Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
Sci Rep ; 14(1): 17532, 2024 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-39080433

RESUMEN

In the last two decades, Nipah virus (NiV) has emerged as a significant paramyxovirus transmitted by bats, causing severe respiratory illness and encephalitis in humans. NiV has been included in the World Health Organization's Blueprint list of priority pathogens due its potential for human-to-human transmission and zoonotic characteristics. In this paper, a mathematical model is formulated to analyze the dynamics and optimal control of NiV. In formulation of the model we consider two modes of transmission: human-to-human and food-borne. Further, the impact of contact with an infected corpse as a potential route for virus transmission is also consider in the model. The analysis identifies the model with constant controls has three equilibrium states: the NiV-free equilibrium, the infected flying foxes-free equilibrium, and the NiV-endemic equilibrium state. Furthermore, a theoretical analysis is conducted to presents the stability of the model equilibria. The model fitting to the reported cases in Bangladesh from 2001 to 2015, and the estimation of parameters are performed using the standard least squares technique. Sensitivity analysis of the model-embedded parameters is provided to set the optimal time-dependent controls for the disease eradication. The necessary optimality conditions are derived using Pontryagin's maximum principle. Finally, numerical simulation is performed to determine the most effective strategy for disease eradication and to confirm the theoretical results.


Asunto(s)
Infecciones por Henipavirus , Virus Nipah , Vacunación , Infecciones por Henipavirus/prevención & control , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/transmisión , Humanos , Animales , Quirópteros/virología , Modelos Teóricos , Bangladesh/epidemiología
2.
Lancet Planet Health ; 8(7): e463-e475, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38969474

RESUMEN

BACKGROUND: Nipah virus is a zoonotic paramyxovirus responsible for disease outbreaks with high fatality rates in south and southeast Asia. However, knowledge of the potential geographical extent and risk patterns of the virus is poor. We aimed to establish an integrated spatiotemporal and phylogenetic database of Nipah virus infections in humans and animals across south and southeast Asia. METHODS: In this geospatial modelling analysis, we developed an integrated database containing information on the distribution of Nipah virus infections in humans and animals from 1998 to 2021. We conducted phylodynamic analysis to examine the evolution and migration pathways of the virus and meta-analyses to estimate the adjusted case-fatality rate. We used two boosted regression tree models to identify the potential ecological drivers of Nipah virus occurrences in spillover events and endemic areas, and mapped potential risk areas for Nipah virus endemicity. FINDINGS: 749 people and eight bat species across nine countries were documented as being infected with Nipah virus. On the basis of 66 complete genomes of the virus, we identified two clades-the Bangladesh clade and the Malaysia clade-with the time of the most recent common ancestor estimated to be 1863. Adjusted case-fatality rates varied widely between countries and were higher for the Bangladesh clade than for the Malaysia clade. Multivariable meta-regression analysis revealed significant relationships between case-fatality rate estimates and viral clade (p=0·0021), source country (p=0·016), proportion of male patients (p=0·036), and travel time to health-care facilities (p=0·036). Temperature-related bioclimate variables and the probability of occurrence of Pteropus medius were important contributors to both the spillover and the endemic infection models. INTERPRETATION: The suitable niches for Nipah virus are more extensive than previously reported. Future surveillance efforts should focus on high-risk areas informed by updated projections. Specifically, intensifying zoonotic surveillance efforts, enhancing laboratory testing capacity, and implementing public health education in projected high-risk areas where no human cases have been reported to date will be crucial. Additionally, strengthening wildlife surveillance and investigating potential modes of transmission in regions with documented human cases is needed. FUNDING: The Key Research and Development Program of China.


Asunto(s)
Infecciones por Henipavirus , Virus Nipah , Virus Nipah/fisiología , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/transmisión , Humanos , Animales , Quirópteros/virología , Asia Sudoriental/epidemiología , Filogenia , Zoonosis/epidemiología , Zoonosis/virología
3.
Comp Immunol Microbiol Infect Dis ; 109: 102183, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38640700

RESUMEN

Henipavirus (HNV) is well known for two zoonotic viruses in the genus, Hendra virus (HeV) and Nipah virus (NiV), which pose serious threat to human and animal health. In August 2022, a third zoonotic virus in the genus Henipavirus, Langya virus (LayV), was discovered in China. The emergence of HeV, NiV, and LayV highlights the persistent threat of HNV to human and animal health. In addition to the above three HNVs, new species within this genus are still being discovered. Although they have not yet caused a pandemic in humans or livestock, they still have the risk of spillover as a potential threat to the health of humans and animals. It's important to understand the infection and transmission of different HNV in animals for the prevention and control of current or future HNV epidemics. Therefore, this review mainly summarizes the animal origin, animal infection and transmission of HNV that have been found worldwide, and further analyzes and summarizes the rules of infection and transmission, so as to provide a reference for relevant scientific researchers. Furthermore, it can provide a direction for epidemic prevention and control, and animal surveillance to reduce the risk of the global pandemic of HNV.


Asunto(s)
Infecciones por Henipavirus , Henipavirus , Animales , Infecciones por Henipavirus/transmisión , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/veterinaria , Infecciones por Henipavirus/virología , Humanos , Zoonosis/transmisión , Zoonosis/virología , Zoonosis Virales/transmisión , Zoonosis Virales/virología , Virus Nipah/patogenicidad , Virus Hendra
4.
J Infect Dis ; 229(3): 733-742, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-37925626

RESUMEN

Nipah virus Bangladesh (NiVB) is a bat-borne zoonosis transmitted between people through the respiratory route. The risk posed by related henipaviruses, including Hendra virus (HeV) and Nipah virus Malaysia (NiVM), is less clear. We conducted a broad search of the literature encompassing both human infections and animal models to synthesize evidence about potential for person-to-person spread. More than 600 human infections have been reported in the literature, but information on viral shedding was only available for 40 case-patients. There is substantial evidence demonstrating person-to-person transmission of NiVB, and some evidence for NiVM. Less direct evidence is available about the risk for person-to-person transmission of HeV, but animals infected with HeV shed more virus in the respiratory tract than those infected with NiVM, suggesting potential for transmission. As the group of known henipaviruses continues to grow, shared protocols for conducting and reporting from human investigations and animal experiments are urgently needed.


Asunto(s)
Virus Hendra , Infecciones por Henipavirus , Virus Nipah , Animales , Humanos , Infecciones por Henipavirus/transmisión , Malasia , Zoonosis/transmisión
5.
Lancet Infect Dis ; 22(1): e13-e27, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34735799

RESUMEN

Henipaviruses, including Nipah virus, are regarded as pathogens of notable epidemic potential because of their high pathogenicity and the paucity of specific medical countermeasures to control infections in humans. We review the evidence of medical countermeasures against henipaviruses and project their cost in a post-COVID-19 era. Given the sporadic and unpredictable nature of henipavirus outbreaks, innovative strategies will be needed to circumvent the infeasibility of traditional phase 3 clinical trial regulatory pathways. Stronger partnerships with scientific institutions and regulatory authorities in low-income and middle-income countries can inform coordination of appropriate investments and development of strategies and normative guidelines for the deployment and equitable use of multiple medical countermeasures. Accessible measures should include global, regional, and endemic in-country stockpiles of reasonably priced small molecules, monoclonal antibodies, and vaccines as part of a combined collection of products that could help to control henipavirus outbreaks and prevent future pandemics.


Asunto(s)
Brotes de Enfermedades/prevención & control , Infecciones por Henipavirus/tratamiento farmacológico , Henipavirus/patogenicidad , Contramedidas Médicas , Salud Pública , Animales , COVID-19/prevención & control , Quirópteros/virología , Ensayos Clínicos Fase III como Asunto , Henipavirus/clasificación , Infecciones por Henipavirus/prevención & control , Infecciones por Henipavirus/transmisión , Humanos , Virus Nipah/patogenicidad , SARS-CoV-2/patogenicidad
6.
Viruses ; 13(7)2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34372504

RESUMEN

Pandemics are a consequence of a series of processes that span scales from viral biology at 10-9 m to global transmission at 106 m. The pathogen passes from one host species to another through a sequence of events that starts with an infected reservoir host and entails interspecific contact, innate immune responses, receptor protein structure within the potential host, and the global spread of the novel pathogen through the naive host population. Each event presents a potential barrier to the onward passage of the virus and should be characterized with an integrated transdisciplinary approach. Epidemic control is based on the prevention of exposure, infection, and disease. However, the ultimate pandemic prevention is prevention of the spillover event itself. Here, we focus on the potential for preventing the spillover of henipaviruses, a group of viruses derived from bats that frequently cross species barriers, incur high human mortality, and are transmitted among humans via stuttering chains. We outline the transdisciplinary approach needed to prevent the spillover process and, therefore, future pandemics.


Asunto(s)
Quirópteros/virología , Salud Global , Infecciones por Henipavirus/prevención & control , Henipavirus/patogenicidad , Pandemias/prevención & control , Virosis/prevención & control , Zoonosis/virología , Animales , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/transmisión , Especificidad del Huésped , Humanos , Inmunidad Innata , Virus Nipah/patogenicidad , Virosis/inmunología , Virosis/transmisión , Zoonosis/prevención & control , Zoonosis/transmisión
7.
Proc Natl Acad Sci U S A ; 117(46): 29190-29201, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33139552

RESUMEN

Nipah virus (NiV) is an emerging bat-borne zoonotic virus that causes near-annual outbreaks of fatal encephalitis in South Asia-one of the most populous regions on Earth. In Bangladesh, infection occurs when people drink date-palm sap contaminated with bat excreta. Outbreaks are sporadic, and the influence of viral dynamics in bats on their temporal and spatial distribution is poorly understood. We analyzed data on host ecology, molecular epidemiology, serological dynamics, and viral genetics to characterize spatiotemporal patterns of NiV dynamics in its wildlife reservoir, Pteropus medius bats, in Bangladesh. We found that NiV transmission occurred throughout the country and throughout the year. Model results indicated that local transmission dynamics were modulated by density-dependent transmission, acquired immunity that is lost over time, and recrudescence. Increased transmission followed multiyear periods of declining seroprevalence due to bat-population turnover and individual loss of humoral immunity. Individual bats had smaller host ranges than other Pteropus species (spp.), although movement data and the discovery of a Malaysia-clade NiV strain in eastern Bangladesh suggest connectivity with bats east of Bangladesh. These data suggest that discrete multiannual local epizootics in bat populations contribute to the sporadic nature of NiV outbreaks in South Asia. At the same time, the broad spatial and temporal extent of NiV transmission, including the recent outbreak in Kerala, India, highlights the continued risk of spillover to humans wherever they may interact with pteropid bats and the importance of limiting opportunities for spillover throughout Pteropus's range.


Asunto(s)
Quirópteros/virología , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/transmisión , Infecciones por Henipavirus/veterinaria , Infecciones por Henipavirus/virología , Virus Nipah/clasificación , Virus Nipah/genética , Animales , Asia , Bangladesh/epidemiología , Brotes de Enfermedades , Femenino , Especificidad del Huésped , Humanos , Inmunidad , Masculino , Modelos Biológicos , Epidemiología Molecular , Virus Nipah/inmunología , Filogenia , Zoonosis/epidemiología , Zoonosis/inmunología , Zoonosis/transmisión , Zoonosis/virología
8.
Bull World Health Organ ; 98(8): 539-547, 2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32773899

RESUMEN

OBJECTIVE: To better understand the potential risks of Nipah virus emergence in Cambodia by studying different components of the interface between humans and bats. METHODS: From 2012 to 2016, we conducted a study at two sites in Kandal and Battambang provinces where fruit bats (Pteropus lylei) roost. We combined research on: bat ecology (reproductive phenology, population dynamics and diet); human practices and perceptions (ethnographic research and a knowledge, attitude and practice study); and Nipah virus circulation in bat and human populations (virus monitoring in bat urine and anti-Nipah-virus antibody detection in human serum). FINDINGS: Our results confirmed circulation of Nipah virus in fruit bats (28 of 3930 urine samples positive by polymerase chain reaction testing). We identified clear potential routes for virus transmission to humans through local practices, including fruit consumed by bats and harvested by humans when Nipah virus is circulating, and palm juice production. Nevertheless, in the serological survey of 418 potentially exposed people, none of them were seropositive to Nipah virus. Differences in agricultural practices among the regions where Nipah virus has emerged may explain the situation in Cambodia and point to actions to limit the risks of virus transmission to humans. CONCLUSION: Human practices are key to understanding transmission risks associated with emerging infectious diseases. Social science disciplines such as anthropology need to be integrated in health programmes targeting emerging infectious diseases. As bats are hosts of major zoonotic pathogens, such integrated studies would likely also help to reduce the risk of emergence of other bat-borne diseases.


Asunto(s)
Quirópteros/virología , Infecciones por Henipavirus/psicología , Infecciones por Henipavirus/transmisión , Virus Nipah/aislamiento & purificación , Animales , Antropología Cultural , Anticuerpos Antivirales , Cambodia/epidemiología , Femenino , Frutas , Conocimientos, Actitudes y Práctica en Salud , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/orina , Humanos , Masculino , Virus Nipah/inmunología , Factores de Riesgo , Zoonosis/virología
9.
BMC Vet Res ; 16(1): 300, 2020 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-32838786

RESUMEN

BACKGROUND: Nipah virus (NiV) is a fatal zoonotic agent that was first identified amongst pig farmers in Malaysia in 1998, in an outbreak that resulted in 105 fatal human cases. That epidemic arose from a chain of infection, initiating from bats to pigs, and which then spilled over from pigs to humans. In Thailand, bat-pig-human communities can be observed across the country, particularly in the central plain. The present study therefore aimed to identify high-risk areas for potential NiV outbreaks and to model how the virus is likely to spread. Multi-criteria decision analysis (MCDA) and weighted linear combination (WLC) were employed to produce the NiV risk map. The map was then overlaid with the nationwide pig movement network to identify the index subdistricts in which NiV may emerge. Subsequently, susceptible-exposed-infectious-removed (SEIR) modeling was used to simulate NiV spread within each subdistrict, and network modeling was used to illustrate how the virus disperses across subdistricts. RESULTS: Based on the MCDA and pig movement data, 14 index subdistricts with a high-risk of NiV emergence were identified. We found in our infectious network modeling that the infected subdistricts clustered in, or close to the central plain, within a range of 171 km from the source subdistricts. However, the virus may travel as far as 528.5 km (R0 = 5). CONCLUSIONS: In conclusion, the risk of NiV dissemination through pig movement networks in Thailand is low but not negligible. The risk areas identified in our study can help the veterinary authority to allocate financial and human resources to where preventive strategies, such as pig farm regionalization, are required and to contain outbreaks in a timely fashion once they occur.


Asunto(s)
Infecciones por Henipavirus/veterinaria , Virus Nipah , Enfermedades de los Porcinos/epidemiología , Animales , Quirópteros/virología , Técnicas de Apoyo para la Decisión , Brotes de Enfermedades/prevención & control , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/transmisión , Humanos , Porcinos , Enfermedades de los Porcinos/virología , Tailandia/epidemiología , Transportes
10.
Infect Genet Evol ; 85: 104442, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32622923

RESUMEN

Little is known about the genetic features of Nipah virus (NiV) associated with virulence and transmission. Herein, phylogenetic and genetic analyses for all available NiV strains revealed sequence variations between the two genetic lineages of NiV with pathogenic differences, as well as among different strains within Bangladesh lineage. A total of 143 conserved amino acid differences, distributed among viral nucleocapsid (N), phosphoprotein (P), matrix protein (M), fusion protein (F) and glycoprotein (G), were revealed. Structural modeling revealed one key substitution (S3554N) in the viral G protein that might mediate a 12-amino-acid structural change from a loop into a ß sheet. Multiple key amino acids substitutions in viral G protein were observed, which may alter viral fitness and transmissibility from bats to humans.


Asunto(s)
Variación Genética , Infecciones por Henipavirus/transmisión , Infecciones por Henipavirus/virología , Virus Nipah/genética , Virus Nipah/patogenicidad , Filogenia , Proteínas Virales/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Bangladesh , Quirópteros/virología , Evolución Molecular , Humanos , Malasia , Virulencia
11.
J Infect Dis ; 221(Suppl 4): S363-S369, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32392322

RESUMEN

It is of uttermost importance that the global health community develops the surveillance capability to effectively monitor emerging zoonotic pathogens that constitute a major and evolving threat for human health. In this study, we propose a comprehensive framework to measure changes in (1) spillover risk, (2) interhuman transmission, and (3) morbidity/mortality associated with infections based on 6 epidemiological key indicators derived from routine surveillance. We demonstrate the indicators' value for the retrospective or real-time assessment of changes in transmission and epidemiological characteristics using data collected through a long-standing, systematic, hospital-based surveillance system for Nipah virus in Bangladesh. We show that although interhuman transmission and morbidity/mortality indicators were stable, the number and geographic extent of spillovers varied significantly over time. This combination of systematic surveillance and active tracking of transmission and epidemiological indicators should be applied to other high-risk emerging pathogens to prevent public health emergencies.


Asunto(s)
Enfermedades Transmisibles Emergentes/virología , Infecciones por Henipavirus/transmisión , Infecciones por Henipavirus/virología , Virus Nipah/aislamiento & purificación , Animales , Bangladesh/epidemiología , Análisis por Conglomerados , Infecciones por Henipavirus/epidemiología , Humanos , Modelos Biológicos , Factores de Riesgo , Zoonosis
12.
Viruses ; 12(4)2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32325930

RESUMEN

Viral outbreaks of varying frequencies and severities have caused panic and havoc across the globe throughout history. Influenza, small pox, measles, and yellow fever reverberated for centuries, causing huge burden for economies. The twenty-first century witnessed the most pathogenic and contagious virus outbreaks of zoonotic origin including severe acute respiratory syndrome coronavirus (SARS-CoV), Ebola virus, Middle East respiratory syndrome coronavirus (MERS-CoV) and Nipah virus. Nipah is considered one of the world's deadliest viruses with the heaviest mortality rates in some instances. It is known to cause encephalitis, with cases of acute respiratory distress turning fatal. Various factors contribute to the onset and spread of the virus. All through the infected zone, various strategies to tackle and enhance the surveillance and awareness with greater emphasis on personal hygiene has been formulated. This review discusses the recent outbreaks of Nipah virus in Malaysia, Bangladesh and India, the routes of transmission, prevention and control measures employed along with possible reasons behind the outbreaks, and the precautionary measures to be ensured by private-public undertakings to contain and ensure a lower incidence in the future.


Asunto(s)
Encefalitis Viral/epidemiología , Encefalitis Viral/transmisión , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/transmisión , Virus Nipah/clasificación , Animales , Bangladesh/epidemiología , Quirópteros/virología , Brotes de Enfermedades , Encefalitis Viral/prevención & control , Infecciones por Henipavirus/prevención & control , Humanos , India/epidemiología , Control de Infecciones , Malasia/epidemiología , Virus Nipah/genética , Proteínas Estructurales Virales/genética
13.
J Infect Dis ; 222(3): 438-442, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32115627

RESUMEN

Contact patterns play a key role in disease transmission, and variation in contacts during the course of illness can influence transmission, particularly when accompanied by changes in host infectiousness. We used surveys among 1642 contacts of 94 Nipah virus case patients in Bangladesh to determine how contact patterns (physical and with bodily fluids) changed as disease progressed in severity. The number of contacts increased with severity and, for case patients who died, peaked on the day of death. Given transmission has only been observed among fatal cases of Nipah virus infection, our findings suggest that changes in contact patterns during illness contribute to risk of infection.


Asunto(s)
Líquidos Corporales/virología , Trazado de Contacto/estadística & datos numéricos , Infecciones por Henipavirus/transmisión , Virus Nipah , Conducta Social , Adolescente , Adulto , Bangladesh/epidemiología , Progresión de la Enfermedad , Transmisión de Enfermedad Infecciosa/prevención & control , Femenino , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/prevención & control , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo , Adulto Joven
14.
Transbound Emerg Dis ; 67(1): 121-132, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31408582

RESUMEN

Since its first emergence in 1998 in Malaysia, Nipah virus (NiV) has become a great threat to domestic animals and humans. Sporadic outbreaks associated with human-to-human transmission caused hundreds of human fatalities. Here, we collected all available NiV sequences and combined phylogenetics, molecular selection, structural biology and receptor analysis to study the emergence and adaptive evolution of NiV. NiV can be divided into two main lineages including the Bangladesh and Malaysia lineages. We formly confirmed a significant association with geography which is probably the result of long-term evolution of NiV in local bat population. The two NiV lineages differ in many amino acids; one change in the fusion protein might be involved in its activation via binding to the G protein. We also identified adaptive and positively selected sites in many viral proteins. In the receptor-binding G protein, we found that sites 384, 386 and especially 498 of G protein might modulate receptor-binding affinity and thus contribute to the host jump from bats to humans via the adaption to bind the human ephrin-B2 receptor. We also found that site 1645 in the connector domain of L was positive selected and involved in adaptive evolution; this site might add methyl groups to the cap structure present at the 5'-end of the RNA and thus modulate its activity. This study provides insight to assist the design of early detection methods for NiV to assess its epidemic potential in humans.


Asunto(s)
Adaptación Biológica , Quirópteros/virología , Brotes de Enfermedades , Infecciones por Henipavirus/virología , Virus Nipah/genética , Polimorfismo Genético , Animales , Bangladesh/epidemiología , Evolución Biológica , Biología Computacional , Geografía , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/transmisión , Especificidad del Huésped , Humanos , Malasia/epidemiología , Modelos Moleculares , Virus Nipah/aislamiento & purificación , Virus Nipah/patogenicidad , Virus Nipah/fisiología , Filogenia , Proteínas Virales/genética
15.
Sci Rep ; 9(1): 11171, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31371748

RESUMEN

Nipah virus (NiV) has emerged as a highly lethal zoonotic paramyxovirus that is capable of causing a febrile encephalitis and/or respiratory disease in humans for which no vaccines or licensed treatments are currently available. There are two genetically and geographically distinct lineages of NiV: NiV-Malaysia (NiV-M), the strain that caused the initial outbreak in Malaysia, and NiV-Bangladesh (NiV-B), the strain that has been implicated in subsequent outbreaks in India and Bangladesh. NiV-B appears to be both more lethal and have a greater propensity for person-to-person transmission than NiV-M. Here we describe the generation and characterization of stable RNA polymerase II-driven infectious cDNA clones of NiV-M and NiV-B. In vitro, reverse genetics-derived NiV-M and NiV-B were indistinguishable from a wildtype isolate of NiV-M, and both viruses were pathogenic in the Syrian hamster model of NiV infection. We also describe recombinant NiV-M and NiV-B with enhanced green fluorescent protein (EGFP) inserted between the G and L genes that enable rapid and sensitive detection of NiV infection in vitro. This panel of molecular clones will enable studies to investigate the virologic determinants of henipavirus pathogenesis, including the pathogenic differences between NiV-M and NiV-B, and the high-throughput screening of candidate therapeutics.


Asunto(s)
Virus Nipah/genética , Animales , Bangladesh , Brotes de Enfermedades , Infecciones por Henipavirus/transmisión , Infecciones por Henipavirus/virología , Humanos , Malasia , Mesocricetus/virología , ARN Polimerasa II , Genética Inversa
16.
Philos Trans R Soc Lond B Biol Sci ; 374(1782): 20190016, 2019 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-31401955

RESUMEN

Dose is the nexus between exposure and all upstream processes that determine pathogen pressure, and is thereby an important element underlying disease dynamics. Understanding the relationship between dose and disease is particularly important in the context of spillover, where nonlinearities in the dose-response could determine the likelihood of transmission. There is a need to explore dose-response models for directly transmitted and zoonotic pathogens, and how these interactions integrate within-host factors to consider, for example, heterogeneity in host susceptibility and dose-dependent antagonism. Here, we review the dose-response literature and discuss the unique role dose-response models have to play in understanding and predicting spillover events. We present a re-analysis of dose-response experiments for two important zoonotic pathogens (Middle East respiratory syndrome coronavirus and Nipah virus), to exemplify potential difficulties in differentiating between appropriate models with small exposure experiment datasets. We also discuss the data requirements needed for robust selection between dose-response models. We then suggest how these processes could be modelled to gain more realistic predictions of zoonotic transmission outcomes and highlight the exciting opportunities that could arise with increased collaboration between the virology and epidemiology disciplines. This article is part of the theme issue 'Dynamic and integrative approaches to understanding pathogen spillover'.


Asunto(s)
Infecciones por Coronavirus/transmisión , Reservorios de Enfermedades/virología , Ambiente , Infecciones por Henipavirus/transmisión , Interacciones Huésped-Patógeno , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Virus Nipah/fisiología , Animales , Animales Salvajes , Humanos
17.
Philos Trans R Soc Lond B Biol Sci ; 374(1782): 20190224, 2019 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-31401958

RESUMEN

Disease emergence events, epidemics and pandemics all underscore the need to predict zoonotic pathogen spillover. Because cross-species transmission is inherently hierarchical, involving processes that occur at varying levels of biological organization, such predictive efforts can be complicated by the many scales and vastness of data potentially required for forecasting. A wide range of approaches are currently used to forecast spillover risk (e.g. macroecology, pathogen discovery, surveillance of human populations, among others), each of which is bound within particular phylogenetic, spatial and temporal scales of prediction. Here, we contextualize these diverse approaches within their forecasting goals and resulting scales of prediction to illustrate critical areas of conceptual and pragmatic overlap. Specifically, we focus on an ecological perspective to envision a research pipeline that connects these different scales of data and predictions from the aims of discovery to intervention. Pathogen discovery and predictions focused at the phylogenetic scale can first provide coarse and pattern-based guidance for which reservoirs, vectors and pathogens are likely to be involved in spillover, thereby narrowing surveillance targets and where such efforts should be conducted. Next, these predictions can be followed with ecologically driven spatio-temporal studies of reservoirs and vectors to quantify spatio-temporal fluctuations in infection and to mechanistically understand how pathogens circulate and are transmitted to humans. This approach can also help identify general regions and periods for which spillover is most likely. We illustrate this point by highlighting several case studies where long-term, ecologically focused studies (e.g. Lyme disease in the northeast USA, Hendra virus in eastern Australia, Plasmodium knowlesi in Southeast Asia) have facilitated predicting spillover in space and time and facilitated the design of possible intervention strategies. Such studies can in turn help narrow human surveillance efforts and help refine and improve future large-scale, phylogenetic predictions. We conclude by discussing how greater integration and exchange between data and predictions generated across these varying scales could ultimately help generate more actionable forecasts and interventions. This article is part of the theme issue 'Dynamic and integrative approaches to understanding pathogen spillover'.


Asunto(s)
Enfermedades Transmisibles Emergentes , Reservorios de Enfermedades , Infecciones por Henipavirus , Enfermedad de Lyme , Malaria , Zoonosis , Animales , Asia Sudoriental/epidemiología , Australia/epidemiología , Borrelia burgdorferi/fisiología , Enfermedades Transmisibles Emergentes/epidemiología , Enfermedades Transmisibles Emergentes/transmisión , Reservorios de Enfermedades/microbiología , Reservorios de Enfermedades/parasitología , Reservorios de Enfermedades/virología , Virus Hendra/fisiología , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/transmisión , Humanos , Enfermedad de Lyme/epidemiología , Enfermedad de Lyme/transmisión , Malaria/epidemiología , Malaria/transmisión , Plasmodium knowlesi/fisiología , Estados Unidos/epidemiología , Zoonosis/epidemiología , Zoonosis/transmisión
18.
Philos Trans R Soc Lond B Biol Sci ; 374(1782): 20190021, 2019 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-31401962

RESUMEN

Pathogen circulation among reservoir hosts is a precondition for zoonotic spillover. Unlike the acute, high morbidity infections typical in spillover hosts, infected reservoir hosts often exhibit low morbidity and mortality. Although it has been proposed that reservoir host infections may be persistent with recurrent episodes of shedding, direct evidence is often lacking. We construct a generalized SEIR (susceptible, exposed, infectious, recovered) framework encompassing 46 sub-models representing the full range of possible transitions among those four states of infection and immunity. We then use likelihood-based methods to fit these models to nine years of longitudinal data on henipavirus serology from a captive colony of Eidolon helvum bats in Ghana. We find that reinfection is necessary to explain observed dynamics; that acute infectious periods may be very short (hours to days); that immunity, if present, lasts about 1-2 years; and that recurring latent infection is likely. Although quantitative inference is sensitive to assumptions about serology, qualitative predictions are robust. Our novel approach helps clarify mechanisms of viral persistence and circulation in wild bats, including estimated ranges for key parameters such as the basic reproduction number and the duration of the infectious period. Our results inform how future field-based and experimental work could differentiate the processes of viral recurrence and reinfection in reservoir hosts. This article is part of the theme issue 'Dynamic and integrative approaches to understanding pathogen spillover'.


Asunto(s)
Quirópteros , Reservorios de Enfermedades/veterinaria , Infecciones por Henipavirus/veterinaria , Henipavirus/fisiología , Animales , Animales de Zoológico , Reservorios de Enfermedades/virología , Ghana/epidemiología , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/transmisión , Infecciones por Henipavirus/virología , Prevalencia , Estudios Seroepidemiológicos
19.
Infez Med ; 27(2): 117-127, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-31205033

RESUMEN

The Nipah virus was discovered twenty years ago, and there is considerable information available regarding the specificities surrounding this virus such as transmission, pathogenesis and genome. Belonging to the Henipavirus genus, this virus can cause fever, encephalitis and respiratory disorders. The first cases were reported in Malaysia and Singapore in 1998, when affected individuals presented with severe febrile encephalitis. Since then, much has been identified about this virus. These single-stranded RNA viruses gain entry into target cells via a process known as macropinocytosis. The viral genome is released into the cell cytoplasm via a cascade of processes that involves conformational changes in G and F proteins which allow for attachment of the viral membrane to the cell membrane. In addition to this, the natural reservoirs of this virus have been identified to be fruit bats from the genus Pteropus. Five of the 14 species of bats in Malaysia have been identified as carriers, and this virus affects horses, cats, dogs, pigs and humans. Various mechanisms of transmission have been proposed such as contamination of date palm saps by bat feces and saliva, nosocomial and human-to-human transmissions. Physical contact was identified as the strongest risk factor for developing an infection in the 2004 Faridpur outbreak. Geographically, the virus seems to favor the Indian sub-continent, Indonesia, Southeast Asia, Pakistan, southern China, northern Australia and the Philippines, as demonstrated by the multiple outbreaks in 2001, 2004, 2007, 2012 in Bangladesh, India and Pakistan as well as the initial outbreaks in Malaysia and Singapore. Multiple routes of the viremic spread in the human body have been identified such as the central nervous system (CNS) and respiratory system, while virus levels in the body remain low, detection in the cerebrospinal fluid is comparatively high. The virus follows an incubation period of 4 days to 2 weeks which is followed by the development of symptoms. The primary clinical signs include fever, headache, vomiting and dizziness, while the characteristic symptoms consist of segmental myoclonus, tachycardia, areflexia, hypotonia, abnormal pupillary reflexes and hypertension. The serum neutralization test (SNT) is the gold standard of diagnosis followed by ELISA if SNT cannot be carried out. On the other hand, treatment is supportive since there a lack of effective pharmacological therapy and only one equine vaccine is currently licensed for use. Prevention of outbreaks seems to be a more viable approach until specific therapeutic strategies are devised.


Asunto(s)
Enfermedades Transmisibles Emergentes/epidemiología , Epidemias/estadística & datos numéricos , Infecciones por Henipavirus/epidemiología , Virus Nipah , Animales , Asia/epidemiología , Gatos , Quirópteros/virología , Enfermedades Transmisibles Emergentes/terapia , Enfermedades Transmisibles Emergentes/transmisión , Enfermedades Transmisibles Emergentes/veterinaria , Reservorios de Enfermedades , Perros , Infecciones por Henipavirus/terapia , Infecciones por Henipavirus/transmisión , Infecciones por Henipavirus/veterinaria , Caballos , Humanos , Virus Nipah/genética , Virus Nipah/aislamiento & purificación , Pinocitosis , Porcinos , Evaluación de Síntomas , Vacunación/métodos , Vacunación/veterinaria , Internalización del Virus
20.
PLoS Negl Trop Dis ; 13(6): e0007393, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31246966

RESUMEN

The 2018 outbreak of Nipah virus in Kerala, India, highlights the need for global surveillance of henipaviruses in bats, which are the reservoir hosts for this and other viruses. Nipah virus, an emerging paramyxovirus in the genus Henipavirus, causes severe disease and stuttering chains of transmission in humans and is considered a potential pandemic threat. In May 2018, an outbreak of Nipah virus began in Kerala, > 1800 km from the sites of previous outbreaks in eastern India in 2001 and 2007. Twenty-three people were infected and 21 people died (16 deaths and 18 cases were laboratory confirmed). Initial surveillance focused on insectivorous bats (Megaderma spasma), whereas follow-up surveys within Kerala found evidence of Nipah virus in fruit bats (Pteropus medius). P. medius is the confirmed host in Bangladesh and is now a confirmed host in India. However, other bat species may also serve as reservoir hosts of henipaviruses. To inform surveillance of Nipah virus in bats, we reviewed and analyzed the published records of Nipah virus surveillance globally. We applied a trait-based machine learning approach to a subset of species that occur in Asia, Australia, and Oceana. In addition to seven species in Kerala that were previously identified as Nipah virus seropositive, we identified at least four bat species that, on the basis of trait similarity with known Nipah virus-seropositive species, have a relatively high likelihood of exposure to Nipah or Nipah-like viruses in India. These machine-learning approaches provide the first step in the sequence of studies required to assess the risk of Nipah virus spillover in India. Nipah virus surveillance not only within Kerala but also elsewhere in India would benefit from a research pipeline that included surveys of known and predicted reservoirs for serological evidence of past infection with Nipah virus (or cross reacting henipaviruses). Serosurveys should then be followed by longitudinal spatial and temporal studies to detect shedding and isolate virus from species with evidence of infection. Ecological studies will then be required to understand the dynamics governing prevalence and shedding in bats and the contacts that could pose a risk to public health.


Asunto(s)
Quirópteros/virología , Control de Enfermedades Transmisibles/organización & administración , Transmisión de Enfermedad Infecciosa , Monitoreo Epidemiológico , Infecciones por Henipavirus/epidemiología , Virus Nipah/crecimiento & desarrollo , Zoonosis/epidemiología , Animales , Reservorios de Enfermedades/virología , Infecciones por Henipavirus/transmisión , Infecciones por Henipavirus/veterinaria , Humanos , India/epidemiología , Virus Nipah/inmunología , Virus Nipah/aislamiento & purificación , Medición de Riesgo , Estudios Seroepidemiológicos , Zoonosis/transmisión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...