Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Zoonoses Public Health ; 71(1): 1-17, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37933425

RESUMO

The attainment of the global target of zero dog-mediated human rabies by 2030 depends on functional rabies programmes. Nigeria, a rabies-endemic country, and the most populous country in Africa has a very poor rabies control strategy with a score of 1.5 out of 5 based on the Stepwise Approach towards Rabies Elimination (SARE). In this article, we report a scoping review that we conducted to highlight the strengths, weaknesses, opportunities and threats as well as situational analysis of rabies control in Nigeria and suggest a timeline for key activities that are needed to ensure zero by 30. Our findings reveal that rabies is grossly under-reported as only 998 human and 273 dog-suspected rabies cases were reported across Nigeria between 2017 and 2022. Our literature review also demonstrates a paucity of information on rabies in both human and animal health sectors. A total of 49 studies on dog rabies in Nigeria, with a predominance of reports from the North Central geopolitical region (48%, n = 23) were therefore included in this study. Currently, only 16.2% (n = 6/37) of Nigerian states have available data related to the estimated dog populations, the dog ownership rates, the vaccination status of dogs or the incidence of dog bites. Based on a dog-to-human ratio of 1:16.3, we estimated that the dog population in Nigeria was 12,969,368 (95% CI: 12,320,900-13,617,836). Thus, to attain herd immunity and dog rabies control in Nigeria, at least 9.1 million dogs must be vaccinated annually. Our review reveals that, despite the strengths and available opportunities to achieve rabies control in Nigeria by 2030, the weaknesses and challenges will make the attainment of zero by 30 very difficult or impossible. Nigeria's best-case scenario by the year 2030 is SARE stage 3-4 (control-elimination) out of 5. Otherwise, the rabies control programme might not surpass SARE stages 2-3. To attain zero by 30, Nigeria must re-strategize its current rabies control programme by funding and implementing the national strategic plan for rabies control, creating a rabies desk office in the 37 states (FCT inclusive), rigorously conducting mass vaccination campaigns, providing post-exposure prophylaxis, prioritizing mass enlightenment with a focus on responsible pet ownership and conduct baseline national rabies surveillance in the animal and human health sectors.


Assuntos
Mordeduras e Picadas , Doenças do Cão , Vacina Antirrábica , Raiva , Animais , Humanos , Cães , Raiva/epidemiologia , Raiva/prevenção & controle , Raiva/veterinária , Nigéria/epidemiologia , Doenças do Cão/epidemiologia , Doenças do Cão/prevenção & controle , Profilaxia Pós-Exposição , Mordeduras e Picadas/epidemiologia , Mordeduras e Picadas/prevenção & controle , Mordeduras e Picadas/veterinária
2.
Med Sci (Paris) ; 39(12): 945-952, 2023 Dec.
Artigo em Francês | MEDLINE | ID: mdl-38108725

RESUMO

In recent decades, bats have been associated with numerous viral pandemics. Bats harbor a large variety of viruses, some of which have a high zoonotic potential for humans. While infection with these viruses can be fatal in other mammals, bats are often infected asymptomatically. It is hypothesized that a balanced immune response would enable them to maintain homeostasis during infection, thus limiting viral replication while avoiding the impact of excessive inflammation. Deciphering these mechanisms, using adapted in vitro models, will help assess and avoid the potential zoonotic risk of these animals, while paving the way for the development of therapeutics for infectious and inflammatory diseases.


Title: Des chauves-souris et des virus - Entre contrôle de l'infection et tolérance immunitaire. Abstract: Durant les dernières décennies, les chauves-souris ont été associées à de nombreuses pandémies virales. Ces animaux hébergent en effet une diversité importante de virus, certains à potentiel zoonotique pour l'homme. Alors que ces virus peuvent être mortels chez d'autres mammifères, les chauves-souris sont souvent infectées de façon asymptomatique. La mise en place d'une réponse immunitaire équilibrée leur permettrait de maintenir l'homéostasie lors de l'infection, en limitant la réplication virale tout en évitant l'impact d'une inflammation trop importante. Le décryptage de ces mécanismes, à l'aide de modèles in vitro adaptés, devrait contribuer à évaluer et à éviter le risque zoonotique potentiel de ces animaux, tout en ouvrant la voie au développement de thérapeutiques pour les maladies infectieuses et inflammatoires.


Assuntos
Quirópteros , Vírus , Animais , Humanos , Tolerância Imunológica , Homeostase , Controle de Infecções
3.
Mol Ecol ; 32(18): 5140-5155, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37540190

RESUMO

In epidemiology, endemicity characterizes sustained pathogen circulation in a geographical area, which involves a circulation that is not being maintained by external introductions. Because it could potentially shape the design of public health interventions, there is an interest in fully uncovering the endemic pattern of a disease. Here, we use a phylogeographic approach to investigate the endemic signature of rabies virus (RABV) circulation in Cambodia. Cambodia is located in one of the most affected regions by rabies in the world, but RABV circulation between and within Southeast Asian countries remains understudied. Our analyses are based on a new comprehensive data set of 199 RABV genomes collected between 2014 and 2017 as well as previously published Southeast Asian RABV sequences. We show that most Cambodian sequences belong to a distinct clade that has been circulating almost exclusively in Cambodia. Our results thus point towards rabies circulation in Cambodia that does not rely on external introductions. We further characterize within-Cambodia RABV circulation by estimating lineage dispersal metrics that appear to be similar to other settings, and by performing landscape phylogeographic analyses to investigate environmental factors impacting the dispersal dynamic of viral lineages. The latter analyses do not lead to the identification of environmental variables that would be associated with the heterogeneity of viral lineage dispersal velocities, which calls for a better understanding of local dog ecology and further investigations of the potential drivers of RABV spread in the region. Overall, our study illustrates how phylogeographic investigations can be performed to assess and characterize viral endemicity in a context of relatively limited data.


Assuntos
Vírus da Raiva , Raiva , Animais , Cães , Raiva/epidemiologia , Raiva/veterinária , Camboja/epidemiologia , Vírus da Raiva/genética , Filogeografia , Análise de Sequência de DNA , Filogenia
4.
BMC Immunol ; 24(1): 7, 2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-37085747

RESUMO

BACKGROUND: Coevolution between pathogens and their hosts decreases host morbidity and mortality. Bats host and can tolerate viruses which can be lethal to other vertebrate orders, including humans. Bat adaptations to infection include localized immune response, early pathogen sensing, high interferon expression without pathogen stimulation, and regulated inflammatory response. The immune reaction is costly, and bats suppress high-cost metabolism during torpor. In the temperate zone, bats hibernate in winter, utilizing a specific behavioural adaptation to survive detrimental environmental conditions and lack of energy resources. Hibernation torpor involves major physiological changes that pose an additional challenge to bat-pathogen coexistence. Here, we compared bat cellular reaction to viral challenge under conditions simulating hibernation, evaluating the changes between torpor and euthermia. RESULTS: We infected the olfactory nerve-derived cell culture of Myotis myotis with an endemic bat pathogen, European bat lyssavirus 1 (EBLV-1). After infection, the bat cells were cultivated at two different temperatures, 37 °C and 5 °C, to examine the cell response during conditions simulating euthermia and torpor, respectively. The mRNA isolated from the cells was sequenced and analysed for differential gene expression attributable to the temperature and/or infection treatment. In conditions simulating euthermia, infected bat cells produce an excess signalling by multitude of pathways involved in apoptosis and immune regulation influencing proliferation of regulatory cell types which can, in synergy with other produced cytokines, contribute to viral tolerance. We found no up- or down-regulated genes expressed in infected cells cultivated at conditions simulating torpor compared to non-infected cells cultivated under the same conditions. When studying the reaction of uninfected cells to the temperature treatment, bat cells show an increased production of heat shock proteins (HSPs) with chaperone activity, improving the bat's ability to repair molecular structures damaged due to the stress related to the temperature change. CONCLUSIONS: The lack of bat cell reaction to infection in conditions simulating hibernation may contribute to the virus tolerance or persistence in bats. Together with the cell damage repair mechanisms induced in response to hibernation, the immune regulation may promote bats' ability to act as reservoirs of zoonotic viruses such as lyssaviruses.


Assuntos
Quirópteros , Hibernação , Lyssavirus , Vírus , Animais , Quirópteros/fisiologia , Transcriptoma
5.
Arch Virol ; 167(12): 2857-2906, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36437428

RESUMO

In March 2022, following the annual International Committee on Taxonomy of Viruses (ICTV) ratification vote on newly proposed taxa, the phylum Negarnaviricota was amended and emended. The phylum was expanded by two new families (bunyaviral Discoviridae and Tulasviridae), 41 new genera, and 98 new species. Three hundred forty-nine species were renamed and/or moved. The accidentally misspelled names of seven species were corrected. This article presents the updated taxonomy of Negarnaviricota as now accepted by the ICTV.


Assuntos
Mononegavirais , Vírus , Humanos , Mononegavirais/genética , Filogenia
6.
J Virol ; 96(14): e0060822, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35862713

RESUMO

Bats are natural reservoirs of numerous coronaviruses, including the potential ancestor of SARS-CoV-2. Knowledge concerning the interaction between coronaviruses and bat cells is sparse. We investigated the ability of primary cells from Rhinolophus and Myotis species, as well as of established and novel cell lines from Myotis myotis, Eptesicus serotinus, Tadarida brasiliensis, and Nyctalus noctula, to support SARS-CoV-2 replication. None of these cells were permissive to infection, not even the ones expressing detectable levels of angiotensin-converting enzyme 2 (ACE2), which serves as the viral receptor in many mammalian species. The resistance to infection was overcome by expression of human ACE2 (hACE2) in three cell lines, suggesting that the restriction to viral replication was due to a low expression of bat ACE2 (bACE2) or the absence of bACE2 binding in these cells. Infectious virions were produced but not released from hACE2-transduced M. myotis brain cells. E. serotinus brain cells and M. myotis nasal epithelial cells expressing hACE2 efficiently controlled viral replication, which correlated with a potent interferon response. Our data highlight the existence of species-specific and cell-specific molecular barriers to viral replication in bat cells. These novel chiropteran cellular models are valuable tools to investigate the evolutionary relationships between bats and coronaviruses. IMPORTANCE Bats are host ancestors of several viruses that cause serious disease in humans, as illustrated by the ongoing SARS-CoV-2 pandemic. Progress in investigating bat-virus interactions has been hampered by a limited number of available bat cellular models. We have generated primary cells and cell lines from several bat species that are relevant for coronavirus research. The various permissivities of the cells to SARS-CoV-2 infection offered the opportunity to uncover some species-specific molecular restrictions to viral replication. All bat cells exhibited a potent entry-dependent restriction. Once this block was overcome by overexpression of human ACE2, which serves at the viral receptor, two bat cell lines controlled well viral replication, which correlated with the inability of the virus to counteract antiviral responses. Other cells potently inhibited viral release. Our novel bat cellular models contribute to a better understanding of the molecular interplays between bat cells and viruses.


Assuntos
Quirópteros , SARS-CoV-2 , Replicação Viral , Enzima de Conversão de Angiotensina 2/genética , Animais , Quirópteros/virologia , Humanos , Receptores Virais/metabolismo , SARS-CoV-2/fisiologia , Especificidade da Espécie , Glicoproteína da Espícula de Coronavírus/metabolismo
7.
Animals (Basel) ; 12(11)2022 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-35681827

RESUMO

The Rhabdoviridae is a large family of negative-sense (-) RNA viruses that includes important pathogens of ray-finned fish and marine mammals. As for all viruses, the taxonomic assignment of rhabdoviruses occurs through a process implemented by the International Committee on Taxonomy of Viruses (ICTV). A recent revision of taxonomy conducted in conjunction with the ICTV Rhabdoviridae Study Group has resulted in the establishment of three new subfamilies (Alpharhabdovirinae, Betarhabdovirinae, and Gammarhabdovirinae) within the Rhabdoviridae, as well as three new genera (Cetarhavirus, Siniperhavirus, and Scophrhavirus) and seven new species for viruses infecting fish or marine mammals. All rhabdovirus species have also now been named or renamed to comply with the binomial format adopted by the ICTV in 2021, comprising the genus name followed by a species epithet. Phylogenetic analyses of L protein (RNA-dependent RNA polymerase) sequences of (-) RNA viruses indicate that members of the genus Novirhabdovirus (subfamily Gammarhabdovirinae) do not cluster within the Rhabdoviridae, suggesting the need for a review of their current classification.

8.
Microbiol Resour Announc ; 11(1): e0104721, 2022 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-35049353

RESUMO

As in other African countries, canine rabies is endemic in Liberia. However, data concerning the genetic diversity of rabies virus isolates circulating in this country remain limited. We report here the complete genome sequences of five rabies viruses obtained from domestic animals. All of them belonged to subgroup H within the Africa 2 clade.

9.
Microbiol Resour Announc ; 11(1): e0110921, 2022 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-34989606

RESUMO

In this report, we describe eight nearly complete genome sequences of rabies virus strains collected in the Democratic Republic of the Congo from domestic carnivores in 2017 and 2018. All of them clustered into a specific phylogroup among the Africa 1b lineage in the Cosmopolitan clade.

10.
Clin Infect Dis ; 74(3): 461-466, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33991184

RESUMO

BACKGROUND: Inaccurate diagnosis of encephalitis is a major issue as immunosuppressive treatments can be deleterious in case of viral infection. The European bat lyssavirus type 1 (EBLV-1), a virus related to rabies virus, is endemic in European bats. No human case has yet been reported in Western Europe. A 59-year-old patient without specific past medical history died from encephalitis. A colony of bats lived in an outbuilding of his house. No diagnosis was made using standard procedures. METHODS: We used a next generation sequencing (NGS) based transcriptomic protocol to search for pathogens in autopsy samples (meninges and brain frontal lobe). Results were confirmed by polymerase chain reaction (PCR) and by antibody testing in serum. Immunochemistry was used to characterize inflammatory cells and viral antigens in brain lesions. Cells and mice were inoculated with brain extracts for virus isolation. RESULTS: The patient's brain lesions were severe and diffuse in white and gray matter. Perivascular inflammatory infiltrates were abundant and rich in plasma cells. NGS identified European bat lyssavirus type 1a in brain, which was confirmed by PCR. A high titer of neutralizing antibodies was found in serum. No viral antigen was detected, and the virus could not be isolated by cell culture or by mouse inoculation. CONCLUSIONS: The patient died from European bat lyssavirus type 1a infection. NGS was key to identifying this unexpected viral etiology in an epidemiological context that did not suggest rabies. People exposed to bats should be strongly advised to be vaccinated with rabies vaccines, which are effective against EBLV-1.


Assuntos
Quirópteros , Encefalite , Lyssavirus , Raiva , Infecções por Rhabdoviridae , Animais , Europa (Continente)/epidemiologia , Humanos , Lyssavirus/genética , Camundongos , Raiva/diagnóstico , Raiva/veterinária , Infecções por Rhabdoviridae/diagnóstico , Infecções por Rhabdoviridae/epidemiologia , Infecções por Rhabdoviridae/veterinária
11.
Viruses ; 13(11)2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34834974

RESUMO

Rhabdoviridae is the most diverse family of the negative, single-stranded RNA viruses, which includes 40 ecologically different genera that infect plants, insects, reptiles, fishes, and mammals, including humans, and birds. To date, only a few bird-related rhabdoviruses among the genera Sunrhavirus, Hapavirus, and Tupavirus have been described and analyzed at the molecular level. In this study, we characterized seven additional and previously unclassified rhabdoviruses, which were isolated from various bird species collected in Africa during the 1960s and 1970s. Based on the analysis of their genome sequences obtained by next generation sequencing, we observed a classical genomic structure, with the presence of the five canonical rhabdovirus genes, i.e., nucleoprotein (N), phosphoprotein (P), matrix protein (M), glycoprotein (G), and polymerase (L). In addition, different additional open reading frames which code putative proteins of unknown function were identified, with the common presence of the C and the SH proteins, within the P gene and between the M and G genes, respectively. Genetic comparisons and phylogenetic analysis demonstrated that these seven bird-related rhabdoviruses could be considered as putative new species within the genus Sunrhavirus, where they clustered into a single group (named Clade III), a companion to two other groups that encompass mainly insect-related viruses. The results of this study shed light on the high diversity of the rhabdoviruses circulating in birds, mainly in Africa. Their close relationship with other insect-related sunrhaviruses raise questions about their potential role and impact as arboviruses that affect bird communities.


Assuntos
Aves/virologia , Genoma Viral , Infecções por Rhabdoviridae/veterinária , Infecções por Rhabdoviridae/virologia , Rhabdoviridae/classificação , Rhabdoviridae/genética , África , Animais , Sequência de Bases , Variação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Fases de Leitura Aberta , Filogenia , RNA Viral/genética , Análise de Sequência , Análise de Sequência de DNA , Proteínas Virais/genética
12.
Front Cell Infect Microbiol ; 11: 761074, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34804996

RESUMO

Rabies virus (RABV), the causative agent for rabies disease is still presenting a major public health concern causing approximately 60,000 deaths annually. This neurotropic virus (genus Lyssavirus, family Rhabdoviridae) induces an acute and almost always fatal form of encephalomyelitis in humans. Despite the lethal consequences associated with clinical symptoms of rabies, RABV limits neuro-inflammation without causing major histopathological lesions in humans. Nevertheless, information about the mechanisms of infection and cellular response in the central nervous system (CNS) remain scarce. Here, we investigated the expression of inflammatory genes involved in immune response to RABV (dog-adapted strain Tha) in mice, the most common animal model used to study rabies. To better elucidate the pathophysiological mechanisms during natural RABV infection, we compared the inflammatory transcriptome profile observed at the late stage of infection in the mouse brain (cortex and brain stem/cerebellum) with the ortholog gene expression in post-mortem brain biopsies of rabid patients. Our data indicate that the inflammatory response associated with rabies is more pronounced in the murine brain compared to the human brain. In contrast to murine transcription profiles, we identified CXC motif chemokine ligand 16 (CXCL16) as the only significant differentially expressed gene in post-mortem brains of rabid patients. This result was confirmed in vitro, in which Tha suppressed interferon alpha (IFN-α)-induced CXCL16 expression in human CNS cell lines but induced CXCL16 expression in IFN-α-stimulated murine astrocytes. We hypothesize that RABV-induced modulation of the CXCL16 pathway in the brain possibly affects neurotransmission, natural killer (NK) and T cell recruitment and activation. Overall, we show species-specific differences in the inflammatory response of the brain, highlighted the importance of understanding the potential limitations of extrapolating data from animal models to humans.


Assuntos
Vírus da Raiva , Raiva , Animais , Encéfalo , Quimiocina CXCL16 , Modelos Animais de Doenças , Cães , Humanos , Camundongos , Vírus da Raiva/genética , Transcriptoma
13.
Pathogens ; 10(10)2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34684205

RESUMO

A perhabdovirus was isolated from a mortality episode affecting a fish farm in 2019 in Western Europe. This virus was produced in cell culture and was readily detected by a species-specific real-time PCR assay. The near-complete sequence of the virus obtained showed some relatedness with viruses of the species Perhabdovirus perca. However, it was distinct enough from these viruses to form a separate genetic lineage. Multiple substitutions along the genome caused non-detection using a range of conventional PCRs previously shown to target four known genogroups of perhabdoviruses. However, various generic PCRs efficiently detected the isolated virus. The origin of this virus remains to be elucidated. It may have been introduced into the farm via wild genitors. This finding provides new evidence of the high genetic diversity of percid perhabdoviruses and the potential of new genotypes to emerge as threats for fish farming. Efforts to improve the existing diagnostic methods and control this large group of viruses are still needed.

14.
Vaccines (Basel) ; 9(4)2021 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-33805019

RESUMO

Pre-exposure rabies prophylaxis (PrEP) is recommended for people at frequent or increased risk of professional exposure to lyssavirus (including rabies virus). PrEP provides protection against unrecognized exposure. After the primary vaccination, one's immune response against rabies may decline over time. We aimed to evaluate the immune response to rabies in individuals immunized for occupational reasons before and after a booster dose of the rabies vaccine. With this aim, we retrospectively documented factors associated with an inadequate response in individuals vaccinated for occupational purposes. Our findings analyzed data from 498 vaccinated individuals and found that 17.2% of participants had an inadequate antibody titration documented after their primary vaccination without the booster, while inadequate response after an additional booster of the vaccine was evidenced in 0.5% of tested participants. This study showed that a single booster dose of vaccine after PrEP conferred a high and long-term immune response in nearly all individuals except for rare, low responders. A systematic rabies booster after primary vaccination may result in alleviating the monitoring strategy of post-PrEP antibody titers among exposed professionals.

15.
Acta Trop ; 216: 105787, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33385361

RESUMO

Despite declaration as a national priority disease, dog rabies remains endemic in Liberia, with surveillance systems and disease control activities still developing. The objective of these initial efforts was to establish animal rabies diagnostics, foster collaboration between all rabies control stakeholders, and develop a short-term action plan with estimated costs for rabies control and elimination in Liberia. Four rabies diagnostic tests, the direct fluorescent antibody (DFA) test, the direct immunohistochemical test (dRIT), the reverse transcriptase polymerase chain reaction (RT-PCR) assay and the rapid immunochromatographic diagnostic test (RIDT), were implemented at the Central Veterinary Laboratory (CVL) in Monrovia between July 2017 and February 2018. Seven samples (n=7) out of eight suspected animals were confirmed positive for rabies lyssavirus, and molecular analyses revealed that all isolates belonged to the Africa 2 lineage, subgroup H. During a comprehensive in-country One Health rabies stakeholder meeting in 2018, a practical workplan, a short-term action plan and an accurately costed mass dog vaccination strategy were developed. Liberia is currently at stage 1.5/5 of the Stepwise Approach towards Rabies Elimination (SARE) tool, which corresponds with countries that are scaling up local-level interventions (e.g. dog vaccination campaigns) to the national level. Overall an estimated 5.3 - 8 million USD invested over 13 years is needed to eliminate rabies in Liberia by 2030. Liberia still has a long road to become free from dog-rabies. However, the dialogue between all relevant stakeholders took place, and disease surveillance considerably improved through implementing rabies diagnosis at the CVL. The joint efforts of diverse national and international stakeholders laid important foundations to achieve the goal of zero dog-mediated human rabies deaths by 2030.


Assuntos
Testes Diagnósticos de Rotina/veterinária , Vacina Antirrábica/administração & dosagem , Raiva/diagnóstico , Raiva/prevenção & controle , Animais , DNA Viral , Testes Diagnósticos de Rotina/métodos , Doenças do Cão/diagnóstico , Doenças do Cão/prevenção & controle , Doenças do Cão/virologia , Cães/virologia , Feminino , Humanos , Libéria/epidemiologia , Masculino , Vacinação em Massa/veterinária , Técnicas de Diagnóstico Molecular/métodos , Técnicas de Diagnóstico Molecular/veterinária , Filogenia , Reação em Cadeia da Polimerase , Vacina Antirrábica/economia , Vírus da Raiva/genética , Vírus da Raiva/isolamento & purificação
16.
Viruses ; 13(1)2021 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-33466539

RESUMO

Bats, the second largest order of mammals worldwide, harbor specific characteristics such as sustaining flight, a special immune system, unique habits, and ecological niches. In addition, they are the natural reservoirs of a variety of emerging or re-emerging zoonotic pathogens. Rhabdoviridae is one of the most diverse families of RNA viruses, which consists of 20 ecologically diverse genera, infecting plants, mammals, birds, reptiles, and fish. To date, three bat-related genera are described, named Lyssavirus, Vesiculovirus, and Ledantevirus. However, the prevalence and the distribution of these bat-related rhabdoviruses remain largely unknown, especially in China. To fill this gap, we performed a large molecular retrospective study based on the real-time reverse transcription polymerase chain reaction (RT-qPCR) detection of lyssavirus in bat samples (1044 brain and 3532 saliva samples, from 63 different bat species) originating from 21 provinces of China during 2006-2018. None of them were positive for lyssavirus, but six bat brains (0.6%) of Rhinolophus bat species, originating from Hubei and Hainan provinces, were positive for vesiculoviruses or ledanteviruses. Based on complete genomes, these viruses were phylogenetically classified into three putative new species, tentatively named Yinshui bat virus (YSBV), Taiyi bat virus (TYBV), and Qiongzhong bat virus (QZBV). These results indicate the novel rhabdoviruses circulated in different Chinese bat populations.


Assuntos
Quirópteros/virologia , Genoma Viral , Filogenia , Infecções por Rhabdoviridae/veterinária , Rhabdoviridae/classificação , Animais , Encéfalo/virologia , China/epidemiologia , Estudos Retrospectivos , Rhabdoviridae/isolamento & purificação , Infecções por Rhabdoviridae/epidemiologia , Infecções por Rhabdoviridae/virologia , Saliva/virologia , Vesiculovirus/classificação
17.
Zoonoses Public Health ; 67(7): 774-784, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32770828

RESUMO

Since the elimination of dog and terrestrial wild animal rabies, bat exposures remain the only source of autochthonous lyssavirus transmission to humans in Western Europe. European bats have already been found infected with several lyssaviruses, and human-bat interactions represent a risk of viral transmission and fatal encephalitis for humans. In this study, we aim to better characterize exposures to bats in metropolitan France from 2003 to 2016 and to identify circumstances associated with exposures to lyssavirus-positive bats. Two complementary sources of data were analysed: 1/ data associated with bats responsible for human exposure received for Lyssavirus testing by the French National Reference Centre for Rabies (NRCR); and 2/ data pertaining to individuals seeking medical care through the French Anti-Rabies Clinics network after contact with a bat. From 2003 to 2016, 425 bats originating from metropolitan France were submitted to the NRCR and 16 (4%) were found positive with a lyssavirus (EBLV-1b was diagnosed in 9 bats, EBLV-1a in 6 and BBLV in one specimen). The two factors associated with bat positivity in our study were the female sex and the bat belonging to the E. serotinus species. During the same study period, 1718 individuals sought care at an Anti-Rabies Clinic after exposure to a bat resulting in an estimated incidence of human-bat interactions of 1.96 per 106 person-years. The two most frequent circumstances of exposure were handling or bites. Interactions mostly involved one adult human being and one live and non-sick-looking bat. Our study provides new insights about circumstances of human-bat interactions and may be helpful to target prevention interventions to improve the awareness of the population of the risk of lyssavirus transmission.


Assuntos
Quirópteros/virologia , Lyssavirus/genética , Lyssavirus/isolamento & purificação , Infecções por Rhabdoviridae/veterinária , Adolescente , Adulto , Animais , Cidades , Feminino , França/epidemiologia , Humanos , Masculino , Filogenia , RNA Viral/genética , RNA Viral/isolamento & purificação , Infecções por Rhabdoviridae/epidemiologia , Infecções por Rhabdoviridae/transmissão , Infecções por Rhabdoviridae/virologia , Zoonoses
18.
J Vis Exp ; (160)2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32658185

RESUMO

Functional rabies surveillance systems are crucial to provide reliable data and increase the political commitment necessary for disease control. To date, animals suspected as rabies-positive must be submitted to a postmortem confirmation using classical or molecular laboratory methods. However, most endemic areas are in low- and middle-income countries where animal rabies diagnosis is restricted to central veterinary laboratories. Poor availability of surveillance infrastructure leads to serious disease underreporting from remote areas. Several diagnostic protocols requiring low technical expertise have been recently developed, providing opportunity to establish rabies diagnosis in decentralized laboratories. We present here a complete protocol for field postmortem diagnosis of animal rabies using a rapid immunochromatographic diagnostic test (RIDT), from brain biopsy sampling to the final interpretation. We complete the protocol by describing a further use of the device for molecular analysis and viral genotyping. RIDT easily detects rabies virus and other lyssaviruses in brain samples. The principle of such tests is simple: brain material is applied on a test strip where gold conjugated antibodies bind specifically to rabies antigens. The antigen-antibody complexes bind further to fixed antibodies on the test line, resulting in a clearly visible purple line. The virus is inactivated in the test strip, but viral RNA can be subsequently extracted. This allows the test strip, rather than the infectious brain sample, to be safely and easily sent to an equipped laboratory for confirmation and molecular typing. Based on a modification of the manufacturer's protocol, we found increased test sensitivity, reaching 98% compared to the gold standard reference method, the direct immunofluorescence antibody test. The advantages of the test are numerous: rapid, easy-to-use, low cost and no requirement for laboratory infrastructure, such as microscopy or cold-chain compliance. RIDTs represent a useful alternative for areas where reference diagnostic methods are not available.


Assuntos
Testes Diagnósticos de Rotina/métodos , Vírus da Raiva/imunologia , Raiva/imunologia , Animais , Diagnóstico , Imunoensaio , Raiva/veterinária
19.
Viruses ; 12(6)2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32560066

RESUMO

Perhabdoviruses are a threat to some freshwater fish species raised in aquaculture farms in Europe. Although the genetic diversity of these viruses is suspected to be high, the classification of isolates is still in its infancy, with just one full-length genome available and only partial sequences for a limited number of others. Here, we characterized a series of viruses isolated from percids in France from 1999 to 2009 by sequencing the nucleoprotein (N) gene. Four main clusters were distinguished, all related at varying levels of similarity to one of the two already-recognized species, namely Perch perhabdovirus and Sea trout perhabdovirus. Furthermore, we obtained the complete genome of five isolates, including one belonging to Sea trout rhabdovirus. The analysis of the complete L genes and the concatenated open reading frames confirmed the existence of four main genetic clusters, sharing 69 to 74% similarity. We propose the assignation of all these viral isolates into four species, including two new ones: Perch perhabdovirus 1, Perch perhabdovirus 2, Sea trout perhabdovirus 1 and Sea trout perhabdovirus 2. In addition, we developed new primers to readily amplify specific portions of the N gene of any isolate of each species by conventional PCR. The presence of such genetically diverse viruses in France is likely due to divergent viral populations maintained in the wild and then introduced to experimental facilities or farms, as well as via trade between farms across the European continent. It is now urgent to improve the identification tools for this large group of viruses to prevent their unchecked dissemination.


Assuntos
Doenças dos Peixes/virologia , Genoma Viral , Infecções por Rhabdoviridae/veterinária , Rhabdoviridae/classificação , Rhabdoviridae/genética , Sequência de Aminoácidos , Animais , Peixes , Filogenia , Rhabdoviridae/química , Rhabdoviridae/isolamento & purificação , Infecções por Rhabdoviridae/virologia , Alinhamento de Sequência , Proteínas Virais/química , Proteínas Virais/genética
20.
Travel Med Infect Dis ; 36: 101766, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32525075

RESUMO

BACKGROUND: Sixty cases of rabies in international travellers from 1990 to 2012 were previously reviewed. We present here an update of rabies cases in international travellers from 2013 to 2019. METHODS: We systematically reviewed the existing literature and collected 23 cases of rabies in individuals who crossed an international border between the time of infection and diagnosis, or who were infected following expatriation or migration. RESULTS: Most cases were in male adult travellers and diagnosed in Europe and the Middle East, with most exposures in Asia or in Africa. Migrants originating from rabies-endemic low-and-middle income countries and their descendants accounted for two thirds of cases. Other cases were in tourists, business travellers and expatriates. Median travel duration (excluding migration trip) was 60 days (range 7-240 days). Most cases were due to dog bites and most common clinical presentation was furious rabies. In most patients (74%), no rabies post-exposure prophylaxis (RPEP) was administered before rabies symptoms appeared. Other patients received incomplete RPEP series. CONCLUSION: Rabies should be suspected in any patient with encephalitis or paralysis who travelled to, or migrated from a rabies-endemic country. Comprehensive information about a rabies risk should be given to travellers to rabies endemic countries, notably migrants visiting friends and relatives.


Assuntos
Raiva , Doença Relacionada a Viagens , Adulto , África , Animais , Ásia , Cães , Europa (Continente) , Humanos , Masculino , Oriente Médio , Raiva/epidemiologia , Estudos Retrospectivos , Viagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...