , Influenza Humana/virologia , Feminino , Humanos
8.
J Virol ; 98(7): e0040924, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38869284

RESUMO

Aerosol transmission remains a major challenge for control of respiratory viruses, particularly those causing recurrent epidemics, like influenza A virus (IAV). These viruses are rarely expelled alone, but instead are embedded in a consortium of microorganisms that populate the respiratory tract. The impact of microbial communities and inter-pathogen interactions upon stability of transmitted viruses is well-characterized for enteric pathogens, but is under-studied in the respiratory niche. Here, we assessed whether the presence of five different species of commensal respiratory bacteria could influence the persistence of IAV within phosphate-buffered saline and artificial saliva droplets deposited on surfaces at typical indoor air humidity, and within airborne aerosol particles. In droplets, presence of individual species or a mixed bacterial community resulted in 10- to 100-fold more infectious IAV remaining after 1 h, due to bacterial-mediated flattening of drying droplets and early efflorescence. Even when no efflorescence occurred at high humidity or the bacteria-induced changes in droplet morphology were abolished by aerosolization instead of deposition on a well plate, the bacteria remained protective. Staphylococcus aureus and Streptococcus pneumoniae were the most stabilizing compared to other commensals at equivalent density, indicating the composition of an individual's respiratory microbiota is a previously unconsidered factor influencing expelled virus persistence.IMPORTANCEIt is known that respiratory infections such as coronavirus disease 2019 and influenza are transmitted by release of virus-containing aerosols and larger droplets by an infected host. The survival time of viruses expelled into the environment can vary depending on temperature, room air humidity, UV exposure, air composition, and suspending fluid. However, few studies consider the fact that respiratory viruses are not alone in the respiratory tract-we are constantly colonized by a plethora of bacteria in our noses, mouth, and lower respiratory system. In the gut, enteric viruses are known to be stabilized against inactivation and environmental decay by gut bacteria. Despite the presence of a similarly complex bacterial microbiota in the respiratory tract, few studies have investigated whether viral stabilization could occur in this niche. Here, we address this question by investigating influenza A virus stabilization by a range of commensal bacteria in systems representing respiratory aerosols and droplets.


Assuntos
Aerossóis , Vírus da Influenza A , Vírus da Influenza A/fisiologia , Humanos , Staphylococcus aureus/fisiologia , Streptococcus pneumoniae/fisiologia , Sistema Respiratório/microbiologia , Sistema Respiratório/virologia , Animais , Influenza Humana/virologia , Influenza Humana/transmissão , Bactérias , Microbiota , Cães , Simbiose , Células Madin Darby de Rim Canino
9.
PLoS Comput Biol ; 20(7): e1012311, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39083536

RESUMO

Like other tropical and subtropical regions, influenza viruses can circulate year-round in Hong Kong. However, during the COVID-19 pandemic, there was a significant decrease in influenza activity. The objective of this study was to retrospectively forecast influenza activity during the year 2020 and assess the impact of COVID-19 public health social measures (PHSMs) on influenza activity and hospital admissions in Hong Kong. Using weekly surveillance data on influenza virus activity in Hong Kong from 2010 to 2019, we developed a statistical modeling framework to forecast influenza virus activity and associated hospital admissions. We conducted short-term forecasts (1-4 weeks ahead) and medium-term forecasts (1-13 weeks ahead) for the year 2020, assuming no PHSMs were implemented against COVID-19. We estimated the reduction in transmissibility, peak magnitude, attack rates, and influenza-associated hospitalization rate resulting from these PHSMs. For short-term forecasts, mean ambient ozone concentration and school holidays were found to contribute to better prediction performance, while absolute humidity and ozone concentration improved the accuracy of medium-term forecasts. We observed a maximum reduction of 44.6% (95% CI: 38.6% - 51.9%) in transmissibility, 75.5% (95% CI: 73.0% - 77.6%) in attack rate, 41.5% (95% CI: 13.9% - 55.7%) in peak magnitude, and 63.1% (95% CI: 59.3% - 66.3%) in cumulative influenza-associated hospitalizations during the winter-spring period of the 2019/2020 season in Hong Kong. The implementation of PHSMs to control COVID-19 had a substantial impact on influenza transmission and associated burden in Hong Kong. Incorporating information on factors influencing influenza transmission improved the accuracy of our predictions.


Assuntos
COVID-19 , Previsões , Hospitalização , Influenza Humana , Pandemias , SARS-CoV-2 , Estações do Ano , Humanos , Hong Kong/epidemiologia , Influenza Humana/epidemiologia , Influenza Humana/transmissão , COVID-19/epidemiologia , COVID-19/transmissão , Hospitalização/estatística & dados numéricos , Previsões/métodos , Estudos Retrospectivos , Modelos Estatísticos , Biologia Computacional
12.
Proc Natl Acad Sci U S A ; 119(37): e2203019119, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36074818

RESUMO

The global spread of coronavirus disease 2019 (COVID-19) has emphasized the need for evidence-based strategies for the safe operation of schools during pandemics that balance infection risk with the society's responsibility of allowing children to attend school. Due to limited empirical data, existing analyses assessing school-based interventions in pandemic situations often impose strong assumptions, for example, on the relationship between class size and transmission risk, which could bias the estimated effect of interventions, such as split classes and staggered attendance. To fill this gap in school outbreak studies, we parameterized an individual-based model that accounts for heterogeneous contact rates within and between classes and grades to a multischool outbreak data of influenza. We then simulated school outbreaks of respiratory infectious diseases of ongoing threat (i.e., COVID-19) and potential threat (i.e., pandemic influenza) under a variety of interventions (changing class structures, symptom screening, regular testing, cohorting, and responsive class closures). Our results suggest that interventions changing class structures (e.g., reduced class sizes) may not be effective in reducing the risk of major school outbreaks upon introduction of a case and that other precautionary measures (e.g., screening and isolation) need to be employed. Class-level closures in response to detection of a case were also suggested to be effective in reducing the size of an outbreak.


Assuntos
Surtos de Doenças , Pandemias , Infecções Respiratórias , Instituições Acadêmicas , COVID-19/prevenção & controle , COVID-19/transmissão , Criança , Simulação por Computador , Surtos de Doenças/prevenção & controle , Humanos , Influenza Humana/prevenção & controle , Influenza Humana/transmissão , Pandemias/prevenção & controle , Infecções Respiratórias/prevenção & controle , Infecções Respiratórias/transmissão
13.
Emerg Infect Dis ; 30(8): 1721-1723, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38914418

RESUMO

Examining the persistence of highly pathogenic avian influenza A(H5N1) from cattle and human influenza A(H1N1)pdm09 pandemic viruses in unpasteurized milk revealed that both remain infectious on milking equipment materials for several hours. Those findings highlight the risk for H5N1 virus transmission to humans from contaminated surfaces during the milking process.


Assuntos
Indústria de Laticínios , Vírus da Influenza A Subtipo H1N1 , Virus da Influenza A Subtipo H5N1 , Influenza Humana , Leite , Animais , Leite/virologia , Bovinos , Humanos , Indústria de Laticínios/instrumentação , Influenza Humana/transmissão , Influenza Humana/virologia , Infecções por Orthomyxoviridae/transmissão , Infecções por Orthomyxoviridae/virologia
14.
J Gen Virol ; 105(7)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38980150

RESUMO

Between 2013 and 2017, the A/Anhui/1/13-lineage (H7N9) low-pathogenicity avian influenza virus (LPAIV) was epizootic in chickens in China, causing mild disease, with 616 fatal human cases. Despite poultry vaccination, H7N9 has not been eradicated. Previously, we demonstrated increased pathogenesis in turkeys infected with H7N9, correlating with the emergence of the L217Q (L226Q H3 numbering) polymorphism in the haemagglutinin (HA) protein. A Q217-containing virus also arose and is now dominant in China following vaccination. We compared infection and transmission of this Q217-containing 'turkey-adapted' (ty-ad) isolate alongside the H7N9 (L217) wild-type (wt) virus in different poultry species and investigated the zoonotic potential in the ferret model. Both wt and ty-ad viruses demonstrated similar shedding and transmission in turkeys and chickens. However, the ty-ad virus was significantly more pathogenic than the wt virus in turkeys but not in chickens, causing 100 and 33% mortality in turkeys respectively. Expanded tissue tropism was seen for the ty-ad virus in turkeys but not in chickens, yet the viral cell receptor distribution was broadly similar in the visceral organs of both species. The ty-ad virus required exogenous trypsin for in vitro replication yet had increased replication in primary avian cells. Replication was comparable in mammalian cells, and the ty-ad virus replicated successfully in ferrets. The L217Q polymorphism also affected antigenicity. Therefore, H7N9 infection in turkeys can generate novel variants with increased risk through altered pathogenicity and potential HA antigenic escape. These findings emphasize the requirement for enhanced surveillance and understanding of A/Anhui/1/13-lineage viruses and their risk to different species.


Assuntos
Galinhas , Furões , Subtipo H7N9 do Vírus da Influenza A , Influenza Aviária , Perus , Animais , Perus/virologia , Influenza Aviária/virologia , Influenza Aviária/transmissão , Subtipo H7N9 do Vírus da Influenza A/genética , Subtipo H7N9 do Vírus da Influenza A/patogenicidade , Galinhas/virologia , Virulência , China/epidemiologia , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/transmissão , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Eliminação de Partículas Virais , Replicação Viral , Zoonoses/virologia , Influenza Humana/virologia , Influenza Humana/transmissão
15.
Curr Opin Infect Dis ; 37(5): 431-435, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-38975631

RESUMO

PURPOSE OF REVIEW: This review aims to discuss the current state of human infections with Avian Influenza A (H5) and (H9) viruses, to support awareness of the global epidemiology among clinicians and public health professionals interested in emerging respiratory infections. RECENT FINDINGS: Among increasing numbers of detections in avian species of Avian Influenza A(H5N1) clade 2.3.4.4b globally, reported human cases of severe infection have been rare.Enhanced surveillance of persons exposed to avian species infected with Influenza A (H5N1) clade 2.3.4.4b in different countries has identified small numbers of asymptomatic individuals with Avian Influenza A (H5N1) detected by PCR from the upper respiratory tract; some of these instances have been considered to represent contamination rather than infection.There have also been recent sporadic human cases of Avian Influenza A(H9N2) internationally, including in China and Cambodia. SUMMARY: Human infections with Avian Influenza A(H5) and (H9) viruses remain of interest as an emerging infection both to clinicians and public health professionals. While maintaining effective surveillance is essential, one health strategies to control infection in avian species will be key to mitigating these risks.


Assuntos
Virus da Influenza A Subtipo H5N1 , Influenza Aviária , Influenza Humana , Zoonoses , Humanos , Animais , Influenza Humana/epidemiologia , Influenza Humana/transmissão , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Influenza Aviária/epidemiologia , Influenza Aviária/transmissão , Influenza Aviária/virologia , Zoonoses/epidemiologia , Zoonoses/virologia , Zoonoses/transmissão , Aves/virologia , Vírus da Influenza A Subtipo H9N2 , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/virologia , Doenças Transmissíveis Emergentes/transmissão , Zoonoses Virais/transmissão , Zoonoses Virais/epidemiologia , Zoonoses Virais/virologia , Saúde Global
16.
J Med Virol ; 96(9): e29922, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39295292

RESUMO

This study retrospectively analyzed the genetic characteristics of influenza A H3N2 (A/H3N2) viruses circulating in New South Wales (NSW), the Australian state with the highest number of influenza cases in 2022, and explored the phylodynamics of A/H3N2 transmission within Australia during this period. Sequencing was performed on 217 archived specimens, and A/H3N2 evolution and spread within Australia were analyzed using phylogenetic and phylodynamic methods. Hemagglutinin genes of all analyzed NSW viruses belonged to subclade 3C.2a1b.2a.2 and clustered together with the 2022 vaccine strain. Complete genome analysis of NSW viruses revealed highly frequent interclade reassortments between subclades 3C.2a1b.2a.2 and 3C.2a1b.1a. The estimated earliest introduction time of the dominant subgroup 3C.2a1b.2a.2a.1 in Australia was February 22, 2022 (95% highest posterior density: December 19, 2021-March 13, 2022), following the easing of Australian travel restrictions, suggesting a possible international source. Phylogeographic analysis revealed that Victoria drove the transmission of A/H3N2 viruses across the country during this season, while NSW did not have a dominant role in viral dissemination to other regions. This study highlights the importance of continuous surveillance and genomic characterization of influenza viruses in the postpandemic era, which can inform public health decision-making and enable early detection of novel strains with pandemic potential.


Assuntos
COVID-19 , Vírus da Influenza A Subtipo H3N2 , Influenza Humana , Filogenia , Humanos , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/classificação , Vírus da Influenza A Subtipo H3N2/isolamento & purificação , Influenza Humana/epidemiologia , Influenza Humana/virologia , Influenza Humana/transmissão , Estudos Retrospectivos , COVID-19/epidemiologia , COVID-19/transmissão , COVID-19/virologia , COVID-19/prevenção & controle , Austrália/epidemiologia , New South Wales/epidemiologia , SARS-CoV-2/genética , SARS-CoV-2/classificação , Filogeografia , Estações do Ano , Genoma Viral/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus Reordenados/genética , Vírus Reordenados/classificação
17.
J Theor Biol ; 587: 111817, 2024 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-38599566

RESUMO

The recent global COVID-19 pandemic resulted in governments enacting non-pharmaceutical interventions (NPIs) targeted at reducing transmission of SARS-CoV-2. But the NPIs also affected the transmission of viruses causing non-target seasonal respiratory diseases, including influenza and respiratory syncytial virus (RSV). In many countries, the NPIs were found to reduce cases of such seasonal respiratory diseases, but there is also evidence that subsequent relaxation of NPIs led to outbreaks of these diseases that were larger than pre-pandemic ones, due to the accumulation of susceptible individuals prior to relaxation. Therefore, the net long-term effects of NPIs on the total disease burden of non-target diseases remain unclear. Knowledge of this is important for infectious disease management and maintenance of public health. In this study, we shed light on this issue for the simplified scenario of a set of NPIs that prevent or reduce transmission of a seasonal respiratory disease for about a year and are then removed, using mathematical analyses and numerical simulations of a suite of four epidemiological models with varying complexity and generality. The model parameters were estimated using empirical data pertaining to seasonal respiratory diseases and covered a wide range. Our results showed that NPIs reduced the total disease burden of a non-target seasonal respiratory disease in the long-term. Expressed as a percentage of population size, the reduction was greater for larger values of the basic reproduction number and the immunity loss rate, reflecting larger outbreaks and hence more infections averted by imposition of NPIs. Our study provides a foundation for exploring the effects of NPIs on total disease burden in more-complex scenarios.


Assuntos
COVID-19 , Modelos Epidemiológicos , SARS-CoV-2 , Humanos , COVID-19/epidemiologia , COVID-19/prevenção & controle , COVID-19/transmissão , Pandemias/prevenção & controle , Infecções por Vírus Respiratório Sincicial/epidemiologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Estações do Ano , Influenza Humana/epidemiologia , Influenza Humana/prevenção & controle , Influenza Humana/transmissão , Efeitos Psicossociais da Doença
18.
Cell ; 136(3): 402-10, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19203576

RESUMO

Both seasonal and pandemic influenza continue to challenge both scientists and clinicians. Drug-resistant H1N1 influenza viruses have dominated the 2009 flu season, and the H5N1 avian influenza virus continues to kill both people and poultry in Eurasia. Here, we discuss the pathogenesis and transmissibility of influenza viruses and we emphasize the need to find better predictors of both seasonal and potentially pandemic influenza.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/virologia , Animais , Aves , Reservatórios de Doenças , Farmacorresistência Viral , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H3N2/fisiologia , Virus da Influenza A Subtipo H5N1/fisiologia , Vírus da Influenza A/patogenicidade , Vacinas contra Influenza/imunologia , Influenza Aviária/virologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Influenza Humana/transmissão , Infecções por Orthomyxoviridae/virologia , Vigilância da População
20.
Bull Math Biol ; 86(6): 71, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38719993

RESUMO

Due to the complex interactions between multiple infectious diseases, the spreading of diseases in human bodies can vary when people are exposed to multiple sources of infection at the same time. Typically, there is heterogeneity in individuals' responses to diseases, and the transmission routes of different diseases also vary. Therefore, this paper proposes an SIS disease spreading model with individual heterogeneity and transmission route heterogeneity under the simultaneous action of two competitive infectious diseases. We derive the theoretical epidemic spreading threshold using quenched mean-field theory and perform numerical analysis under the Markovian method. Numerical results confirm the reliability of the theoretical threshold and show the inhibitory effect of the proportion of fully competitive individuals on epidemic spreading. The results also show that the diversity of disease transmission routes promotes disease spreading, and this effect gradually weakens when the epidemic spreading rate is high enough. Finally, we find a negative correlation between the theoretical spreading threshold and the average degree of the network. We demonstrate the practical application of the model by comparing simulation outputs to temporal trends of two competitive infectious diseases, COVID-19 and seasonal influenza in China.


Assuntos
COVID-19 , Simulação por Computador , Influenza Humana , Cadeias de Markov , Conceitos Matemáticos , Modelos Biológicos , SARS-CoV-2 , Humanos , COVID-19/transmissão , COVID-19/epidemiologia , COVID-19/prevenção & controle , Influenza Humana/epidemiologia , Influenza Humana/transmissão , China/epidemiologia , Número Básico de Reprodução/estatística & dados numéricos , Modelos Epidemiológicos , Pandemias/estatística & dados numéricos , Pandemias/prevenção & controle , Epidemias/estatística & dados numéricos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA