Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 17.737
Filtrar
Mais filtros

Coleções SMS-SP
Intervalo de ano de publicação
1.
Cell ; 181(1): 6, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32243796

RESUMO

Ervebo is the first licensed vaccine for prevention of Ebola virus disease. The vaccine, originally developed by the Public Health Agency of Canada, is delivered in a single 1 mL dose and has been delivered to >200,000 people in an ongoing 2018-2020 outbreak of disease. To view this Bench to Bedside, open or download the PDF.


Assuntos
Anticorpos Antivirais/imunologia , Surtos de Doenças/prevenção & controle , Vacinas contra Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Vacinação em Massa , Aprovação de Drogas , Ebolavirus , Humanos , Proteínas do Envelope Viral/imunologia
2.
Nature ; 622(7984): 810-817, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37853121

RESUMO

Highly pathogenic avian influenza (HPAI) H5N1 activity has intensified globally since 2021, increasingly causing mass mortality in wild birds and poultry and incidental infections in mammals1-3. However, the ecological and virological properties that underscore future mitigation strategies still remain unclear. Using epidemiological, spatial and genomic approaches, we demonstrate changes in the origins of resurgent HPAI H5 and reveal significant shifts in virus ecology and evolution. Outbreak data show key resurgent events in 2016-2017 and 2020-2021, contributing to the emergence and panzootic spread of H5N1 in 2021-2022. Genomic analysis reveals that the 2016-2017 epizootics originated in Asia, where HPAI H5 reservoirs are endemic. In 2020-2021, 2.3.4.4b H5N8 viruses emerged in African poultry, featuring mutations altering HA structure and receptor binding. In 2021-2022, a new H5N1 virus evolved through reassortment in wild birds in Europe, undergoing further reassortment with low-pathogenic avian influenza in wild and domestic birds during global dissemination. These results highlight a shift in the HPAI H5 epicentre beyond Asia and indicate that increasing persistence of HPAI H5 in wild birds is facilitating geographic and host range expansion, accelerating dispersion velocity and increasing reassortment potential. As earlier outbreaks of H5N1 and H5N8 were caused by more stable genomic constellations, these recent changes reflect adaptation across the domestic-bird-wild-bird interface. Elimination strategies in domestic birds therefore remain a high priority to limit future epizootics.


Assuntos
Aves , Surtos de Doenças , Virus da Influenza A Subtipo H5N1 , Influenza Aviária , Internacionalidade , Animais , África/epidemiologia , Animais Selvagens/virologia , Ásia/epidemiologia , Aves/virologia , Surtos de Doenças/prevenção & controle , Surtos de Doenças/estatística & dados numéricos , Surtos de Doenças/veterinária , Europa (Continente)/epidemiologia , Evolução Molecular , Especificidade de Hospedeiro , Virus da Influenza A Subtipo H5N1/classificação , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Virus da Influenza A Subtipo H5N1/patogenicidade , Vírus da Influenza A Subtipo H5N8/genética , Vírus da Influenza A Subtipo H5N8/isolamento & purificação , Influenza Aviária/epidemiologia , Influenza Aviária/mortalidade , Influenza Aviária/transmissão , Influenza Aviária/virologia , Mamíferos/virologia , Mutação , Filogenia , Aves Domésticas/virologia
3.
N Engl J Med ; 388(12): 1101-1110, 2023 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-36947467

RESUMO

BACKGROUND: Despite widespread adoption of surveillance testing for coronavirus disease 2019 (Covid-19) among staff members in skilled nursing facilities, evidence is limited regarding its relationship with outcomes among facility residents. METHODS: Using data obtained from 2020 to 2022, we performed a retrospective cohort study of testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) among staff members in 13,424 skilled nursing facilities during three pandemic periods: before vaccine approval, before the B.1.1.529 (omicron) variant wave, and during the omicron wave. We assessed staff testing volumes during weeks without Covid-19 cases relative to other skilled nursing facilities in the same county, along with Covid-19 cases and deaths among residents during potential outbreaks (defined as the occurrence of a case after 2 weeks with no cases). We reported adjusted differences in outcomes between high-testing facilities (90th percentile of test volume) and low-testing facilities (10th percentile). The two primary outcomes were the weekly cumulative number of Covid-19 cases and related deaths among residents during potential outbreaks. RESULTS: During the overall study period, 519.7 cases of Covid-19 per 100 potential outbreaks were reported among residents of high-testing facilities as compared with 591.2 cases among residents of low-testing facilities (adjusted difference, -71.5; 95% confidence interval [CI], -91.3 to -51.6). During the same period, 42.7 deaths per 100 potential outbreaks occurred in high-testing facilities as compared with 49.8 deaths in low-testing facilities (adjusted difference, -7.1; 95% CI, -11.0 to -3.2). Before vaccine availability, high- and low-testing facilities had 759.9 cases and 1060.2 cases, respectively, per 100 potential outbreaks (adjusted difference, -300.3; 95% CI, -377.1 to -223.5), along with 125.2 and 166.8 deaths (adjusted difference, -41.6; 95% CI, -57.8 to -25.5). Before the omicron wave, the numbers of cases and deaths were similar in high- and low-testing facilities; during the omicron wave, high-testing facilities had fewer cases among residents, but deaths were similar in the two groups. CONCLUSIONS: Greater surveillance testing of staff members at skilled nursing facilities was associated with clinically meaningful reductions in Covid-19 cases and deaths among residents, particularly before vaccine availability.


Assuntos
COVID-19 , Surtos de Doenças , Pessoal de Saúde , Vigilância da População , Instituições de Cuidados Especializados de Enfermagem , Humanos , COVID-19/diagnóstico , COVID-19/epidemiologia , COVID-19/mortalidade , COVID-19/prevenção & controle , Surtos de Doenças/prevenção & controle , Surtos de Doenças/estatística & dados numéricos , Estudos Retrospectivos , SARS-CoV-2 , Instituições de Cuidados Especializados de Enfermagem/normas , Instituições de Cuidados Especializados de Enfermagem/estatística & dados numéricos , Pessoal de Saúde/normas , Pessoal de Saúde/estatística & dados numéricos , Vigilância da População/métodos , Pacientes/estatística & dados numéricos , Pandemias/prevenção & controle , Pandemias/estatística & dados numéricos
4.
Nature ; 584(7821): 425-429, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32604404

RESUMO

On 21 February 2020, a resident of the municipality of Vo', a small town near Padua (Italy), died of pneumonia due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection1. This was the first coronavirus disease 19 (COVID-19)-related death detected in Italy since the detection of SARS-CoV-2 in the Chinese city of Wuhan, Hubei province2. In response, the regional authorities imposed the lockdown of the whole municipality for 14 days3. Here we collected information on the demography, clinical presentation, hospitalization, contact network and the presence of SARS-CoV-2 infection in nasopharyngeal swabs for 85.9% and 71.5% of the population of Vo' at two consecutive time points. From the first survey, which was conducted around the time the town lockdown started, we found a prevalence of infection of 2.6% (95% confidence interval (CI): 2.1-3.3%). From the second survey, which was conducted at the end of the lockdown, we found a prevalence of 1.2% (95% CI: 0.8-1.8%). Notably, 42.5% (95% CI: 31.5-54.6%) of the confirmed SARS-CoV-2 infections detected across the two surveys were asymptomatic (that is, did not have symptoms at the time of swab testing and did not develop symptoms afterwards). The mean serial interval was 7.2 days (95% CI: 5.9-9.6). We found no statistically significant difference in the viral load of symptomatic versus asymptomatic infections (P = 0.62 and 0.74 for E and RdRp genes, respectively, exact Wilcoxon-Mann-Whitney test). This study sheds light on the frequency of asymptomatic SARS-CoV-2 infection, their infectivity (as measured by the viral load) and provides insights into its transmission dynamics and the efficacy of the implemented control measures.


Assuntos
Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/prevenção & controle , Surtos de Doenças/prevenção & controle , Pandemias/prevenção & controle , Pneumonia Viral/epidemiologia , Pneumonia Viral/prevenção & controle , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Infecções Assintomáticas/epidemiologia , Betacoronavirus/enzimologia , Betacoronavirus/genética , Betacoronavirus/isolamento & purificação , COVID-19 , Criança , Pré-Escolar , Proteínas do Envelope de Coronavírus , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/virologia , RNA-Polimerase RNA-Dependente de Coronavírus , Surtos de Doenças/estatística & dados numéricos , Feminino , Humanos , Lactente , Recém-Nascido , Itália/epidemiologia , Masculino , Pessoa de Meia-Idade , Pneumonia Viral/transmissão , Pneumonia Viral/virologia , Prevalência , RNA Polimerase Dependente de RNA/genética , SARS-CoV-2 , Proteínas do Envelope Viral/genética , Carga Viral , Proteínas não Estruturais Virais/genética , Adulto Jovem
5.
Proc Natl Acad Sci U S A ; 120(48): e2305227120, 2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-37983514

RESUMO

Disease surveillance systems provide early warnings of disease outbreaks before they become public health emergencies. However, pandemics containment would be challenging due to the complex immunity landscape created by multiple variants. Genomic surveillance is critical for detecting novel variants with diverse characteristics and importation/emergence times. Yet, a systematic study incorporating genomic monitoring, situation assessment, and intervention strategies is lacking in the literature. We formulate an integrated computational modeling framework to study a realistic course of action based on sequencing, analysis, and response. We study the effects of the second variant's importation time, its infectiousness advantage and, its cross-infection on the novel variant's detection time, and the resulting intervention scenarios to contain epidemics driven by two-variants dynamics. Our results illustrate the limitation in the intervention's effectiveness due to the variants' competing dynamics and provide the following insights: i) There is a set of importation times that yields the worst detection time for the second variant, which depends on the first variant's basic reproductive number; ii) When the second variant is imported relatively early with respect to the first variant, the cross-infection level does not impact the detection time of the second variant. We found that depending on the target metric, the best outcomes are attained under different interventions' regimes. Our results emphasize the importance of sustained enforcement of Non-Pharmaceutical Interventions on preventing epidemic resurgence due to importation/emergence of novel variants. We also discuss how our methods can be used to study when a novel variant emerges within a population.


Assuntos
COVID-19 , Pandemias , Humanos , Pandemias/prevenção & controle , Saúde Pública , Surtos de Doenças/prevenção & controle , Genômica
6.
Proc Natl Acad Sci U S A ; 120(41): e2305451120, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37788317

RESUMO

In the era of living with COVID-19, the risk of localised SARS-CoV-2 outbreaks remains. Here, we develop a multiscale modelling framework for estimating the local outbreak risk for a viral disease (the probability that a major outbreak results from a single case introduced into the population), accounting for within-host viral dynamics. Compared to population-level models previously used to estimate outbreak risks, our approach enables more detailed analysis of how the risk can be mitigated through pre-emptive interventions such as antigen testing. Considering SARS-CoV-2 as a case study, we quantify the within-host dynamics using data from individuals with omicron variant infections. We demonstrate that regular antigen testing reduces, but may not eliminate, the outbreak risk, depending on characteristics of local transmission. In our baseline analysis, daily antigen testing reduces the outbreak risk by 45% compared to a scenario without antigen testing. Additionally, we show that accounting for heterogeneity in within-host dynamics between individuals affects outbreak risk estimates and assessments of the impact of antigen testing. Our results therefore highlight important factors to consider when using multiscale models to design pre-emptive interventions against SARS-CoV-2 and other viruses.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/epidemiologia , Surtos de Doenças/prevenção & controle , Probabilidade
7.
Proc Natl Acad Sci U S A ; 120(18): e2207537120, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-37098064

RESUMO

Policymakers must make management decisions despite incomplete knowledge and conflicting model projections. Little guidance exists for the rapid, representative, and unbiased collection of policy-relevant scientific input from independent modeling teams. Integrating approaches from decision analysis, expert judgment, and model aggregation, we convened multiple modeling teams to evaluate COVID-19 reopening strategies for a mid-sized United States county early in the pandemic. Projections from seventeen distinct models were inconsistent in magnitude but highly consistent in ranking interventions. The 6-mo-ahead aggregate projections were well in line with observed outbreaks in mid-sized US counties. The aggregate results showed that up to half the population could be infected with full workplace reopening, while workplace restrictions reduced median cumulative infections by 82%. Rankings of interventions were consistent across public health objectives, but there was a strong trade-off between public health outcomes and duration of workplace closures, and no win-win intermediate reopening strategies were identified. Between-model variation was high; the aggregate results thus provide valuable risk quantification for decision making. This approach can be applied to the evaluation of management interventions in any setting where models are used to inform decision making. This case study demonstrated the utility of our approach and was one of several multimodel efforts that laid the groundwork for the COVID-19 Scenario Modeling Hub, which has provided multiple rounds of real-time scenario projections for situational awareness and decision making to the Centers for Disease Control and Prevention since December 2020.


Assuntos
COVID-19 , Humanos , COVID-19/epidemiologia , COVID-19/prevenção & controle , Incerteza , Surtos de Doenças/prevenção & controle , Saúde Pública , Pandemias/prevenção & controle
8.
Lancet ; 403(10427): 667-682, 2024 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-38280388

RESUMO

Dengue, caused by four closely related viruses, is a growing global public health concern, with outbreaks capable of overwhelming health-care systems and disrupting economies. Dengue is endemic in more than 100 countries across tropical and subtropical regions worldwide, and the expanding range of the mosquito vector, affected in part by climate change, increases risk in new areas such as Spain, Portugal, and the southern USA, while emerging evidence points to silent epidemics in Africa. Substantial advances in our understanding of the virus, immune responses, and disease progression have been made within the past decade. Novel interventions have emerged, including partially effective vaccines and innovative mosquito control strategies, although a reliable immune correlate of protection remains a challenge for the assessment of vaccines. These developments mark the beginning of a new era in dengue prevention and control, offering promise in addressing this pressing global health issue.


Assuntos
Aedes , Vírus da Dengue , Dengue , Vacinas , Animais , Humanos , Dengue/epidemiologia , Dengue/prevenção & controle , Surtos de Doenças/prevenção & controle , Saúde Pública
9.
J Virol ; 98(2): e0168323, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38226809

RESUMO

Emerging and endemic zoonotic diseases continue to threaten human and animal health, our social fabric, and the global economy. Zoonoses frequently emerge from congregate interfaces where multiple animal species and humans coexist, including farms and markets. Traditional food markets are widespread across the globe and create an interface where domestic and wild animals interact among themselves and with humans, increasing the risk of pathogen spillover. Despite decades of evidence linking markets to disease outbreaks across the world, there remains a striking lack of pathogen surveillance programs that can relay timely, cost-effective, and actionable information to decision-makers to protect human and animal health. However, the strategic incorporation of environmental surveillance systems in markets coupled with novel pathogen detection strategies can create an early warning system capable of alerting us to the risk of outbreaks before they happen. Here, we explore the concept of "smart" markets that utilize continuous surveillance systems to monitor the emergence of zoonotic pathogens with spillover potential.IMPORTANCEFast detection and rapid intervention are crucial to mitigate risks of pathogen emergence, spillover and spread-every second counts. However, comprehensive, active, longitudinal surveillance systems at high-risk interfaces that provide real-time data for action remain lacking. This paper proposes "smart market" systems harnessing cutting-edge tools and a range of sampling techniques, including wastewater and air collection, multiplex assays, and metagenomic sequencing. Coupled with robust response pathways, these systems could better enable Early Warning and bolster prevention efforts.


Assuntos
Doenças Transmissíveis Emergentes , Monitoramento Epidemiológico , Animais , Humanos , Animais Selvagens , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/prevenção & controle , Doenças Transmissíveis Emergentes/veterinária , Surtos de Doenças/prevenção & controle , Zoonoses/epidemiologia , Zoonoses/prevenção & controle
10.
PLoS Comput Biol ; 20(8): e1012345, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39116182

RESUMO

Human behaviors have non-negligible impacts on spread of contagious disease. For instance, large-scale gathering and high mobility of population could lead to accelerated disease transmission, while public behavioral changes in response to pandemics may effectively reduce contacts and suppress the peak of the outbreak. In order to understand how spatial characteristics like population mobility and clustering interplay with epidemic outbreaks, we formulate a stochastic-statistical environment-epidemic dynamic system (SEEDS) via an agent-based biased random walk model on a two-dimensional lattice. The "popularity" and "awareness" variables are taken into consideration to capture human natural and preventive behavioral factors, which are assumed to guide and bias agent movement in a combined way. It is found that the presence of the spatial heterogeneity, like social influence locality and spatial clustering induced by self-aggregation, potentially suppresses the contacts between agents and consequently flats the epidemic curve. Surprisedly, disease responses might not necessarily reduce the susceptibility of informed individuals and even aggravate disease outbreak if each individual responds independently upon their awareness. The disease control is achieved effectively only if there are coordinated public-health interventions and public compliance to these measures. Therefore, our model may be useful for quantitative evaluations of a variety of public-health policies.


Assuntos
Doenças Transmissíveis , Humanos , Doenças Transmissíveis/epidemiologia , Doenças Transmissíveis/transmissão , Biologia Computacional/métodos , Surtos de Doenças/estatística & dados numéricos , Surtos de Doenças/prevenção & controle , Processos Estocásticos
11.
PLoS Comput Biol ; 20(8): e1012358, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39146377

RESUMO

Reducing spillover of zoonotic pathogens is an appealing approach to preventing human disease and minimizing the risk of future epidemics and pandemics. Although the immediate human health benefit of reducing spillover is clear, over time, spillover reduction could lead to counterintuitive negative consequences for human health. Here, we use mathematical models and computer simulations to explore the conditions under which unanticipated consequences of spillover reduction can occur in systems where the severity of disease increases with age at infection. Our results demonstrate that, because the average age at infection increases as spillover is reduced, programs that reduce spillover can actually increase population-level disease burden if the clinical severity of infection increases sufficiently rapidly with age. If, however, immunity wanes over time and reinfection is possible, our results reveal that negative health impacts of spillover reduction become substantially less likely. When our model is parameterized using published data on Lassa virus in West Africa, it predicts that negative health outcomes are possible, but likely to be restricted to a small subset of populations where spillover is unusually intense. Together, our results suggest that adverse consequences of spillover reduction programs are unlikely but that the public health gains observed immediately after spillover reduction may fade over time as the age structure of immunity gradually re-equilibrates to a reduced force of infection.


Assuntos
Simulação por Computador , Zoonoses , Humanos , Animais , Zoonoses/transmissão , Zoonoses/epidemiologia , Zoonoses/prevenção & controle , Zoonoses/virologia , Biologia Computacional , Saúde Pública , Febre Lassa/epidemiologia , Febre Lassa/prevenção & controle , Febre Lassa/transmissão , Surtos de Doenças/prevenção & controle , Surtos de Doenças/estatística & dados numéricos , Medição de Risco , África Ocidental/epidemiologia
12.
PLoS Comput Biol ; 20(7): e1012010, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39024382

RESUMO

During an infectious disease outbreak, public health policy makers are tasked with strategically implementing interventions whilst balancing competing objectives. To provide a quantitative framework that can be used to guide these decisions, it is helpful to devise a clear and specific objective function that can be evaluated to determine the optimal outbreak response. In this study, we have developed a mathematical modelling framework representing outbreaks of a novel emerging pathogen for which non-pharmaceutical interventions (NPIs) are imposed or removed based on thresholds for hospital occupancy. These thresholds are set at different levels to define four unique strategies for disease control. We illustrate that the optimal intervention strategy is contingent on the choice of objective function. Specifically, the optimal strategy depends on the extent to which policy makers prioritise reducing health costs due to infection over the costs associated with maintaining interventions. Motivated by the scenario early in the COVID-19 pandemic, we incorporate the development of a vaccine into our modelling framework and demonstrate that a policy maker's belief about when a vaccine will become available in future, and its eventual coverage (and/or effectiveness), affects the optimal strategy to adopt early in the outbreak. Furthermore, we show how uncertainty in these quantities can be accounted for when deciding which interventions to introduce. This research highlights the benefits of policy makers being explicit about the precise objectives of introducing interventions.


Assuntos
COVID-19 , Análise Custo-Benefício , Humanos , COVID-19/prevenção & controle , COVID-19/epidemiologia , Incerteza , SARS-CoV-2 , Vacinas contra COVID-19/economia , Pandemias/prevenção & controle , Quarentena , Biologia Computacional , Surtos de Doenças/prevenção & controle , Modelos Teóricos , Epidemias/prevenção & controle
13.
Rev Med Virol ; 34(4): e2559, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38886173

RESUMO

The World Organization for Animal Health defines Avian Influenza Virus as a highly infectious disease caused by diverse subtypes that continue to evolve rapidly, impacting poultry species, pet birds, wild birds, non-human mammals, and occasionally humans. The effects of Avian influenza viruses have been recognised as a precursor for serious health concerns among affected birds, poultry, and human populations in the Middle East. Furthermore, low and high pathogenic avian influenza viruses lead to respiratory illness with varying severity, depending on the virus subtype (e.g., H5, H7, H9, etc.). Possible future outbreaks and endemics of newly emerging subtypes are expected to occur, as many studies have reported the emergence of novel mutations and viral subtypes. However, proper surveillance programs and biosecurity applications should be developed, and countries with incapacitated defences against such outbreaks should be encouraged to undergo complete reinstation and reinforcement in their health and research sectors. Public education regarding biosafety and virus prevention is necessary to ensure minimal spread of avian influenza endemic.


Assuntos
Aves , Vírus da Influenza A , Influenza Aviária , Influenza Humana , Animais , Influenza Aviária/epidemiologia , Influenza Aviária/virologia , Influenza Aviária/prevenção & controle , Influenza Aviária/transmissão , Humanos , Influenza Humana/prevenção & controle , Influenza Humana/epidemiologia , Influenza Humana/virologia , Região do Mediterrâneo/epidemiologia , Aves/virologia , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Vírus da Influenza A/patogenicidade , Surtos de Doenças/prevenção & controle , Surtos de Doenças/veterinária
14.
Rev Med Virol ; 34(3): e2541, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38743385

RESUMO

As the mankind counters the ongoing COVID-19 pandemic by the novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), it simultaneously witnesses the emergence of mpox virus (MPXV) that signals at global spread and could potentially lead to another pandemic. Although MPXV has existed for more than 50 years now with most of the human cases being reported from the endemic West and Central African regions, the disease is recently being reported in non-endemic regions too that affect more than 50 countries. Controlling the spread of MPXV is important due to its potential danger of a global spread, causing severe morbidity and mortality. The article highlights the transmission dynamics, zoonosis potential, complication and mitigation strategies for MPXV infection, and concludes with suggested 'one health' approach for better management, control and prevention. Bibliometric analyses of the data extend the understanding and provide leads on the research trends, the global spread, and the need to revamp the critical research and healthcare interventions. Globally published mpox-related literature does not align well with endemic areas/regions of occurrence which should ideally have been the scenario. Such demographic and geographic gaps between the location of the research work and the endemic epicentres of the disease need to be bridged for greater and effective translation of the research outputs to pubic healthcare systems, it is suggested.


Assuntos
Bibliometria , Humanos , Surtos de Doenças/prevenção & controle , Animais , Mpox/epidemiologia , Mpox/transmissão , Mpox/prevenção & controle , Mpox/virologia , COVID-19/transmissão , COVID-19/epidemiologia , COVID-19/prevenção & controle , COVID-19/virologia , SARS-CoV-2 , Zoonoses/epidemiologia , Zoonoses/virologia , Zoonoses/transmissão , Zoonoses/prevenção & controle , Pandemias/prevenção & controle
17.
Nature ; 566(7745): 467-474, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30814711

RESUMO

Mobile health, or 'mHealth', is the application of mobile devices, their components and related technologies to healthcare. It is already improving patients' access to treatment and advice. Now, in combination with internet-connected diagnostic devices, it offers novel ways to diagnose, track and control infectious diseases and to improve the efficiency of the health system. Here we examine the promise of these technologies and discuss the challenges in realizing their potential to increase patients' access to testing, aid in their treatment and improve the capability of public health authorities to monitor outbreaks, implement response strategies and assess the impact of interventions across the world.


Assuntos
Doenças Transmissíveis/diagnóstico , Doenças Transmissíveis/terapia , Telemedicina/métodos , Telemedicina/organização & administração , Controle de Doenças Transmissíveis/métodos , Controle de Doenças Transmissíveis/organização & administração , Doenças Transmissíveis/epidemiologia , Doenças Transmissíveis/transmissão , Busca de Comunicante , Análise de Dados , Surtos de Doenças/prevenção & controle , Surtos de Doenças/estatística & dados numéricos , Humanos , Sistemas Automatizados de Assistência Junto ao Leito , Saúde Pública/métodos , Saúde Pública/tendências , Smartphone , Telemedicina/tendências
20.
Proc Natl Acad Sci U S A ; 119(23): e2121335119, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35639694

RESUMO

Many pathogenic viruses are endemic among human populations and can cause a broad variety of diseases, some potentially leading to devastating pandemics. How virus populations maintain diversity and what selective pressures drive population turnover is not thoroughly understood. We conducted a large-scale phylodynamic analysis of 27 human pathogenic RNA viruses spanning diverse life history traits, in search of unifying trends that shape virus evolution. For most virus species, we identify multiple, cocirculating lineages with low turnover rates. These lineages appear to be largely noncompeting and likely occupy semiindependent epidemiological niches that are not regionally or seasonally defined. Typically, intralineage mutational signatures are similar to interlineage signatures. The principal exception are members of the family Picornaviridae, for which mutations in capsid protein genes are primarily lineage defining. Interlineage turnover is slower than expected under a neutral model, whereas intralineage turnover is faster than the neutral expectation, further supporting the existence of independent niches. The persistence of virus lineages appears to stem from limited outbreaks within small communities, so that only a small fraction of the global susceptible population is infected at any time. As disparate communities become increasingly connected through globalization, interaction and competition between lineages might increase as well, which could result in changing selective pressures and increased diversification and/or pathogenicity. Thus, in addition to zoonotic events, ongoing surveillance of familiar, endemic viruses appears to merit global attention with respect to the prevention or mitigation of future pandemics.


Assuntos
Vírus de RNA , RNA , Viroses , Surtos de Doenças/prevenção & controle , Saúde Global , Humanos , Internacionalidade , Pandemias , Vírus de RNA/genética , Vírus de RNA/patogenicidade , Estações do Ano , Viroses/epidemiologia , Viroses/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA