Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Emerg Infect Dis ; 30(4): 681-690, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38526081

ABSTRACT

Although pigs are naturally susceptible to Reston virus and experimentally to Ebola virus (EBOV), their role in Orthoebolavirus ecology remains unknown. We tested 888 serum samples collected from pigs in Guinea during 2017-2019 (between the 2013-16 epidemic and its resurgence in 2021) by indirect ELISA against the EBOV nucleoprotein. We identified 2 hotspots of possible pig exposure by IgG titer levels: the northern coast had 48.7% of positive serum samples (37/76), and Forest Guinea, bordering Sierra Leone and Liberia, where the virus emerged and reemerged, had 50% of positive serum samples (98/196). The multitarget Luminex approach confirms ELISA results against Ebola nucleoprotein and highlights cross-reactivities to glycoprotein of EBOV, Reston virus, and Bundibugyo virus. Those results are consistent with previous observations of the circulation of Orthoebolavirus species in pig farming regions in Sierra Leone and Ghana, suggesting potential risk for Ebola virus disease in humans, especially in Forest Guinea.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Swine , Animals , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/veterinary , Guinea/epidemiology , Sus scrofa , Sierra Leone/epidemiology , Nucleoproteins/genetics
2.
Math Biosci Eng ; 20(11): 19686-19709, 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-38052620

ABSTRACT

Past works on partially diffusive models of diseases typically rely on a strong assumption regarding the initial data of their infection-related compartments in order to demonstrate uniform persistence in the case that the basic reproduction number $ \mathcal{R}_0 $ is above 1. Such a model for avian influenza was proposed, and its uniform persistence was proven for the case $ \mathcal{R}_0 > 1 $ when all of the infected bird population, recovered bird population and virus concentration in water do not initially vanish. Similarly, a work regarding a model of the Ebola virus disease required that the infected human population does not initially vanish to show an analogous result. We introduce a modification on the standard method of proving uniform persistence, extending both of these results by weakening their respective assumptions to requiring that only one (rather than all) infection-related compartment is initially non-vanishing. That is, we show that, given $ \mathcal{R}_0 > 1 $, if either the infected bird population or the viral concentration are initially nonzero anywhere in the case of avian influenza, or if any of the infected human population, viral concentration or population of deceased individuals who are under care are initially nonzero anywhere in the case of the Ebola virus disease, then their respective models predict uniform persistence. The difficulty which we overcome here is the lack of diffusion, and hence the inability to apply the minimum principle, in the equations of the avian influenza virus concentration in water and of the population of the individuals deceased due to the Ebola virus disease who are still in the process of caring.


Subject(s)
Communicable Diseases , Hemorrhagic Fever, Ebola , Influenza A virus , Influenza in Birds , Influenza, Human , Animals , Humans , Influenza in Birds/epidemiology , Hemorrhagic Fever, Ebola/veterinary , Birds , Water , Influenza, Human/epidemiology
3.
Vet Pathol ; 60(4): 473-487, 2023 07.
Article in English | MEDLINE | ID: mdl-37170900

ABSTRACT

The liver is an early systemic target of Ebola virus (EBOV), but characterization beyond routine histopathology and viral antigen distribution is limited. We hypothesized Ebola virus disease (EVD) systemic proinflammatory responses would be reflected in temporally altered liver myeloid phenotypes. We utilized multiplex fluorescent immunohistochemistry (mfIHC), multispectral whole slide imaging, and image analysis to quantify molecular phenotypes of myeloid cells in the liver of rhesus macaques (Macaca mulatta; n = 21) infected with EBOV Kikwit. Liver samples included uninfected controls (n = 3), 3 days postinoculation (DPI; n = 3), 4 DPI (n = 3), 5 DPI (n = 3), 6 DPI (n = 3), and terminal disease (6-8 DPI; n = 6). Alterations in hepatic macrophages occurred at ≥ 5 DPI characterized by a 1.4-fold increase in CD68+ immunoreactivity and a transition from primarily CD14-CD16+ to CD14+CD16- macrophages, with a 2.1-fold decrease in CD163 expression in terminal animals compared with uninfected controls. An increase in the neutrophil chemoattractant and alarmin S100A9 occurred within hepatic myeloid cells at 5 DPI, followed by rapid neutrophil influx at ≥ 6 DPI. An acute rise in the antiviral myxovirus resistance protein 1 (MxA) occurred at ≥ 4 DPI, with a predilection for enhanced expression in uninfected cells. Distinctive expression of major histocompatibility complex (MHC) class II was observed in hepatocytes during terminal disease. Results illustrate that EBOV causes macrophage phenotype alterations as well as neutrophil influx and prominent activation of interferon host responses in the liver. Results offer insight into potential therapeutic strategies to prevent and/or modulate the host proinflammatory response to normalize hepatic myeloid functionality.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Hemorrhagic Fever, Ebola/veterinary , Hemorrhagic Fever, Ebola/pathology , Ebolavirus/physiology , Macaca mulatta , Liver/pathology , Phenotype
4.
Sci Rep ; 13(1): 5675, 2023 04 07.
Article in English | MEDLINE | ID: mdl-37029156

ABSTRACT

Ebola virus is highly lethal for great apes. Estimated mortality rates up to 98% have reduced the global gorilla population by approximately one-third. As mountain gorillas (Gorilla beringei beringei) are endangered, with just over 1000 individuals remaining in the world, an outbreak could decimate the population. Simulation modeling was used to evaluate the potential impact of an Ebola virus outbreak on the mountain gorilla population of the Virunga Massif. Findings indicate that estimated contact rates among gorilla groups are high enough to allow rapid spread of Ebola, with less than 20% of the population projected to survive at 100 days post-infection of just one gorilla. Despite increasing survival with vaccination, no modeled vaccination strategy prevented widespread infection. However, the model projected that survival rates greater than 50% could be achieved by vaccinating at least half the habituated gorillas within 3 weeks of the first infectious individual.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Hominidae , Humans , Animals , Gorilla gorilla , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/veterinary , Disease Outbreaks/veterinary
5.
Viruses ; 15(2)2023 01 26.
Article in English | MEDLINE | ID: mdl-36851566

ABSTRACT

The Marburg and Ebola filoviruses cause a severe, often fatal, disease in humans and nonhuman primates but have only subclinical effects in bats, including Egyptian rousettes, which are a natural reservoir of Marburg virus. A fundamental question is why these viruses are highly pathogenic in humans but fail to cause disease in bats. To address this question, we infected one cohort of Egyptian rousette bats with Marburg virus and another cohort with Ebola virus and harvested multiple tissues for mRNA expression analysis. While virus transcripts were found primarily in the liver, principal component analysis (PCA) revealed coordinated changes across multiple tissues. Gene signatures in kidney and liver pointed at induction of vasodilation, reduction in coagulation, and changes in the regulation of iron metabolism. Signatures of immune response detected in spleen and liver indicated a robust anti-inflammatory state signified by macrophages in the M2 state and an active T cell response. The evolutionary divergence between bats and humans of many responsive genes might provide a framework for understanding the differing outcomes upon infection by filoviruses. In this study, we outline multiple interconnected pathways that respond to infection by MARV and EBOV, providing insights into the complexity of the mechanisms that enable bats to resist the disease caused by filoviral infections. The results have the potential to aid in the development of new strategies to effectively mitigate and treat the disease caused by these viruses in humans.


Subject(s)
Chiroptera , Ebolavirus , Filoviridae Infections , Hemorrhagic Fever, Ebola , Marburgvirus , Humans , Animals , Hemorrhagic Fever, Ebola/veterinary , Ebolavirus/genetics , Liver , Marburgvirus/genetics
6.
J Vet Med Sci ; 84(11): 1491-1494, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36123040

ABSTRACT

Some filoviruses such as ebolaviruses and marburgviruses, cause hemorrhagic fever in humans and nonhuman primates. Pigs are suggested to play a potential role in the filovirus ecology. We investigated the seroprevalence of filovirus infection in pigs in Ghana. Using a viral glycoprotein (GP)-based enzyme-linked immunosorbent assay, we detected filovirus-specific immunoglobulin G antibodies in 5 of 139 samples. These positive sera showed specificities to four different filovirus species. Particularly, two of the positive sera reacted to GPs of two African ebolaviruses (i.e., Ebola virus and Taï Forest virus) in Western blotting. Our results suggest that these Ghanaian pigs were exposed to multiple filoviruses and emphasize the importance of continuous monitoring of filovirus infection in pig populations in West African countries.


Subject(s)
Ebolavirus , Filoviridae Infections , Hemorrhagic Fever, Ebola , Swine Diseases , Swine , Humans , Animals , Ghana/epidemiology , Hemorrhagic Fever, Ebola/diagnosis , Hemorrhagic Fever, Ebola/veterinary , Seroepidemiologic Studies , Antibodies, Viral , Filoviridae Infections/veterinary , Swine Diseases/epidemiology
7.
Vector Borne Zoonotic Dis ; 22(9): 478-490, 2022 09.
Article in English | MEDLINE | ID: mdl-36084314

ABSTRACT

Outbreaks of African filoviruses often have high mortality, including more than 11,000 deaths among 28,562 cases during the West Africa Ebola outbreak of 2014-2016. Numerous studies have investigated the factors that contributed to individual filovirus outbreaks, but there has been little quantitative synthesis of this work. In addition, the ways in which the typical causes of filovirus outbreaks differ from other zoonoses remain poorly described. In this study, we quantify factors associated with 45 outbreaks of African filoviruses (ebolaviruses and Marburg virus) using a rubric of 48 candidate causal drivers. For filovirus outbreaks, we reviewed >700 peer-reviewed and gray literature sources and developed a list of the factors reported to contribute to each outbreak (i.e., a "driver profile" for each outbreak). We compare and contrast the profiles of filovirus outbreaks to 200 background outbreaks, randomly selected from a global database of 4463 outbreaks of bacterial and viral zoonotic diseases. We also test whether the quantitative patterns that we observed were robust to the influences of six covariates, country-level factors such as gross domestic product, population density, and latitude that have been shown to bias global outbreak data. We find that, regardless of whether covariates are included or excluded from models, the driver profile of filovirus outbreaks differs from that of background outbreaks. Socioeconomic factors such as trade and travel, wild game consumption, failures of medical procedures, and deficiencies in human health infrastructure were more frequently reported in filovirus outbreaks than in the comparison group. Based on our results, we also present a review of drivers reported in at least 10% of filovirus outbreaks, with examples of each provided.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Marburg Virus Disease , Marburgvirus , Animals , Disease Outbreaks , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/veterinary , Humans , Marburg Virus Disease/epidemiology
8.
J Gen Virol ; 103(8)2022 08.
Article in English | MEDLINE | ID: mdl-35972225

ABSTRACT

Bats have been implicated as the reservoir hosts of filoviruses in Africa, with serological evidence of filoviruses in various bat species identified in other countries. Here, serum samples from 190 bats, comprising 12 different species, collected in Australia were evaluated for filovirus antibodies. An in-house indirect microsphere assay to detect antibodies that cross-react with Ebola virus (Zaire ebolavirus; EBOV) nucleoprotein (NP) followed by an immunofluorescence assay (IFA) were used to confirm immunoreactivity to EBOV and Reston virus (Reston ebolavirus; RESTV). We found 27 of 102 Yinpterochiroptera and 19 of 88 Yangochiroptera samples were positive to EBOV NP in the microsphere assay. Further testing of these NP positive samples by IFA revealed nine bat sera that showed binding to ebolavirus-infected cells. This is the first report of filovirus-reactive antibodies detected in Australian bat species and suggests that novel filoviruses may be circulating in Australian bats.


Subject(s)
Chiroptera , Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Antibodies, Viral , Australia , Hemorrhagic Fever, Ebola/veterinary , Nucleoproteins
9.
PLoS Negl Trop Dis ; 16(6): e0010504, 2022 06.
Article in English | MEDLINE | ID: mdl-35731800

ABSTRACT

On the 8th of May, 2018, an outbreak of Ebola virus disease (EVD) was declared, originating in the Bikoro region of the Democratic Republic of the Congo (DRC) near the border with neighboring Republic of the Congo (ROC). Frequent trade and migration occur between DRC and ROC-based communities residing along the Congo River. In June 2018, a field team was deployed to determine whether Zaire ebolavirus (Ebola virus (EBOV)) was contemporaneously circulating in local bats at the human-animal interface in ROC near the Bikoro EVD outbreak. Samples were collected from bats in the Cuvette and Likouala departments, ROC, bordering the Équateur Province in DRC where the Bikoro EVD outbreak was first detected. EBOV genomic material was not detected in bat-derived samples by targeted quantitative reverse transcription-polymerase chain reaction or by family-level consensus polymerase chain reaction; however, serological data suggests recent exposure to EBOV in bats in the region. We collected serum from 144 bats in the Cuvette department with 6.9% seropositivity against the EBOV glycoprotein and 14.3% seropositivity for serum collected from 27 fruit bats and one Molossinae in the Likouala department. We conclude that proactive investment in longitudinal sampling for filoviruses at the human-animal interface, coupled with ecological investigations are needed to identify EBOV wildlife reservoirs.


Subject(s)
Chiroptera , Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Democratic Republic of the Congo/epidemiology , Disease Outbreaks , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/veterinary
10.
Vet Med Sci ; 8(4): 1787-1801, 2022 07.
Article in English | MEDLINE | ID: mdl-35537080

ABSTRACT

Bats are the natural reservoir host for many pathogenic and non-pathogenic viruses, potentially spilling over to humans and domestic animals directly or via an intermediate host. The ongoing COVID-19 pandemic is the continuation of virus spillover events that have taken place over the last few decades, particularly in Asia and Africa. Therefore, these bat-associated epidemics provide a significant number of hints, including respiratory cellular tropism, more intense susceptibility to these cell types, and overall likely to become a pandemic for the next spillover. In this systematic review, we analysed data to insight, through bat-originated spillover in Asia and Africa. We used STATA/IC-13 software for descriptive statistics and meta-analysis. The random effect of meta-analysis showed that the pooled estimates of case fatality rates of bat-originated viral zoonotic diseases were higher in Africa (61.06%, 95%CI: 50.26 to 71.85, l2 % = 97.3, p < 0.001). Moreover, estimates of case fatality rates were higher in Ebola (61.06%; 95%CI: 50.26 to 71.85, l2 % = 97.3, p < 0.001) followed by Nipah (55.19%; 95%CI: 39.29 to 71.09, l2 % = 94.2, p < 0.001), MERS (18.49%; 95%CI: 8.19 to 28.76, l2 % = 95.4, p < 0.001) and SARS (10.86%; 95%CI: 6.02 to 15.71, l2 % = 85.7, p < 0.001) with the overall case fatality rates of 29.86 (95%CI: 29.97 to 48.58, l2 % = 99.0, p < 0.001). Bat-originated viruses have caused several outbreaks of deadly diseases, including Nipah, Ebola, SARS and MERS in Asia and Africa in a sequential fashion. Nipah virus emerged first in Malaysia, but later, periodic outbreaks were noticed in Bangladesh and India. Similarly, the Ebola virus was detected in the African continent with neurological disorders in humans, like Nipah, seen in the Asian region. Two important coronaviruses, MERS and SARS, were introduced, both with the potential to infect respiratory passages. This paper explores the dimension of spillover events within and/or between bat-human and the epidemiological risk factors, which may lead to another pandemic occurring. Further, these processes enhance the bat-originated virus, which utilises an intermediate host to jump into human species.


Subject(s)
COVID-19 , Chiroptera , Hemorrhagic Fever, Ebola , Viruses , Africa/epidemiology , Animals , COVID-19/epidemiology , COVID-19/veterinary , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/veterinary , Humans , Pandemics
11.
BMC Genomics ; 22(1): 735, 2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34635054

ABSTRACT

BACKGROUND: Numerous Ebola virus outbreaks have occurred in Equatorial Africa over the past decades. Besides human fatalities, gorillas and chimpanzees have also succumbed to the fatal virus. The 2004 outbreak at the Odzala-Kokoua National Park (Republic of Congo) alone caused a severe decline in the resident western lowland gorilla (Gorilla gorilla gorilla) population, with a 95% mortality rate. Here, we explore the immediate genetic impact of the Ebola outbreak in the western lowland gorilla population. RESULTS: Associations with survivorship were evaluated by utilizing DNA obtained from fecal samples from 16 gorilla individuals declared missing after the outbreak (non-survivors) and 15 individuals observed before and after the epidemic (survivors). We used a target enrichment approach to capture the sequences of 123 genes previously associated with immunology and Ebola virus resistance and additionally analyzed the gut microbiome which could influence the survival after an infection. Our results indicate no changes in the population genetic diversity before and after the Ebola outbreak, and no significant differences in microbial community composition between survivors and non-survivors. However, and despite the low power for an association analysis, we do detect six nominally significant missense mutations in four genes that might be candidate variants associated with an increased chance of survival. CONCLUSION: This study offers the first insight to the genetics of a wild great ape population before and after an Ebola outbreak using target capture experiments from fecal samples, and presents a list of candidate loci that may have facilitated their survival.


Subject(s)
Gastrointestinal Microbiome , Hemorrhagic Fever, Ebola , Animals , Disease Outbreaks , Gorilla gorilla/genetics , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/veterinary , Humans , Pan troglodytes
12.
Emerg Microbes Infect ; 10(1): 1320-1330, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34112056

ABSTRACT

Ebola virus (EBOV) is a negative single-stranded RNA virus within the Filoviridae family and the causative agent of Ebola virus disease (EVD). Nonhuman primates (NHPs), including cynomolgus and rhesus macaques, are considered the gold standard animal model to interrogate mechanisms of EBOV pathogenesis. However, despite significant genetic similarity (>90%), NHP species display different clinical presentation following EBOV infection, notably a ∼1-2 days delay in disease progression. Consequently, evaluation of therapeutics is generally conducted in rhesus macaques, whereas cynomolgus macaques are utilized to determine efficacy of preventative treatments, notably vaccines. This observation is in line with reported differences in disease severity and host responses between these two NHP following infection with simian varicella virus, influenza A and SARS-CoV-2. However, the molecular underpinnings of these differential outcomes following viral infections remain poorly defined. In this study, we compared published transcriptional profiles obtained from cynomolgus and rhesus macaques infected with the EBOV-Makona Guinea C07 using bivariate and regression analyses to elucidate differences in host responses. We report the presence of a shared core of differentially expressed genes (DEGs) reflecting EVD pathology, including aberrant inflammation, lymphopenia, and coagulopathy. However, the magnitudes of change differed between the two macaque species. These findings suggest that the differential clinical presentation of EVD in these two species is mediated by altered transcriptional responses.


Subject(s)
Gene Expression Regulation/immunology , Hemorrhagic Fever, Ebola/veterinary , Macaca fascicularis , Macaca mulatta , Monkey Diseases/immunology , Transcription, Genetic/immunology , Animals , COVID-19 , Ebolavirus , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/mortality , Humans , Immunity , Monkey Diseases/genetics , Monkey Diseases/mortality , RNA, Viral/metabolism , SARS-CoV-2 , Species Specificity
13.
PLoS Negl Trop Dis ; 15(3): e0009300, 2021 03.
Article in English | MEDLINE | ID: mdl-33780452

ABSTRACT

Ebola virus has been responsible for two major epidemics over the last several years and there has been a strong effort to find potential treatments that can improve the disease outcome. Antiviral favipiravir was thus tested on non-human primates infected with Ebola virus. Half of the treated animals survived the Ebola virus challenge, whereas the infection was fully lethal for the untreated ones. Moreover, the treated animals that did not survive died later than the controls. We evaluated the hematological, virological, biochemical, and immunological parameters of the animals and performed proteomic analysis at various timepoints of the disease. The viral load strongly correlated with dysregulation of the biological functions involved in pathogenesis, notably the inflammatory response, hemostatic functions, and response to stress. Thus, the management of viral replication in Ebola virus disease is of crucial importance in preventing the immunopathogenic disorders and septic-like shock syndrome generally observed in Ebola virus-infected patients.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Cytokine Release Syndrome/prevention & control , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Pyrazines/pharmacology , Viral Load/drug effects , Animals , Cytokines/blood , Disease Models, Animal , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/veterinary , Macaca fascicularis , T-Lymphocytes/immunology , Viremia/blood , Viremia/pathology , Virus Replication/drug effects
14.
Transbound Emerg Dis ; 68(3): 1521-1530, 2021 May.
Article in English | MEDLINE | ID: mdl-32915496

ABSTRACT

In 2008, an outbreak of Reston ebolavirus (RESTV) in pigs in the Philippines expanded our understanding of the host range of ebolaviruses. Subsequent experimental infections with the human-pathogenic species Zaire ebolavirus (EBOV) confirmed that pigs are susceptible to African species of ebolaviruses. Pig keeping has become an increasingly important livelihood strategy throughout parts of sub-Saharan Africa, driven by increasing demand for pork. The growth in pig keeping is particularly rapid in Uganda, which has the highest per capita pork consumption in East Africa and a history of sporadic human outbreaks of Ebola virus disease (EVD). Using a systematic sampling protocol, we collected sera from 658 pigs presented for slaughter in Uganda between December 2015 and October 2016. Forty-six pigs (7%) were seropositive based on ELISA tests at two different institutions. Seropositive pigs had antibodies that bound to Sudan NP (n = 27), Zaire NP (Kikwit; n = 8) or both NPs (n = 11). Sera from 4 of the ELISA-positive pigs reacted in Western blot (EBOV NP = 1; RESTV NP = 2; both NPs = 2), and one sample had full neutralizing antibody against Sudan ebolavirus (SUDV) in virus neutralization tests. Pigs sampled in June 2016 were significantly more likely to be seropositive than pigs sampled in October 2016 (p = .03). Seropositive pigs were sourced from all regions except Western region. These observed temporal and spatial variations are suggestive of multiple introductions of ebolaviruses into the pig population in Uganda. This is the first report of exposure of pigs in Uganda to ebolaviruses and the first to employ systematic abattoir sampling for ebolavirus surveillance during a non-outbreak period. Future studies will be necessary to further define the role pigs play (if any) in ebolavirus maintenance and transmission so that potential risks can be mitigated.


Subject(s)
Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/veterinary , Swine Diseases/epidemiology , Abattoirs , Animals , Female , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Male , Prevalence , Risk Factors , Seroepidemiologic Studies , Spatio-Temporal Analysis , Sus scrofa , Swine , Swine Diseases/virology , Uganda/epidemiology
15.
Viruses ; 12(12)2020 11 24.
Article in English | MEDLINE | ID: mdl-33255243

ABSTRACT

Based on a large study conducted on wild great ape fecal samples collected in regions of Gabon where previous human outbreaks of Ebola virus disease have occurred between 1994 and 2002, we provide evidence for prevalence of Zaire ebolavirus (EBOV)-specific antibodies of 3.9% (immunoglobulin G (IgG)) and 3.5% (immunoglobulin M (IgM)) in chimpanzees and 8.8% (IgG) and 2.4% (IgM) in gorillas. Importantly, we observed a high local prevalence (31.2%) of anti-EBOV IgG antibodies in gorilla samples. This high local rate of positivity among wild great apes raises the question of a spatially and temporally localized increase in EBOV exposure risk and the role that can be played by these animals as sentinels of the virus's spread or reemergence in a given area.


Subject(s)
Ape Diseases/immunology , Ape Diseases/virology , Ebolavirus , Gorilla gorilla/immunology , Gorilla gorilla/virology , Hemorrhagic Fever, Ebola/veterinary , Animals , Antibodies, Viral , Ape Diseases/diagnosis , Ape Diseases/epidemiology , Feces/virology , Gabon/epidemiology , Geography , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Pan troglodytes/immunology , RNA, Viral , Sequence Analysis, DNA
16.
Rev Sci Tech ; 39(2): 579-590, 2020 Aug.
Article in English | MEDLINE | ID: mdl-33046918

ABSTRACT

All-hazards preparedness and response planning requires ongoing individual, organisational and multi-jurisdictional learning. Disaster after-action reviews are an established emergency management practice to acquire knowledge through a process of analysing what happened and why, to improve the emergency response before the next crisis. After-action reviews help individuals and organisations learn, and are an essential step in the preparedness cycle. Human and animal health authorities have begun to employ after-action reviews for disaster preparedness and response among public health and Veterinary Services. The World Organisation for Animal Health (OIE) encourages Members to establish after-action reviews and share best practice. The adoption of afteraction review is an essential step for all provincial, national and multinational emergency management authorities to mitigate the impact of disasters on human and animal health. Emerging and re-emerging infectious diseases with pandemic potential pose unique preparedness challenges, requiring high-level policy attention to close long-standing gaps. A review of after-action reports from the 2001 anthrax bioterror attacks and of naturally occurring infectious disease crises, from the 2003 outbreak of severe acute respiratory syndrome (SARS) to the 2014 Ebola epidemic, reveal a similar pattern of repeated weakness and failures. These phenomena are described as 'lessons observed but not lessons learned'. Most infectious disease outbreaks with pandemic potential are zoonotic and require a One Health approach to prevent, prepare for and respond to global health security crises. After-action reviews in a One Health security context are essential to improve the pandemic preparedness of public health and Veterinary Services. After-action reviews can also provide the evidence-based 'feedback loop' needed to galvanise public policy and political will to translate lessons observed into sustained and applied lessons learned.


La planification de la préparation et de la réponse à tous les risques est un processus qui exige un apprentissage permanent tant à l'échelle des individus que des organisations et des différentes autorités compétentes. Les retours d'expérience (ou « revues après action¼) suite à une catastrophe constituent un exercice éprouvé de gestion des urgences visant à acquérir de nouvelles connaissances en procédant à l'analyse de ce qui est arrivé et des raisons pour lesquelles c'est arrivé, dans le but d'améliorer les capacités d'intervention d'urgence avant que ne survienne la prochaine crise. Les individus et les organisations trouvent dans ces retours un cadre pour tirer des enseignements de leur expérience, ce qui constitue une étape essentielle du cycle de préparation. Les autorités en charge de la santé humaine et de la santé animale ont commencé à utiliser les retours d'expérience pour planifier la préparation et la réponse au sein des Services de santé publique et des Services vétérinaires. L'Organisation mondiale de la santé animale (OIE) encourage ses Membres à mettre en place des retours d'expérience et à partager les meilleures pratiques en la matière. L'analyse des retours d'expérience est une étape cruciale pour que les autorités en charge de la gestion des urgences à l'échelle provinciale, nationale et internationale puissent atténuer l'impact des catastrophes sur la santé humaine et animale. Les maladies émergentes et ré-émergentes ayant un potentiel pandémique posent des défis exceptionnels en termes de préparation et exigent des prises de décision de haut niveau afin de pallier des lacunes souvent anciennes. L'examen des retours d'expérience datant des attentats terroristes à l'anthrax de 2001 et des crises sanitaires dues à des maladies infectieuses d'origine naturelle (depuis l'épidémie du syndrome respiratoire aigu sévère [SRAS] en 2003 jusqu'à l'épidémie d'Ebola en 2014) révèle des caractéristiques toujours similaires, avec à chaque fois les mêmes faiblesses et les mêmes écueils. Ce phénomène correspond à ce que l'on peut appeler des « leçons observées mais non apprises ¼. Compte tenu de la nature zoonotique de la plupart des foyers de maladies infectieuses ayant un potentiel pandémique, c'est l'approche Une seule santé qui doit prévaloir en matière de prévention, de préparation et de réponse aux crises de sécurité sanitaire d'envergure mondiale. Les retours d'expérience dans un contexte de sécurité Une seule santé sont essentiels pour améliorer la préparation des Services de santé publique et des Services vétérinaires aux pandémies. En outre, les « boucles de réaction¼ fondées sur des éléments factuels résultant des retours d'expérience apportent un éclairage indispensable pour inciter les pouvoirs publics à élaborer des mesures appropriées et pour créer la volonté politique de traduire les leçons observées en leçons durablement apprises et appliquées.


La planificación de las labores de preparación y respuesta ante toda clase de peligros exige un permanente aprendizaje tanto personal como institucional y desde múltiples competencias. El examen de las intervenciones tras un desastre constituye un arraigado proceder de gestión de emergencias que sirve para aprender de la experiencia analizando el cómo y el porqué de lo sucedido y, a partir de ahí, mejorar los procesos de respuesta de emergencia antes de que advenga la siguiente crisis. Estos exámenes posteriores a las intervenciones, que ayudan a las personas y organizaciones a aprender, son una etapa fundamental del ciclo de preparación. Las autoridades sanitarias y zoosanitarias han empezado a utilizarlos en los servicios de salud pública y los Servicios Veterinarios con fines de preparación y respuesta para casos de desastre. La Organización Mundial de Sanidad Animal (OIE) alienta a sus Miembros a que establezcan este tipo de exámenes y pongan en común prácticas óptimas al respecto. La implantación del examen posterior a las intervenciones es un paso esencial para que todas las autoridades provinciales, nacionales e internacionales de gestión de emergencias estén en condiciones de mitigar los efectos sanitarios y zoosanitarios de un desastre. Las enfermedades infecciosas emergentes y reemergentes con potencial pandémico plantean singulares problemas de preparación, que requieren una atención y una labor normativa de alto nivel para solventar carencias históricas. El examen de los informes posteriores a las actuaciones de respuesta a los ataques bioterroristas perpetrados en 2001 con bacterias de carbunco (ántrax) y a crisis infecciosas de origen natural, desde el brote registrado en 2003 de síndrome respiratorio agudo severo (SRAS) hasta la epidemia causada por el virus del Ébola en 2014, revela un parecido patrón de fallos y carencias que se van repitiendo, fenómeno que se describe como «hechos observados sin enseñanzas extraídas¼. La mayoría de los brotes de enfermedades infecciosas con potencial pandémico son zoonóticos y exigen la aplicación de la lógica de Una sola salud para prevenir crisis sanitarias de dimensión mundial, prepararse para ellas y darles respuesta. Los exámenes posteriores a una intervención inscritos en el contexto de seguridad sanitaria de Una sola salud son esenciales para mejorar la preparación de los servicios de salud pública y los Servicios Veterinarios para episodios de pandemia. Estos procesos de examen también pueden alimentar con datos científicos el «ciclo de retroalimentación¼ que se requiere para galvanizar las políticas públicas y la voluntad política de traducir los hechos observados en enseñanzas extraídas que sean duraderas y se apliquen en la práctica.


Subject(s)
Disasters , Hemorrhagic Fever, Ebola , Animals , Disease Outbreaks/prevention & control , Disease Outbreaks/veterinary , Global Health , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/veterinary , Humans , Public Health
17.
J Virol ; 94(11)2020 05 18.
Article in English | MEDLINE | ID: mdl-32213610

ABSTRACT

Ebola virus (EBOV) continues to pose a significant threat to human health, as evidenced by the 2013-2016 epidemic in West Africa and the ongoing outbreak in the Democratic Republic of the Congo. EBOV causes hemorrhagic fever, organ damage, and shock culminating in death, with case fatality rates as high as 90%. This high lethality combined with the paucity of licensed medical countermeasures makes EBOV a critical human pathogen. Although EBOV infection results in significant damage to the liver and the adrenal glands, little is known about the molecular signatures of injury in these organs. Moreover, while changes in peripheral blood cells are becoming increasingly understood, the host responses within organs and lymphoid tissues remain poorly characterized. To address this knowledge gap, we tracked longitudinal transcriptional changes in tissues collected from EBOV-Makona-infected cynomolgus macaques. Following infection, both liver and adrenal glands exhibited significant and early downregulation of genes involved in metabolism, coagulation, hormone synthesis, and angiogenesis; upregulated genes were associated with inflammation. Analysis of lymphoid tissues showed early upregulation of genes that play a role in innate immunity and inflammation and downregulation of genes associated with cell cycle and adaptive immunity. Moreover, transient activation of innate immune responses and downregulation of humoral immune responses in lymphoid tissues were confirmed with flow cytometry. Together, these data suggest that the liver, adrenal gland, and lymphatic organs are important sites of EBOV infection and that dysregulating the function of these vital organs contributes to the development of Ebola virus disease.IMPORTANCE Ebola virus (EBOV) remains a high-priority pathogen since it continues to cause outbreaks with high case fatality rates. Although it is well established that EBOV results in severe organ damage, our understanding of tissue injury in the liver, adrenal glands, and lymphoid tissues remains limited. We begin to address this knowledge gap by conducting longitudinal gene expression studies in these tissues, which were collected from EBOV-infected cynomolgus macaques. We report robust and early gene expression changes within these tissues, indicating they are primary sites of EBOV infection. Furthermore, genes involved in metabolism, coagulation, and adaptive immunity were downregulated, while inflammation-related genes were upregulated. These results indicate significant tissue damage consistent with the development of hemorrhagic fever and lymphopenia. Our study provides novel insight into EBOV-host interactions and elucidates how host responses within the liver, adrenal glands, and lymphoid tissues contribute to EBOV pathogenesis.


Subject(s)
Adrenal Glands , Ebolavirus , Gene Expression Regulation, Viral/immunology , Hemorrhagic Fever, Ebola , Liver , Lymphoid Tissue , Monkey Diseases , Transcription, Genetic/immunology , Adrenal Glands/immunology , Adrenal Glands/metabolism , Adrenal Glands/pathology , Adrenal Glands/virology , Animals , Ebolavirus/immunology , Ebolavirus/metabolism , Female , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/veterinary , Liver/immunology , Liver/metabolism , Liver/pathology , Liver/virology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Macaca fascicularis , Male , Monkey Diseases/immunology , Monkey Diseases/metabolism , Monkey Diseases/pathology , Monkey Diseases/virology
18.
Emerg Infect Dis ; 26(4): 760-763, 2020 04.
Article in English | MEDLINE | ID: mdl-32186496

ABSTRACT

Ebola virus (EBOV) is a highly pathogenic zoonotic virus for which the reservoir host has not been identified. To study the role of dogs as potential hosts, we screened 300 serum samples from dogs in Sierra Leone and found EBOV neutralizing antibodies in 12, suggesting their susceptibility to natural infection.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Antibodies, Neutralizing , Disease Outbreaks , Dogs , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/veterinary , Sierra Leone/epidemiology
19.
PLoS Negl Trop Dis ; 14(3): e0008117, 2020 03.
Article in English | MEDLINE | ID: mdl-32130210

ABSTRACT

Ebolaviruses pose a substantial threat to wildlife populations and to public health in Africa. Evolutionary analyses of virus genome sequences can contribute significantly to elucidate the origin of new outbreaks, which can help guide surveillance efforts. The reconstructed between-outbreak evolutionary history of Zaire ebolavirus so far has been highly consistent. By removing the confounding impact of population growth bursts during local outbreaks on the free mixing assumption that underlies coalescent-based demographic reconstructions, we find-contrary to what previous results indicated-that the circulation dynamics of Ebola virus in its animal reservoir are highly uncertain. Our findings also accentuate the need for a more fine-grained picture of the Ebola virus diversity in its reservoir to reliably infer the reservoir origin of outbreak lineages. In addition, the recent appearance of slower-evolving variants is in line with latency as a survival mechanism and with bats as the natural reservoir host.


Subject(s)
Animal Diseases/epidemiology , Chiroptera/virology , Disease Reservoirs/virology , Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/veterinary , Africa , Animal Diseases/virology , Animals , Ebolavirus/classification , Ebolavirus/genetics , Genotype , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Humans , Phylogeny
20.
Transbound Emerg Dis ; 67(2): 724-732, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31627257

ABSTRACT

The genus Ebolavirus comprises several virus species with zoonotic potential and varying pathogenicity for humans. Ebolaviruses are considered to circulate in wildlife with occasional spillover events into the human population which then often leads to severe disease outbreaks. Several studies indicate a significant role of bats as reservoir hosts in the ebolavirus ecology. However, pigs from the Philippines have been found to be naturally infected with Reston virus (RESTV), an ebolavirus that is thought to only cause asymptomatic infections in humans. The recent report of ebolavirus-specific antibodies in pigs from Sierra Leone further supports natural infection of pigs with ebolaviruses. However, susceptibility of pigs to highly pathogenic Ebola virus (EBOV) was only shown under experimental settings and evidence for natural infection of pigs with EBOV is currently lacking. Between October and December 2017, we collected 308 serum samples from pigs in Guinea, West Africa, and tested for the presence of ebolavirus-specific antibodies with different serological assays. Besides reactivity to EBOV nucleoproteins in ELISA and Western blot for 19 (6.2%) and 13 (4.2%) samples, respectively, four sera recognized Sudan virus (SUDV) NP in Western blot. Furthermore, four samples specifically detected EBOV or SUDV glycoprotein (GP) in an indirect immunofluorescence assay under native conditions. Virus neutralization assay based on EBOV (Mayinga isolate) revealed five weakly neutralizing sera. The finding of (cross-) reactive and weakly neutralizing antibodies suggests the exposure of pigs from Guinea to ebolaviruses or ebola-like viruses with their pathogenicity as well as their zoonotic potential remaining unknown. Future studies should investigate whether pigs can act as an amplifying host for ebolaviruses and whether there is a risk for spillover events.


Subject(s)
Antibodies, Viral/blood , Disease Outbreaks/veterinary , Disease Reservoirs/veterinary , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/veterinary , Immunoglobulin G/blood , Swine Diseases/epidemiology , Animals , Antibodies, Neutralizing/blood , Cross Reactions , Ebolavirus/isolation & purification , Enzyme-Linked Immunosorbent Assay/veterinary , Farms , Female , Guinea/epidemiology , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Humans , Male , Nucleoproteins/immunology , Sus scrofa , Swine , Swine Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...