Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Microbiol ; 15: 1341842, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38435695

RESUMO

As the climate changes, global systems have become increasingly unstable and unpredictable. This is particularly true for many disease systems, including subtypes of highly pathogenic avian influenzas (HPAIs) that are circulating the world. Ecological patterns once thought stable are changing, bringing new populations and organisms into contact with one another. Wild birds continue to be hosts and reservoirs for numerous zoonotic pathogens, and strains of HPAI and other pathogens have been introduced into new regions via migrating birds and transboundary trade of wild birds. With these expanding environmental changes, it is even more crucial that regions or counties that previously did not have surveillance programs develop the appropriate skills to sample wild birds and add to the understanding of pathogens in migratory and breeding birds through research. For example, little is known about wild bird infectious diseases and migration along the Mediterranean and Black Sea Flyway (MBSF), which connects Europe, Asia, and Africa. Focusing on avian influenza and the microbiome in migratory wild birds along the MBSF, this project seeks to understand the determinants of transboundary disease propagation and coinfection in regions that are connected by this flyway. Through the creation of a threat reduction network for avian diseases (Avian Zoonotic Disease Network, AZDN) in three countries along the MBSF (Georgia, Ukraine, and Jordan), this project is strengthening capacities for disease diagnostics; microbiomes; ecoimmunology; field biosafety; proper wildlife capture and handling; experimental design; statistical analysis; and vector sampling and biology. Here, we cover what is required to build a wild bird infectious disease research and surveillance program, which includes learning skills in proper bird capture and handling; biosafety and biosecurity; permits; next generation sequencing; leading-edge bioinformatics and statistical analyses; and vector and environmental sampling. Creating connected networks for avian influenzas and other pathogen surveillance will increase coordination and strengthen biosurveillance globally in wild birds.

2.
Health Secur ; 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38407830

RESUMO

Pathogens threaten human lives and disrupt economies around the world. This has been clearly illustrated by the current COVID-19 pandemic and outbreaks in livestock and food crops. To manage pathogen emergence and spread, cooperative engagement programs develop and strengthen biosafety, biosecurity, and biosurveillance capabilities among local researchers to detect pathogens. In this case study, we describe the efforts of a collaboration between the Los Alamos National Laboratory and the Uganda Virus Research Institute, the primary viral diagnostic laboratory in Uganda, to implement and ensure the sustainability of sequencing for biosurveillance. We describe the process of establishing this capability along with the lessons learned from both sides of the partnership to inform future cooperative engagement efforts in low- and middle-income countries. We found that by strengthening sequencing capabilities at the Uganda Virus Research Institute before the COVID-19 pandemic, the institute was able to successfully sequence SARS-CoV-2 samples and provide data to the scientific community. We highlight the need to strengthen and sustain capabilities through in-country training, collaborative research projects, and trust.

3.
Sci Rep ; 13(1): 20192, 2023 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-37980384

RESUMO

In Sub-Saharan Africa (SSA), effective brucellosis control is limited, in part, by the lack of long-term commitments by governments to control the disease and the absence of reliable national human and livestock population-based data to inform policies. Therefore, we conducted a study to establish the national prevalence and develop a risk map for Brucella spp. in cattle to contribute to plans to eliminate the disease in Kenya by the year 2040. We randomly generated 268 geolocations and distributed them across Kenya, proportionate to the area of each of the five agroecological zones and the associated cattle population. Cattle herds closest to each selected geolocation were identified for sampling. Up to 25 cattle were sampled per geolocation and a semi-structured questionnaire was administered to their owners. We tested 6,593 cattle samples for Brucella immunoglobulin G (IgG) antibodies using an Enzyme-linked immunosorbent assay (ELISA). We assessed potential risk factors and performed spatial analyses and prevalence mapping using approximate Bayesian inference implemented via the integrated nested Laplace approximation (INLA) method. The national Brucella spp. prevalence was 6.8% (95% CI: 6.2-7.4%). Exposure levels varied significantly between agro-ecological zones, with a high of 8.5% in the very arid zone with the lowest agricultural potential relative to a low of 0.0% in the agro-alpine zone with the highest agricultural potential. Additionally, seroprevalence increased with herd size, and the odds of seropositivity were significantly higher for females and adult animals than for males or calves. Similarly, animals with a history of abortion, or with multiple reproductive syndromes had higher seropositivity than those without. At the herd level, the risk of Brucella spp. transmission was higher in larger herds, and herds with a history of reproductive problems such as abortion, giving birth to weak calves, or having swollen testes. Geographic localities with high Brucella seroprevalence occurred in northern, eastern, and southern regions of Kenya all primarily characterized by semi-arid or arid agro-ecological zones dominated by livestock pastoralism interspersed with vast areas with mixed livestock-wildlife systems. The large spatial extent of our survey provides compelling evidence for the widespread geographical distribution of brucellosis risk across Kenya in a manner easily understandable for policymakers. Our findings can provide a basis for risk-stratified pilot studies aiming to investigate the cost-effectiveness and efficacy of singular and combined preventive intervention strategies that seek to inform Kenya's Brucellosis Control Policy.


Assuntos
Brucella , Brucelose , Animais , Bovinos , Feminino , Masculino , Gravidez , Criação de Animais Domésticos , Anticorpos Antibacterianos , Teorema de Bayes , Brucelose/epidemiologia , Brucelose/veterinária , Estudos Transversais , Quênia/epidemiologia , Gado , Fatores de Risco , Estudos Soroepidemiológicos
4.
PLoS Negl Trop Dis ; 17(10): e0011677, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37797043

RESUMO

Brucella spp. and Rift Valley fever virus (RVFV) are classified as priority zoonotic agents in Kenya, based on their public health and socioeconomic impact on the country. Data on the pathogen-specific and co-exposure levels is scarce due to limited active surveillance. This study investigated seroprevalence and co-exposure of Brucella spp. and RVFV and associated risk factors among slaughterhouse workers in Isiolo County, northern Kenya. A cross-sectional serosurvey was done in all 19 slaughterhouses in Isiolo County, enrolling 378 participants into the study. The overall seroprevalences for Brucella spp. and RVFV were 40.2% (95% CI: 35.2-45.4) and 18.3% (95% CI: 14.5-22.5), respectively while 10.3% (95% CI 7.4%-13.8%) of individuals were positive for antibodies against both Brucella spp. and RVFV. Virus neutralisation tests (VNT) confirmed anti-RVFV antibodies in 85% of ELISA-positive samples. Our seroprevalence results were comparable to community-level seroprevalences previously reported in the area. Since most of the study participants were not from livestock-keeping households, our findings attribute most of the detected infections to occupational exposure. The high exposure levels indicate slaughterhouse workers are the most at-risk population and there is need for infection, prevention, and control programs among this high-risk group. This is the first VNT confirmation of virus-neutralising antibodies among slaughterhouse workers in Isiolo County and corroborates reports of the area being a high-risk RVFV area as occasioned by previously reported outbreaks. This necessitates sensitization campaigns to enhance awareness of the risks involved and appropriate mitigation measures.


Assuntos
Brucella , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Matadouros , Estudos Soroepidemiológicos , Quênia/epidemiologia , Estudos Transversais , Anticorpos Antivirais
5.
Front Public Health ; 11: 1194964, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37529427

RESUMO

Abattoirs are facilities where livestock are slaughtered and are an important aspect in the food production chain. There are several types of abattoirs, which differ in infrastructure and facilities, sanitation and PPE practices, and adherence to regulations. In each abattoir facility, worker exposure to animals and animal products increases their risk of infection from zoonotic pathogens. Backyard abattoirs and slaughter slabs have the highest risk of pathogen transmission because of substandard hygiene practices and minimal infrastructure. These abattoir conditions can often contribute to environmental contamination and may play a significant role in disease outbreaks within communities. To assess further the risk of disease, we conducted a scoping review of parasites and pathogens among livestock and human workers in abattoirs across 13 Eastern African countries, which are hotspots for zoonoses. Our search results (n = 104 articles) showed the presence of bacteria, viruses, fungi, and macroparasites (nematodes, cestodes, etc.) in cattle, goats, sheep, pigs, camels, and poultry. Most articles reported results from cattle, and the most frequent pathogen detected was Mycobacterium bovis, which causes bovine tuberculosis. Some articles included worker survey and questionnaires that suggested how the use of PPE along with proper worker training and safe animal handling practices could reduce disease risk. Based on these findings, we discuss ways to improve abattoir biosafety and increase biosurveillance for disease control and mitigation. Abattoirs are a 'catch all' for pathogens, and by surveying animals at abattoirs, health officials can determine which diseases are prevalent in different regions and which pathogens are most likely transmitted from wildlife to livestock. We suggest a regional approach to biosurveillance, which will improve testing and data gathering for enhanced disease risk mapping and forecasting. Next generation sequencing will be key in identifying a wide range of pathogens, rather than a targeted approach.


Assuntos
Parasitos , Humanos , Bovinos , Animais , Suínos , Ovinos , Matadouros , Zoonoses/epidemiologia , África Oriental , Higiene , Gado/microbiologia
6.
PNAS Nexus ; 2(8): pgad241, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37614675

RESUMO

Chelonians (turtles, tortoises, and sea turtles) grow scute keratin in sequential layers over time. Once formed, scute keratin acts as an inert reservoir of environmental information. For chelonians inhabiting areas with legacy or modern nuclear activities, their scute has the potential to act as a time-stamped record of radionuclide contamination in the environment. Here, we measure bulk (i.e. homogenized scute) and sequential samples of chelonian scute from the Republic of the Marshall Islands and throughout the United States of America, including at the Barry M. Goldwater Air Force Range, southwestern Utah, the Savannah River Site, and the Oak Ridge Reservation. We identify legacy uranium (235U and 236U) contamination in bulk and sequential chelonian scute that matches known nuclear histories at these locations during the 20th century. Our results confirm that chelonians bioaccumulate uranium radionuclides and do so sequentially over time. This technique provides both a time series approach for reconstructing nuclear histories from significant past and present contexts throughout the world and the ability to use chelonians for long-term environmental monitoring programs (e.g. sea turtles at Enewetok and Bikini Atolls in the Republic of the Marshall Islands and in Japan near the Fukushima Daiichi reactors).

7.
Front Public Health ; 11: 1018293, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36741948

RESUMO

Climate change impacts global ecosystems at the interface of infectious disease agents and hosts and vectors for animals, humans, and plants. The climate is changing, and the impacts are complex, with multifaceted effects. In addition to connecting climate change and infectious diseases, we aim to draw attention to the challenges of working across multiple disciplines. Doing this requires concentrated efforts in a variety of areas to advance the technological state of the art and at the same time implement ideas and explain to the everyday citizen what is happening. The world's experience with COVID-19 has revealed many gaps in our past approaches to anticipating emerging infectious diseases. Most approaches to predicting outbreaks and identifying emerging microbes of major consequence have been with those causing high morbidity and mortality in humans and animals. These lagging indicators offer limited ability to prevent disease spillover and amplifications in new hosts. Leading indicators and novel approaches are more valuable and now feasible, with multidisciplinary approaches also within our grasp to provide links to disease predictions through holistic monitoring of micro and macro ecological changes. In this commentary, we describe niches for climate change and infectious diseases as well as overarching themes for the important role of collaborative team science, predictive analytics, and biosecurity. With a multidisciplinary cooperative "all call," we can enhance our ability to engage and resolve current and emerging problems.


Assuntos
COVID-19 , Doenças Transmissíveis Emergentes , Doenças Transmissíveis , Humanos , Animais , Ecossistema , Mudança Climática , COVID-19/epidemiologia , Doenças Transmissíveis/epidemiologia , Doenças Transmissíveis Emergentes/epidemiologia
8.
Mol Aspects Med ; 91: 101142, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36116999

RESUMO

Topics expected to influence personalized medicine (PM), where medical decisions, practices, and treatments are tailored to the individual patient, are reviewed. Lack of discrimination due to different biological conditions that express similar values of numerical variables (ambiguity) is regarded to be a major potential barrier for PM. This material explores possible causes and sources of ambiguity and offers suggestions for mitigating the impacts of uncertainties. Three causes of ambiguity are identified: (1) delayed adoption of innovations, (2) inadequate emphases, and (3) inadequate processes used when new medical practices are developed and validated. One example of the first problem is the relative lack of medical research on "compositional data" -the type that characterizes leukocyte data. This omission results in erroneous use of data abundantly utilized in medicine, such as the blood cell differential. Emphasis on data output ‒not biomedical interpretation that facilitates the use of clinical data‒ exemplifies the second type of problems. Reliance on tools generated in other fields (but not validated within biomedical contexts) describes the last limitation. Because reductionism is associated with these problems, non-reductionist alternatives are reviewed as potential remedies. Data structuring (converting data into information) is considered a key element that may promote PM. To illustrate a process that includes data-information-knowledge and decision-making, previously published data on COVID-19 are utilized. It is suggested that ambiguity may be prevented or ameliorated. Provided that validations are grounded on biomedical knowledge, approaches that describe certain criteria - such as non-overlapping data intervals of patients that experience different outcomes, immunologically interpretable data, and distinct graphic patterns - can inform, at personalized bases, earlier and/or with fewer observations.


Assuntos
COVID-19 , Humanos , COVID-19/epidemiologia , Medicina de Precisão/métodos , Leucócitos
9.
Front Vet Sci ; 10: 1270505, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38179332

RESUMO

Introduction: Control of zoonosis can benefit from geo-referenced procedures. Focusing on brucellosis, here the ability of two methods to distinguish disease dissemination patterns and promote cost-effective interventions was compared. Method: Geographical data on bovine, ovine and human brucellosis reported in the country of Georgia between 2014 and 2019 were investigated with (i) the Hot Spot (HS) analysis and (ii) a bio-geographical (BG) alternative. Results: More than one fourth of all sites reported cases affecting two or more species. While ruminant cases displayed different patterns over time, most human cases described similar geo-temporal features, which were associated with the route used by migrant shepherds. Other human cases showed heterogeneous patterns. The BG approach identified small areas with a case density twice as high as the HS method. The BG method also identified, in 2018, a 2.6-2.99 higher case density in zoonotic (human and non-human) sites than in non-zoonotic sites (which only reported cases affecting a single species) -a finding that, if corroborated, could support cost-effective policy-making. Discussion: Three dissemination hypotheses were supported by the data: (i) human cases induced by sheep-related contacts; (ii) human cases probably mediated by contaminated milk or meat; and (iii) cattle and sheep that infected one another. This proof-of-concept provided a preliminary validation for a method that may support cost-effective interventions oriented to control zoonoses. To expand these findings, additional studies on zoonosis-related decision-making are recommended.

10.
PLoS Negl Trop Dis ; 16(8): e0010596, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35939503

RESUMO

BACKGROUND: Co-infection, especially with pathogens of dissimilar genetic makeup, may result in a more devastating impact on the host. Investigations on co-infection with neglected zoonotic pathogens in wildlife are necessary to inform appropriate prevention and control strategies to reduce disease burden in wildlife and the potential transmission of these pathogens between wildlife, livestock and humans. This study assessed co-exposure of various Kenyan wildflife species with Brucella spp, Coxiella burnetii and Rift Valley fever virus (RVFV). METHODOLOGY: A total of 363 sera from 16 different wildlife species, most of them (92.6%) herbivores, were analysed by Enzyme-linked immunosorbent assay (ELISA) for IgG antibodies against Brucella spp, C. burnetii and RVFV. Further, 280 of these were tested by PCR to identify Brucella species. RESULTS: Of the 16 wildlife species tested, 15 (93.8%) were seropositive for at least one of the pathogens. Mean seropositivities were 18.9% (95% CI: 15.0-23.3) for RVFV, 13.7% (95% CI: 10.3-17.7) for Brucella spp and 9.1% (95% CI: 6.3-12.5) for C. burnetii. Buffaloes (n = 269) had higher seropositivity for Brucella spp. (17.1%, 95% CI: 13.0-21.7%) and RVFV (23.4%, 95% CI: 18.6-28.6%), while giraffes (n = 36) had the highest seropositivity for C. burnetii (44.4%, 95% CI: 27.9-61.9%). Importantly, 23 of the 93 (24.7%) animals positive for at least one pathogen were co-exposed, with 25.4% (18/71) of the positive buffaloes positive for brucellosis and RVFV. On molecular analysis, Brucella DNA was detected in 46 (19.5%, CI: 14.9-24.7) samples, with 4 (8.6%, 95% CI: 2.2-15.8) being identified as B. melitensis. The Fisher's Exact test indicated that seropositivity varied significantly within the different animal families, with Brucella (p = 0.013), C. burnetii (p = <0.001) and RVFV (p = 0.007). Location was also significantly associated (p = <0.001) with Brucella spp. and C. burnetii seropositivities. CONCLUSION: Of ~20% of Kenyan wildlife that are seropositive for Brucella spp, C. burnetii and RVFV, almost 25% indicate co-infections with the three pathogens, particularly with Brucella spp and RVFV.


Assuntos
Brucella , Coinfecção , Coxiella burnetii , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Animais Selvagens , Brucella/genética , Búfalos , Coinfecção/epidemiologia , Coinfecção/veterinária , Coxiella burnetii/genética , Humanos , Quênia/epidemiologia , Vírus da Febre do Vale do Rift/genética , Estudos Soroepidemiológicos , Zoonoses
11.
Anim Microbiome ; 4(1): 45, 2022 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-35908068

RESUMO

Microbiomes are essential to a host's physiology and health. Despite the overall importance of microbiomes to animal health, they remain understudied in wildlife. Microbiomes function as physical barriers to invading pathogens, and changes in the diversity or composition of microbes within a host may disrupt this barrier. In order to use microbiomes in wildlife ecology, knowledge of the natural variation within and among species is essential. We compare the diversity and composition of two avian species that share the same habitat and niche in our study area, the western screech-owl (Megascops kennicottii) and the whiskered screech-owl (M. trichopsis). We used a targeted 16S sequencing method to improve the taxonomic resolution of microbiomes. We found similar measures of alpha diversity between species and sample types (cloacal samples vs. fecal samples). However, there were significant differences in bacterial species richness among nestlings from different nest boxes, and the composition differed between the two bird species and among nestlings from different nest boxes. Western screech-owls had more variation in alpha diversity and composition and had fewer bacterial species in their core microbiome than whiskered screech-owls. Siblings are likely to yield similar findings for microbiomes; thus, sampling nestlings from different nests may be most informative for monitoring population-level changes.

12.
Zoonoses Public Health ; 69(3): 175-194, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35034427

RESUMO

Zoonotic diseases, such as brucellosis, Q fever and Rift Valley fever (RVF) caused by Brucella spp., Coxiella burnetii and RVF virus, respectively, can have devastating effects on human, livestock, and wildlife health and cause economic hardship due to morbidity and mortality in livestock. Coinfection with multiple pathogens can lead to more severe disease outcomes and altered transmission dynamics. These three pathogens can alter host immune responses likely leading to increased morbidity, mortality and pathogen transmission during coinfection. Developing countries, such as those commonly afflicted by outbreaks of brucellosis, Q fever and RVF, have high disease burden and thus common coinfections. A literature survey provided information on case reports and studies investigating coinfections involving the three focal diseases. Fifty five studies were collected demonstrating coinfections of Brucella spp., C. burnetii or RVFV with 50 different pathogens, of which 64% were zoonotic. While the literature search criteria involved 'coinfection', only 24/55 studies showed coinfections with direct pathogen detection methods (microbiology, PCR and antigen test), while the rest only reported detection of antibodies against multiple pathogens, which only indicate a history of co-exposure, not concurrent infection. These studies lack the ability to test whether coinfection leads to changes in morbidity, mortality or transmission dynamics. We describe considerations and methods for identifying ongoing coinfections to address this critical blind spot in disease risk management.


Assuntos
Brucella , Brucelose , Coinfecção , Coxiella burnetii , Febre Q , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Animais Selvagens , Brucelose/epidemiologia , Brucelose/veterinária , Coinfecção/epidemiologia , Coinfecção/veterinária , Humanos , Gado/microbiologia , Febre Q/epidemiologia , Febre Q/veterinária , Febre do Vale de Rift/epidemiologia , Estudos Soroepidemiológicos
13.
PLOS Glob Public Health ; 2(7): e0000811, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36962439

RESUMO

Early and accurate diagnosis of respiratory pathogens and associated outbreaks can allow for the control of spread, epidemiological modeling, targeted treatment, and decision making-as is evident with the current COVID-19 pandemic. Many respiratory infections share common symptoms, making them difficult to diagnose using only syndromic presentation. Yet, with delays in getting reference laboratory tests and limited availability and poor sensitivity of point-of-care tests, syndromic diagnosis is the most-relied upon method in clinical practice today. Here, we examine the variability in diagnostic identification of respiratory infections during the annual infection cycle in northern New Mexico, by comparing syndromic diagnostics with polymerase chain reaction (PCR) and sequencing-based methods, with the goal of assessing gaps in our current ability to identify respiratory pathogens. Of 97 individuals that presented with symptoms of respiratory infection, only 23 were positive for at least one RNA virus, as confirmed by sequencing. Whereas influenza virus (n = 7) was expected during this infection cycle, we also observed coronavirus (n = 7), respiratory syncytial virus (n = 8), parainfluenza virus (n = 4), and human metapneumovirus (n = 1) in individuals with respiratory infection symptoms. Four patients were coinfected with two viruses. In 21 individuals that tested positive using PCR, RNA sequencing completely matched in only 12 (57%) of these individuals. Few individuals (37.1%) were diagnosed to have an upper respiratory tract infection or viral syndrome by syndromic diagnostics, and the type of virus could only be distinguished in one patient. Thus, current syndromic diagnostic approaches fail to accurately identify respiratory pathogens associated with infection and are not suited to capture emerging threats in an accurate fashion. We conclude there is a critical and urgent need for layered agnostic diagnostics to track known and unknown pathogens at the point of care to control future outbreaks.

14.
Parasit Vectors ; 14(1): 547, 2021 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-34688314

RESUMO

BACKGROUND: Estimates of the geographical distribution of Culex mosquitoes in the Americas have been limited to state and provincial levels in the United States and Canada and based on data from the 1980s. Since these estimates were made, there have been many more documented observations of mosquitoes and new methods have been developed for species distribution modeling. Moreover, mosquito distributions are affected by environmental conditions, which have changed since the 1980s. This calls for updated estimates of these distributions to understand the risk of emerging and re-emerging mosquito-borne diseases. METHODS: We used contemporary mosquito data, environmental drivers, and a machine learning ecological niche model to create updated estimates of the geographical range of seven predominant Culex species across North America and South America: Culex erraticus, Culex nigripalpus, Culex pipiens, Culex quinquefasciatus, Culex restuans, Culex salinarius, and Culex tarsalis. RESULTS: We found that Culex mosquito species differ in their geographical range. Each Culex species is sensitive to both natural and human-influenced environmental factors, especially climate and land cover type. Some prefer urban environments instead of rural ones, and some are limited to tropical or humid areas. Many are found throughout the Central Plains of the USA. CONCLUSIONS: Our updated contemporary Culex distribution maps may be used to assess mosquito-borne disease risk. It is critical to understand the current geographical distributions of these important disease vectors and the key environmental predictors structuring their distributions not only to assess current risk, but also to understand how they will respond to climate change. Since the environmental predictors structuring the geographical distribution of mosquito species varied, we hypothesize that each species may have a different response to climate change.


Assuntos
Distribuição Animal , Culex/fisiologia , Mosquitos Vetores/fisiologia , América , Animais , Mudança Climática , Culex/classificação , Culex/parasitologia , Culex/virologia , Humanos , Aprendizado de Máquina , Mosquitos Vetores/classificação , Mosquitos Vetores/parasitologia , Mosquitos Vetores/virologia , América do Norte , América do Sul
15.
Front Public Health ; 9: 659695, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34568249

RESUMO

The current COVID-19 pandemic demonstrates the need for urgent and on-demand solutions to provide diagnostics, treatment and preventative measures for infectious disease outbreaks. Once solutions are developed, meeting capacities depends on the ability to mitigate technical, logistical and production issues. While it is difficult to predict the next outbreak, augmenting investments in preparedness, such as infectious disease surveillance, is far more effective than mustering last-minute response funds. Bringing research outputs into practice sooner rather than later is part of an agile approach to pivot and deliver solutions. Cooperative multi- country research programs, especially those funded by global biosecurity programs, develop capacity that can be applied to infectious disease surveillance and research that enhances detection, identification, and response to emerging and re-emerging pathogens with epidemic or pandemic potential. Moreover, these programs enhance trust building among partners, which is essential because setting expectation and commitment are required for successful research and training. Measuring research outputs, evaluating outcomes and justifying continual investments are essential but not straightforward. Lessons learned include those related to reducing biological threats and maturing capabilities for national laboratory diagnostics strategy and related health systems. Challenges, such as growing networks, promoting scientific transparency, data and material sharing, sustaining funds and developing research strategies remain to be fully resolved. Here, experiences from several programs highlight successful partnerships that provide ways forward to address the next outbreak.


Assuntos
COVID-19 , Doenças Transmissíveis , Doenças Transmissíveis/diagnóstico , Surtos de Doenças/prevenção & controle , Humanos , Pandemias , SARS-CoV-2
16.
Animals (Basel) ; 11(8)2021 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-34438914

RESUMO

Geographic ranges of plants and animals are shifting due to environmental change. While some species are shifting towards the poles and upslope in elevation, the processes leading to these patterns are not well known. We analyzed 22 years of western bluebird (Sialia mexicana) data from a large nest box network in northern New Mexico at elevations between 1860 m and 2750 m. This population has shifted to higher elevations over time, but whether this is due to changes in nesting behavior and preference for higher elevation within the population or driven by immigration is unclear. We banded adults and nestlings from nest boxes and examined nesting location and elevation for individual birds captured two or more times. Most recaptured birds nested at the same nest boxes in subsequent years, and the number of birds that moved upslope did not significantly differ from the number that moved downslope. Fledglings moved greater distances and elevations than adults, but these movements were not upslope specific. Female fledglings showed greater changes in elevation and distance compared to male fledglings, but again, movements were not consistently upslope. The upslope shift in this population may be due to birds immigrating into the population and not from changes in individual nesting behavior.

17.
Front Public Health ; 9: 648424, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33732679

RESUMO

The threat of emerging and re-emerging infectious diseases continues to be a challenge to public and global health security. Cooperative biological engagement programs act to build partnerships and collaborations between scientists and health professionals to strengthen capabilities in biosurveillance. Biosurveillance is the systematic process of detecting, reporting, and responding to especially dangerous pathogens and pathogens of pandemic potential before they become outbreaks, epidemics, and pandemics. One important tool in biosurveillance is next generation sequencing. Expensive sequencing machines, reagents, and supplies make it difficult for countries to adopt this technology. Cooperative engagement programs help by providing funding for technical assistance to strengthen sequencing capabilities. Through workshops and training, countries are able to learn sequencing and bioinformatics, and implement these tools in their biosurveillance programs. Cooperative programs have an important role in building and sustaining collaborations among institutions and countries. One of the most important pieces in fostering these collaborations is trust. Trust provides the confidence that a successful collaboration will benefit all parties involved. With sequencing, this enables the sharing of pathogen samples and sequences. Obtaining global sequencing data helps to identify unknown etiological agents, track pathogen evolution and infer transmission networks throughout the duration of a pandemic. Having sequencing technology in place for biosurveillance generates the capacity to provide real-time data to understand and respond to pandemics. We highlight the need for these programs to continue to strengthen sequencing in biosurveillance. By working together to strengthen sequencing capabilities, trust can be formed, benefitting global health in the face of biological threats.


Assuntos
Biovigilância , Surtos de Doenças/prevenção & controle , Saúde Global , Pandemias
18.
Methods ; 195: 77-91, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33744397

RESUMO

The current COVID-19 pandemic contains an unprecedented amount of uncertainty and variability and thus, there is a critical need for understanding of the variation documented in the biological, policy, sociological, and infrastructure responses during an epidemic to support decisions at all levels. With the significant asymptomatic spread of the virus and without an immediate vaccine and pharmaceuticals available, the best feasible strategies for testing and diagnostics, contact tracing, and quarantine need to be optimized. With potentially high false negative test results, infected people would not be enrolled in contact-trace programs and thus, may not be quarantined. Similarly, without broad testing, asymptomatic people are not identified and quarantined. Interconnected system dynamics models can be used to optimize strategies for mitigations for decision support during a pandemic. We use a systems dynamics epidemiology model along with other interconnected system models within public health including hospitals, intensive care units, masks, contact tracing, social distancing, and a newly developed testing and diagnostics model to investigate the uncertainties with testing and to optimize strategies for detecting and diagnosing infected people. Using an orthogonal array Latin Hypercube experimental design, we ran 54 simulations each for two scenarios of 10% and 30% asymptomatic people, varying important inputs for testing and social distancing. Systems dynamics modeling, coupled with computer experimental design and statistical analysis can provide rapid and quantitative results for decision support. Our results show that widespread testing, contacting tracing and quarantine can curtail the pandemic through identifying asymptomatic people in the population.


Assuntos
COVID-19/diagnóstico , COVID-19/epidemiologia , Busca de Comunicante/métodos , Modelos Biológicos , Análise de Sistemas , Incerteza , COVID-19/prevenção & controle , Humanos , Distanciamento Físico , Quarentena/métodos
20.
BMC Res Notes ; 14(1): 122, 2021 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-33785071

RESUMO

OBJECTIVE: Currently, next generation sequencing (NGS) is widely used to decode potential novel or variant pathogens both in emergent outbreaks and in routine clinical practice. However, the efficient identification of novel or diverged pathogenomic compositions remains a big challenge. It is especially true for short DNA sequence fragments from NGS, since sequence similarity searching is vulnerable to false negatives or false positives, as is mismatching or matching with unrelated proteins. Therefore, this study aimed to establish a bioinformatics approach that can generate unique motif sequences for profiling searching, resulting in high specificity and sensitivity. RESULTS: In this study, we introduced a Shortest Unique Representative Hidden Markov Model (HMM) approach to identify bacterial toxin, virulence factor (VF), and antimicrobial resistance (AR) in short sequence reads. We first construct unique representative domain sequences of toxin genes, VFs, and ARs to avoid potential false positives, and then to use HMM models to accurately identify potential toxin, VF, and AR fragments. The benchmark shows this approach can achieve relatively high specificity and sensitivity if the appropriate cutoff value is applied. Our approach can be used to recognize the protein sequences of known toxins and pathogens, identifies their common characteristics and then searches for similar sequences in other organisms.


Assuntos
Antibacterianos , Fatores de Virulência , Antibacterianos/farmacologia , Resistência Microbiana a Medicamentos , Cadeias de Markov , Proteínas , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...