Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
Add more filters

Publication year range
1.
Nature ; 598(7882): 652-656, 2021 10.
Article in English | MEDLINE | ID: mdl-34646009

ABSTRACT

Humans are considered as the main host for Mycobacterium leprae1, the aetiological agent of leprosy, but spillover has occurred to other mammals that are now maintenance hosts, such as nine-banded armadillos and red squirrels2,3. Although naturally acquired leprosy has also been described in captive nonhuman primates4-7, the exact origins of infection remain unclear. Here we describe leprosy-like lesions in two wild populations of western chimpanzees (Pan troglodytes verus) in Cantanhez National Park, Guinea-Bissau and Taï National Park, Côte d'Ivoire, West Africa. Longitudinal monitoring of both populations revealed the progression of disease symptoms compatible with advanced leprosy. Screening of faecal and necropsy samples confirmed the presence of M. leprae as the causative agent at each site and phylogenomic comparisons with other strains from humans and other animals show that the chimpanzee strains belong to different and rare genotypes (4N/O and 2F). These findings suggest that M. leprae may be circulating in more wild animals than suspected, either as a result of exposure to humans or other unknown environmental sources.


Subject(s)
Leprosy/veterinary , Pan troglodytes/microbiology , Animals , Autopsy/veterinary , Cote d'Ivoire , Feces/microbiology , Genotype , Guinea-Bissau , Humans , Leprosy/microbiology , Mycobacterium leprae/genetics , Mycobacterium leprae/isolation & purification , Phylogeny
2.
BMC Infect Dis ; 24(1): 808, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39123104

ABSTRACT

BACKGROUND: Campylobacter spp. is a significant etiological agent of bacterial gastroenteritis globally. In Burkina Faso (BFA), the actual impact of this pathogen on gastroenteritis is considerably underestimated, primarily due to inadequate surveillance systems. OBJECTIVES: This study aimed to investigate the proportion of Campylobacter species responsible for acute gastroenteritis among patients of all ages in urban and rural areas of BFA, using molecular biology techniques. STUDY DESIGN & METHODS: Between 2018 and 2021, faecal specimens were obtained from 1,295 individuals presenting with acute gastroenteritis. These samples underwent screening for the Campylobacter coli/jejuni/lari complex utilizing real-time polymerase chain reaction (PCR) assays. Subsequently, positive samples were subjected to species-level differentiation through the application of species-specific primers. RESULTS: Campylobacter spp. was detected in 25.0% (324/1,295) of the samples analysed. The majority of positive samples (95%, 308/324) were obtained from children under 5 years of age. Species identification was performed on a subset of 114 isolates, revealing 51 Campylobacter jejuni, 10 Campylobacter coli, and 53 Campylobacter isolates that remained unspeciated. CONCLUSIONS: This study reveals a significant prevalence of Campylobacter species among patients with acute gastroenteritis, with a particularly high incidence observed in children under 5 years of age. Based on these findings, the implementation of routine Campylobacter surveillance in public health laboratories is strongly recommended to better monitor and address this health concern.


Subject(s)
Campylobacter Infections , Campylobacter , Feces , Humans , Burkina Faso/epidemiology , Campylobacter Infections/epidemiology , Campylobacter Infections/microbiology , Child, Preschool , Infant , Campylobacter/isolation & purification , Campylobacter/genetics , Campylobacter/classification , Female , Male , Child , Adult , Adolescent , Feces/microbiology , Young Adult , Middle Aged , Gastroenteritis/microbiology , Gastroenteritis/epidemiology , Prevalence , Infant, Newborn , Campylobacter jejuni/isolation & purification , Campylobacter jejuni/genetics , Campylobacter jejuni/classification , Aged , Enteritis/microbiology , Enteritis/epidemiology , Acute Disease , Incidence
3.
Am J Primatol ; 86(8): e23652, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38807168

ABSTRACT

Nematodes belonging to the genus Oesophagostomum frequently infect wild chimpanzees (Pan troglodytes) across widely separated field sites. Nodular lesions (granulomas) containing Oesophagostomum are commonly seen in the abdomen of infected chimpanzees post-mortem. At Taï National Park, Côte d'Ivoire, previous studies have identified larvae of a variety of Oesophagostomum spp. in wild chimpanzee stool, based on sequencing of larval DNA, and nodular lesions associated with Oesophagostomum, identified morphologically to the genus level but not sequenced. Here we present three recent cases of parasitic granulomas found post-mortem in chimpanzees at Taï. We complement descriptions of gross pathology, histopathology and parasitology with PCR and sequencing of DNA isolated from the parasitic nodules and from adult worms found inside the nodules. In all three cases, we identify Oesophagostomum stephanostomum as the causative agent. The sequences from this study were identical to the only other published sequences from nodules in nonhuman primates-those from the wild chimpanzees of Gombe, Tanzania.


Subject(s)
Ape Diseases , Oesophagostomiasis , Oesophagostomum , Pan troglodytes , Animals , Pan troglodytes/parasitology , Oesophagostomum/isolation & purification , Oesophagostomum/genetics , Cote d'Ivoire , Oesophagostomiasis/veterinary , Oesophagostomiasis/parasitology , Ape Diseases/parasitology , Granuloma/veterinary , Granuloma/parasitology , Male , Female , Parks, Recreational
4.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Article in English | MEDLINE | ID: mdl-33876746

ABSTRACT

Humans harbor diverse communities of microorganisms, the majority of which are bacteria in the gastrointestinal tract. These gut bacterial communities in turn host diverse bacteriophage (hereafter phage) communities that have a major impact on their structure, function, and, ultimately, human health. However, the evolutionary and ecological origins of these human-associated phage communities are poorly understood. To address this question, we examined fecal phageomes of 23 wild nonhuman primate taxa, including multiple representatives of all the major primate radiations. We find relatives of the majority of human-associated phages in wild primates. Primate taxa have distinct phageome compositions that exhibit a clear phylosymbiotic signal, and phage-superhost codivergence is often detected for individual phages. Within species, neighboring social groups harbor compositionally and evolutionarily distinct phageomes, which are structured by superhost social behavior. Captive nonhuman primate phageome composition is intermediate between that of their wild counterparts and humans. Phage phylogenies reveal replacement of wild great ape-associated phages with human-associated ones in captivity and, surprisingly, show no signal for the persistence of wild-associated phages in captivity. Together, our results suggest that potentially labile primate-phage associations have persisted across millions of years of evolution. Across primates, these phylosymbiotic and sometimes codiverging phage communities are shaped by transmission between groupmates through grooming and are dramatically modified when primates are moved into captivity.


Subject(s)
Bacteriophages/pathogenicity , Gastrointestinal Microbiome , Hominidae/virology , Virome , Animals , Bacteriophages/genetics , Environment , Evolution, Molecular , Hominidae/classification , Hominidae/genetics , Hominidae/microbiology , Phylogeny , Social Behavior
5.
Virus Genes ; 59(3): 370-376, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36932280

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with increased transmissibility, virulence and immune escape abilities have heavily altered the COVID-19 pandemic's course. Deciphering local and global transmission patterns of those variants is thus key in building a profound understanding of the virus' spread around the globe. In the present study, we investigate SARS-CoV-2 variant epidemiology in Côte d'Ivoire, Western sub-Saharan Africa. We therefore generated 234 full SARS-CoV-2 genomes stemming from Central and Northern Côte d'Ivoire. Covering the first and second pandemic wave the country had been facing, we identified 20 viral lineages and showed that in Côte d'Ivoire the second pandemic wave in 2021 was driven by the spread of the Alpha (B.1.1.7) and Eta (B.1.525) variant. Our analyses are consistent with a limited number of international introductions of Alpha and Eta into Côte d'Ivoire, and those introduction events mostly stemmed from within the West African subregion. This suggests that subregional travel to Côte d'Ivoire had more impact on local pandemic waves than direct intercontinental travel.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Cote d'Ivoire/epidemiology , SARS-CoV-2/genetics , Pandemics , COVID-19/epidemiology
6.
Clin Infect Dis ; 74(5): 882-890, 2022 03 09.
Article in English | MEDLINE | ID: mdl-34089598

ABSTRACT

BACKGROUND: In October 2020, after the first wave of coronavirus disease 2019 (COVID-19), only 8290 confirmed cases were reported in Kinshasa, Democratic Republic of the Congo, but the real prevalence remains unknown. To guide public health policies, we aimed to describe the prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunoglobulin G (IgG) antibodies in the general population in Kinshasa. METHODS: We conducted a cross-sectional, household-based serosurvey between 22 October 2020 and 8 November 2020. Participants were interviewed at home and tested for antibodies against SARS-CoV-2 spike and nucleocapsid proteins in a Luminex-based assay. A positive serology was defined as a sample that reacted with both SARS-CoV-2 proteins (100% sensitivity, 99.7% specificity). The overall weighted, age-standardized prevalence was estimated and the infection-to-case ratio was calculated to determine the proportion of undiagnosed SARS-CoV-2 infections. RESULTS: A total of 1233 participants from 292 households were included (mean age, 32.4 years; 764 [61.2%] women). The overall weighted, age-standardized SARS-CoV-2 seroprevalence was 16.6% (95% CI: 14.0-19.5%). The estimated infection-to-case ratio was 292:1. Prevalence was higher among participants ≥40 years than among those <18 years (21.2% vs 14.9%, respectively; P < .05). It was also higher in participants who reported hospitalization than among those who did not (29.8% vs 16.0%, respectively; P < .05). However, differences were not significant in the multivariate model (P = .1). CONCLUSIONS: The prevalence of SARS-CoV-2 is much higher than the number of COVID-19 cases reported. These results justify the organization of a sequential series of serosurveys by public health authorities to adapt response measures to the dynamics of the pandemic.


Subject(s)
COVID-19 , Adult , Antibodies, Viral , COVID-19/diagnosis , COVID-19/epidemiology , Cross-Sectional Studies , Democratic Republic of the Congo/epidemiology , Female , Humans , Prevalence , SARS-CoV-2 , Seroepidemiologic Studies
7.
Mol Biol Evol ; 38(7): 2818-2830, 2021 06 25.
Article in English | MEDLINE | ID: mdl-33720357

ABSTRACT

Viruses closely related to human pathogens can reveal the origins of human infectious diseases. Human herpes simplexvirus type 1 (HSV-1) and type 2 (HSV-2) are hypothesized to have arisen via host-virus codivergence and cross-species transmission. We report the discovery of novel herpes simplexviruses during a large-scale screening of fecal samples from wild gorillas, bonobos, and chimpanzees. Phylogenetic analysis indicates that, contrary to expectation, simplexviruses from these African apes are all more closely related to HSV-2 than to HSV-1. Molecular clock-based hypothesis testing suggests the divergence between HSV-1 and the African great ape simplexviruses likely represents a codivergence event between humans and gorillas. The simplexviruses infecting African great apes subsequently experienced multiple cross-species transmission events over the past 3 My, the most recent of which occurred between humans and bonobos around 1 Ma. These findings revise our understanding of the origins of human herpes simplexviruses and suggest that HSV-2 is one of the earliest zoonotic pathogens.


Subject(s)
Hominidae/virology , Phylogeny , Simplexvirus/genetics , Viral Zoonoses , Animals , Herpesvirus 2, Human , Humans , Sequence Analysis, DNA
8.
J Gen Virol ; 103(1)2022 01.
Article in English | MEDLINE | ID: mdl-35077341

ABSTRACT

Decades after its discovery in East Africa, Zika virus (ZIKV) emerged in Brazil in 2013 and infected millions of people during intense urban transmission. Whether vertebrates other than humans are involved in ZIKV transmission cycles remained unclear. Here, we investigate the role of different animals as ZIKV reservoirs by testing 1723 sera of pets, peri-domestic animals and African non-human primates (NHP) sampled during 2013-2018 in Brazil and 2006-2016 in Côte d'Ivoire. Exhaustive neutralization testing substantiated co-circulation of multiple flaviviruses and failed to confirm ZIKV infection in pets or peri-domestic animals in Côte d'Ivoire (n=259) and Brazil (n=1416). In contrast, ZIKV seroprevalence was 22.2% (2/9, 95% CI, 2.8-60.1) in West African chimpanzees (Pan troglodytes verus) and 11.1% (1/9, 95% CI, 0.3-48.3) in king colobus (Colobus polycomos). Our results indicate that while NHP may represent ZIKV reservoirs in Africa, pets or peri-domestic animals likely do not play a role in ZIKV transmission cycles.


Subject(s)
Animals, Domestic/virology , Primates/virology , Zika Virus Infection/epidemiology , Zika Virus Infection/virology , Zika Virus , Africa , Animals , Brazil , Cote d'Ivoire , Humans , Neutralization Tests , Seroepidemiologic Studies , Zika Virus Infection/transmission
11.
Proc Natl Acad Sci U S A ; 116(8): 3229-3238, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718403

ABSTRACT

Human and simian immunodeficiency viruses (HIV/SIVs) use CD4 as the primary receptor to enter target cells. Here, we show that the chimpanzee CD4 is highly polymorphic, with nine coding variants present in wild populations, and that this diversity interferes with SIV envelope (Env)-CD4 interactions. Testing the replication fitness of SIVcpz strains in CD4+ T cells from captive chimpanzees, we found that certain viruses were unable to infect cells from certain hosts. These differences were recapitulated in CD4 transfection assays, which revealed a strong association between CD4 genotypes and SIVcpz infection phenotypes. The most striking differences were observed for three substitutions (Q25R, Q40R, and P68T), with P68T generating a second N-linked glycosylation site (N66) in addition to an invariant N32 encoded by all chimpanzee CD4 alleles. In silico modeling and site-directed mutagenesis identified charged residues at the CD4-Env interface and clashes between CD4- and Env-encoded glycans as mechanisms of inhibition. CD4 polymorphisms also reduced Env-mediated cell entry of monkey SIVs, which was dependent on at least one D1 domain glycan. CD4 allele frequencies varied among wild chimpanzees, with high diversity in all but the western subspecies, which appeared to have undergone a selective sweep. One allele was associated with lower SIVcpz prevalence rates in the wild. These results indicate that substitutions in the D1 domain of the chimpanzee CD4 can prevent SIV cell entry. Although some SIVcpz strains have adapted to utilize these variants, CD4 diversity is maintained, protecting chimpanzees against infection with SIVcpz and other SIVs to which they are exposed.


Subject(s)
CD4 Antigens/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/genetics , Viral Envelope Proteins/genetics , Animals , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Evolution, Molecular , Genetic Variation/immunology , HIV/genetics , HIV/pathogenicity , Humans , Pan troglodytes/genetics , Pan troglodytes/immunology , Polysaccharides/genetics , Polysaccharides/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Viral Envelope Proteins/immunology
12.
Gut ; 2021 Apr 22.
Article in English | MEDLINE | ID: mdl-33888516

ABSTRACT

OBJECTIVE: Haemorrhoidal disease (HEM) affects a large and silently suffering fraction of the population but its aetiology, including suspected genetic predisposition, is poorly understood. We report the first genome-wide association study (GWAS) meta-analysis to identify genetic risk factors for HEM to date. DESIGN: We conducted a GWAS meta-analysis of 218 920 patients with HEM and 725 213 controls of European ancestry. Using GWAS summary statistics, we performed multiple genetic correlation analyses between HEM and other traits as well as calculated HEM polygenic risk scores (PRS) and evaluated their translational potential in independent datasets. Using functional annotation of GWAS results, we identified HEM candidate genes, which differential expression and coexpression in HEM tissues were evaluated employing RNA-seq analyses. The localisation of expressed proteins at selected loci was investigated by immunohistochemistry. RESULTS: We demonstrate modest heritability and genetic correlation of HEM with several other diseases from the GI, neuroaffective and cardiovascular domains. HEM PRS validated in 180 435 individuals from independent datasets allowed the identification of those at risk and correlated with younger age of onset and recurrent surgery. We identified 102 independent HEM risk loci harbouring genes whose expression is enriched in blood vessels and GI tissues, and in pathways associated with smooth muscles, epithelial and endothelial development and morphogenesis. Network transcriptomic analyses highlighted HEM gene coexpression modules that are relevant to the development and integrity of the musculoskeletal and epidermal systems, and the organisation of the extracellular matrix. CONCLUSION: HEM has a genetic component that predisposes to smooth muscle, epithelial and connective tissue dysfunction.

14.
BMC Infect Dis ; 21(1): 539, 2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34098893

ABSTRACT

BACKGROUND: In sub-Saharan Africa, acute respiratory infections (ARI), acute gastrointestinal infections (GI) and acute febrile disease of unknown cause (AFDUC) have a large disease burden, especially among children, while respective aetiologies often remain unresolved. The need for robust infectious disease surveillance to detect emerging pathogens along with common human pathogens has been highlighted by the ongoing novel coronavirus disease 2019 (COVID-19) pandemic. The African Network for Improved Diagnostics, Epidemiology and Management of Common Infectious Agents (ANDEMIA) is a sentinel surveillance study on the aetiology and clinical characteristics of ARI, GI and AFDUC in sub-Saharan Africa. METHODS: ANDEMIA includes 12 urban and rural health care facilities in four African countries (Côte d'Ivoire, Burkina Faso, Democratic Republic of the Congo and Republic of South Africa). It was piloted in 2018 in Côte d'Ivoire and the initial phase will run from 2019 to 2021. Case definitions for ARI, GI and AFDUC were established, as well as syndrome-specific sampling algorithms including the collection of blood, naso- and oropharyngeal swabs and stool. Samples are tested using comprehensive diagnostic protocols, ranging from classic bacteriology and antimicrobial resistance screening to multiplex real-time polymerase chain reaction (PCR) systems and High Throughput Sequencing. In March 2020, PCR testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and analysis of full genomic information was included in the study. Standardised questionnaires collect relevant clinical, demographic, socio-economic and behavioural data for epidemiologic analyses. Controls are enrolled over a 12-month period for a nested case-control study. Data will be assessed descriptively and aetiologies will be evaluated using a latent class analysis among cases. Among cases and controls, an integrated analytic approach using logistic regression and Bayesian estimation will be employed to improve the assessment of aetiology and associated risk factors. DISCUSSION: ANDEMIA aims to expand our understanding of ARI, GI and AFDUC aetiologies in sub-Saharan Africa using a comprehensive laboratory diagnostics strategy. It will foster early detection of emerging threats and continued monitoring of important common pathogens. The network collaboration will be strengthened and site diagnostic capacities will be reinforced to improve quality management and patient care.


Subject(s)
Communicable Diseases/diagnosis , Communicable Diseases/epidemiology , Mass Screening , Sentinel Surveillance , Bayes Theorem , Burkina Faso , Case-Control Studies , Cote d'Ivoire , Democratic Republic of the Congo , Fever/epidemiology , Fever/microbiology , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/microbiology , Humans , Real-Time Polymerase Chain Reaction , Respiratory Tract Infections/epidemiology , South Africa
16.
Proc Natl Acad Sci U S A ; 115(36): E8450-E8459, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30127015

ABSTRACT

Wild-living African apes are endemically infected with parasites that are closely related to human Plasmodium vivax, a leading cause of malaria outside Africa. This finding suggests that the origin of P. vivax was in Africa, even though the parasite is now rare in humans there. To elucidate the emergence of human P. vivax and its relationship to the ape parasites, we analyzed genome sequence data of P. vivax strains infecting six chimpanzees and one gorilla from Cameroon, Gabon, and Côte d'Ivoire. We found that ape and human parasites share nearly identical core genomes, differing by only 2% of coding sequences. However, compared with the ape parasites, human strains of P. vivax exhibit about 10-fold less diversity and have a relative excess of nonsynonymous nucleotide polymorphisms, with site-frequency spectra suggesting they are subject to greatly relaxed purifying selection. These data suggest that human P. vivax has undergone an extreme bottleneck, followed by rapid population expansion. Investigating potential host-specificity determinants, we found that ape P. vivax parasites encode intact orthologs of three reticulocyte-binding protein genes (rbp2d, rbp2e, and rbp3), which are pseudogenes in all human P. vivax strains. However, binding studies of recombinant RBP2e and RBP3 proteins to human, chimpanzee, and gorilla erythrocytes revealed no evidence of host-specific barriers to red blood cell invasion. These data suggest that, from an ancient stock of P. vivax parasites capable of infecting both humans and apes, a severely bottlenecked lineage emerged out of Africa and underwent rapid population growth as it spread globally.


Subject(s)
Evolution, Molecular , Genome-Wide Association Study , Plasmodium vivax/genetics , Polymorphism, Genetic , Protozoan Proteins/genetics , Selection, Genetic , Animals , Cameroon , Cote d'Ivoire , Female , Gabon , Gorilla gorilla , Humans , Male , Pan troglodytes , Protozoan Proteins/metabolism , Pseudogenes
17.
Emerg Infect Dis ; 26(6): 1283-1286, 2020 06.
Article in English | MEDLINE | ID: mdl-32441635

ABSTRACT

Yaws-like lesions are widely reported in wild African great apes, yet the causative agent has not been confirmed in affected animals. We describe yaws-like lesions in a wild chimpanzee in Guinea for which we demonstrate infection with Treponema pallidum subsp. pertenue. Assessing the conservation implications of this pathogen requires further research.


Subject(s)
Yaws , Animals , Guinea/epidemiology , Pan troglodytes , Treponema , Treponema pallidum/genetics , Yaws/epidemiology , Yaws/veterinary
18.
Emerg Infect Dis ; 26(9): 2205-2209, 2020 09.
Article in English | MEDLINE | ID: mdl-32818404

ABSTRACT

After the 2017 Ebola virus (EBOV) outbreak in Likati, a district in northern Democratic Republic of the Congo, we sampled small mammals from the location where the primary case-patient presumably acquired the infection. None tested positive for EBOV RNA or antibodies against EBOV, highlighting the ongoing challenge in detecting animal reservoirs for EBOV.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Animals, Wild , Democratic Republic of the Congo/epidemiology , Disease Outbreaks , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/epidemiology , Humans
19.
Arch Virol ; 165(10): 2291-2299, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32754877

ABSTRACT

The multimammate mouse (Mastomys natalensis; M. natalensis) serves as the main reservoir for the zoonotic arenavirus Lassa virus (LASV), and this has led to considerable investigation into the distribution of LASV and other related arenaviruses in this host species. In contrast to the situation with arenaviruses, the presence of other viruses in M. natalensis remains largely unexplored. In this study, herpesviruses and polyomaviruses were identified and partially characterized by PCR methods, sequencing, and phylogenetic analysis. In tissues sampled from M. natalensis populations in Côte d'Ivoire and Mali, six new DNA viruses (four betaherpesviruses, one gammaherpesvirus and one polyomavirus) were identified. Phylogenetic analysis based on glycoprotein B amino acid sequences showed that the herpesviruses clustered with cytomegaloviruses and rhadinoviruses of multiple rodent species. The complete circular genome of the newly identified polyomavirus was amplified by PCR. Amino acid sequence analysis of the large T antigen or VP1 showed that this virus clustered with a known polyomavirus from a house mouse (species Mus musculus polyomavirus 1). These two polyomaviruses form a clade with other rodent polyomaviruses, and the newly identified virus represents the third known polyomavirus of M. natalensis. This study represents the first identification of herpesviruses and the discovery of a novel polyomavirus in M. natalensis. In contrast to arenaviruses, we anticipate that these newly identified viruses represent a low zoonotic risk due to the normally highly restricted specificity of members of these two DNA virus families to their individual mammalian host species.


Subject(s)
Genome, Viral , Herpesviridae Infections/epidemiology , Herpesviridae/genetics , Phylogeny , Polyomavirus Infections/epidemiology , Polyomavirus/genetics , Rodent Diseases/epidemiology , Africa South of the Sahara/epidemiology , Animals , Antigens, Viral, Tumor/genetics , Capsid Proteins/genetics , Disease Reservoirs/virology , Herpesviridae/classification , Herpesviridae/isolation & purification , Herpesviridae Infections/virology , Host Specificity , Molecular Typing , Murinae/virology , Polyomavirus/classification , Polyomavirus/isolation & purification , Polyomavirus Infections/virology , Rodent Diseases/virology , Viral Envelope Proteins/genetics
20.
Virus Genes ; 56(1): 95-98, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31654295

ABSTRACT

To date, only two rodent-borne hantaviruses have been detected in sub-Saharan Africa. Here, we report the detection of a yet unknown hantavirus in a Natal mastomys (Mastomys natalensis) in Méliandou, Guinea, in 2014. The phylogenetic placement of this virus suggests that it might represent a cross-order spillover event from an unknown bat or eulipotyphlan host.


Subject(s)
Hantavirus Infections/veterinary , Murinae/virology , Orthohantavirus/isolation & purification , Rodent Diseases/virology , Animals , Guinea , Orthohantavirus/classification , Orthohantavirus/genetics , Hantavirus Infections/virology , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL