Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Viruses ; 12(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32824946

ABSTRACT

Mammarenaviruses include several known human pathogens, such as the prototypic lymphocytic choriomeningitis virus (LCMV) that can cause neurological diseases and Lassa virus (LASV) that causes endemic hemorrhagic fever infection. LASV-infected patients show diverse clinical manifestations ranging from asymptomatic infection to hemorrhage, multi-organ failures and death, the mechanisms of which have not been well characterized. We have previously shown that the matrix protein Z of pathogenic arenaviruses, including LASV and LCMV, can strongly inhibit the ability of the innate immune protein RIG-I to suppress type I interferon (IFN-I) expression, which serves as a mechanism of viral immune evasion and virulence. Here, we show that Z proteins of diverse LASV isolates derived from rodents and humans have a high degree of sequence variations at their N- and C-terminal regions and produce variable degrees of inhibition of human RIG-I (hRIG-I) function in an established IFN-ß promoter-driven luciferase (LUC) reporter assay. Additionally, we show that Z proteins of four known LCMV strains can also inhibit hRIG-I at variable degrees of efficiency. Collectively, our results confirm that Z proteins of pathogenic LASV and LCMV can inhibit hRIG-I and suggest that strain variations of the Z proteins can influence their efficiency to suppress host innate immunity that might contribute to viral virulence and disease heterogeneity.


Subject(s)
DEAD Box Protein 58/immunology , Lassa Fever/immunology , Lassa Fever/virology , Lassa virus/immunology , Receptors, Immunologic/immunology , Viral Proteins/immunology , Amino Acid Motifs , Cell Line , DEAD Box Protein 58/genetics , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon-beta/genetics , Interferon-beta/immunology , Lassa Fever/genetics , Lassa virus/chemistry , Lassa virus/classification , Lassa virus/genetics , Lymphocytic choriomeningitis virus/chemistry , Lymphocytic choriomeningitis virus/classification , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Receptors, Immunologic/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
2.
Viruses ; 12(4)2020 03 31.
Article in English | MEDLINE | ID: mdl-32244402

ABSTRACT

Lassa virus (LASV), which causes considerable morbidity and mortality annually, has a high genetic diversity across West Africa. LASV glycoprotein (GP) expresses this diversity, but most LASV vaccine candidates utilize only the Lineage IV LASV Josiah strain GP antigen as an immunogen and homologous challenge with Lineage IV LASV. In addition to the sequence variation amongst the LASV lineages, these lineages are also distinguished in their presentations. Inter-lineage variations within previously mapped B-cell and T-cell LASV GP epitopes and the breadth of protection in LASV vaccine/challenge studies were examined critically. Multiple alignments of the GP primary sequence of strains from each LASV lineage showed that LASV GP has diverging degrees of amino acid conservation within known epitopes among LASV lineages. Conformational B-cell epitopes spanning different sites in GP subunits were less impacted by LASV diversity. LASV GP diversity should influence the approach used for LASV vaccine design. Expression of LASV GP on viral vectors, especially in its prefusion configuration, has shown potential for protective LASV vaccines that can overcome LASV diversity. Advanced vaccine candidates should demonstrate efficacy against all LASV lineages for evidence of a pan-LASV vaccine.


Subject(s)
Epitopes/genetics , Lassa virus/immunology , Viral Envelope Proteins/genetics , Viral Vaccines/immunology , Animals , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , Drug Development , Epitopes/chemistry , Epitopes/immunology , Genetic Variation , Humans , Lassa virus/classification , Lassa virus/genetics , Phylogeny , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Viral Vaccines/genetics
3.
Viruses ; 12(4)2020 04 13.
Article in English | MEDLINE | ID: mdl-32294960

ABSTRACT

Lassa mammarenavirus (LASV) is the etiologic agent of Lassa fever. In endemic regions in West Africa, LASV genetic diversity tends to cluster by geographic area. Seven LASV lineages are recognized, but the role of viral genetic determinants on disease presentation in humans is uncertain. We investigated the geographic structure and distribution of LASV in West Africa. We found strong spatial clustering of LASV populations, with two major east-west and north-south diversity gradients. Analysis of ancestry components indicated that known LASV lineages diverged from an ancestral population that most likely circulated in Nigeria, although alternative locations, such as Togo, cannot be excluded. Extant sequences carrying the largest contribution of this ancestral population include the prototype Pinneo strain, the Togo isolates, and a few viruses isolated in Nigeria. The LASV populations that experienced the strongest drift circulate in Mali and the Ivory Coast. By focusing on sequences form a single LASV sublineage (IIg), we identified an ancestry component possibly associated with protection from a fatal disease outcome. Although the same ancestry component tends to associate with lower viral loads in plasma, the small sample size requires that these results are treated with extreme caution.


Subject(s)
Genetics, Population , Lassa Fever/epidemiology , Lassa Fever/virology , Lassa virus/classification , Lassa virus/genetics , Phylogeny , Africa, Western/epidemiology , Biodiversity , Genome, Viral , Humans , Phylogeography
4.
Virology ; 545: 10-15, 2020 06.
Article in English | MEDLINE | ID: mdl-32174454

ABSTRACT

Lassa fever (LF) is a viral hemorrhagic fever that causes high morbidity and severe mortality annually. The disease is endemic to two geographically separate areas within tropical West Africa, one in Nigeria and the second predominantly in Sierra Leone-Guinea-Liberia-Mali. Lassa virus (LASV), the causative agent of the disease, exhibits clear delineation of phylogeography between the endemic areas. In order to characterize the genetic nature of Nigerian-non-Nigerian epidemic split, we performed molecular epidemiological analyses on non-Nigerian isolates (lineage IV as well as lineage V) and their sister group from north-central Nigeria (lineage III). The results showed that adaptive genetic diversification has occurred between these currently circulating clusters in the spread process, and a number of replacement divergences have been fixed between these clusters on the viral RNA-dependent RNA polymerase (L protein). This study highlights the viral L protein could be a determinant factor for the epidemic split.


Subject(s)
Lassa Fever/virology , Lassa virus/genetics , Africa, Western/epidemiology , Epidemics , Genetic Variation , Humans , Lassa Fever/epidemiology , Lassa virus/classification , Lassa virus/isolation & purification , Nigeria/epidemiology , Phylogeny , Phylogeography , Viral Proteins/genetics
5.
Lancet Infect Dis ; 19(12): 1371-1378, 2019 12.
Article in English | MEDLINE | ID: mdl-31588039

ABSTRACT

BACKGROUND: An alarming rise in reported Lassa fever cases continues in west Africa. Liberia has the largest reported per capita incidence of Lassa fever cases in the region, but genomic information on the circulating strains is scarce. The aim of this study was to substantially increase the available pool of data to help foster the generation of targeted diagnostics and therapeutics. METHODS: Clinical serum samples collected from 17 positive Lassa fever cases originating from Liberia (16 cases) and Guinea (one case) within the past decade were processed at the Liberian Institute for Biomedical Research using a targeted-enrichment sequencing approach, producing 17 near-complete genomes. An additional 17 Lassa virus sequences (two from Guinea, seven from Liberia, four from Nigeria, and four from Sierra Leone) were generated from viral stocks at the US Centers for Disease Control and Prevention (Atlanta, GA) from samples originating from the Mano River Union (Guinea, Liberia, and Sierra Leone) region and Nigeria. Sequences were compared with existing Lassa virus genomes and published Lassa virus assays. FINDINGS: The 23 new Liberian Lassa virus genomes grouped within two clades (IV.A and IV.B) and were genetically divergent from those circulating elsewhere in west Africa. A time-calibrated phylogeographic analysis incorporating the new genomes suggests Liberia was the entry point of Lassa virus into the Mano River Union region and estimates the introduction to have occurred between 300-350 years ago. A high level of diversity exists between the Liberian Lassa virus genomes. Nucleotide percent difference between Liberian Lassa virus genomes ranged up to 27% in the L segment and 18% in the S segment. The commonly used Lassa Josiah-MGB assay was up to 25% divergent across the target sites when aligned to the Liberian Lassa virus genomes. INTERPRETATION: The large amount of novel genomic diversity of Lassa virus observed in the Liberian cases emphasises the need to match deployed diagnostic capabilities with locally circulating strains and underscores the importance of evaluating cross-lineage protection in the development of vaccines and therapeutics. FUNDING: Defense Biological Product Assurance Office of the US Department of Defense and the Armed Forces Health Surveillance Branch and its Global Emerging Infections Surveillance and Response Section.


Subject(s)
Lassa Fever/epidemiology , Lassa Fever/virology , Lassa virus/genetics , Genome, Viral , Genomics/methods , Genotype , Humans , Lassa Fever/diagnosis , Lassa virus/classification , Liberia/epidemiology , Phylogeny , Public Health Surveillance
6.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31413134

ABSTRACT

Lassa virus is genetically diverse with several lineages circulating in West Africa. This study aimed at describing the sequence variability of Lassa virus across Nigeria and inferring its spatiotemporal evolution. We sequenced and isolated 77 Lassa virus strains from 16 Nigerian states. The final data set, including previous works, comprised metadata and sequences of 219 unique strains sampled between 1969 and 2018 in 22 states. Most of this data originated from Lassa fever patients diagnosed at Irrua Specialist Teaching Hospital, Edo State, Nigeria. The majority of sequences clustered with the main Nigerian lineages II and III, while a few sequences formed a new cluster related to Lassa virus strains from Hylomyscus pamfi Within lineages II and III, seven and five sublineages, respectively, were distinguishable. Phylogeographic analysis suggests an origin of lineage II in the southeastern part of the country around Ebonyi State and a main vector of dispersal toward the west across the Niger River, through Anambra, Kogi, Delta, and Edo into Ondo State. The frontline of virus dispersal appears to be in Ondo. Minor vectors are directed northeast toward Taraba and Adamawa and south toward Imo and Rivers. Lineage III might have spread from northern Plateau State into Kaduna, Nasarawa, Federal Capital Territory, and Bauchi. One sublineage moved south and crossed the Benue River into Benue State. This study provides a geographic mapping of lineages and phylogenetic clusters in Nigeria at a higher resolution. In addition, we estimated the direction and time frame of virus dispersal in the country.IMPORTANCE Lassa virus is the causative agent of Lassa fever, a viral hemorrhagic fever with a case fatality rate of approximately 30% in Africa. Previous studies disclosed a geographical pattern in the distribution of Lassa virus strains and a westward movement of the virus across West Africa during evolution. Our study provides a deeper understanding of the geography of genetic lineages and sublineages of the virus in Nigeria. In addition, we modeled how the virus spread in the country. This knowledge allows us to predict into which geographical areas the virus might spread in the future and prioritize areas for Lassa fever surveillance. Our study not only aimed to generate Lassa virus sequences from across Nigeria but also to isolate and conserve the respective viruses for future research. Both isolates and sequences are important for the development and evaluation of medical countermeasures to treat and prevent Lassa fever, such as diagnostics, therapeutics, and vaccines.


Subject(s)
Lassa Fever/virology , Lassa virus/classification , Animals , Evolution, Molecular , Genetic Variation , Humans , Lassa Fever/epidemiology , Lassa Fever/transmission , Lassa virus/genetics , Murinae/virology , Nigeria/epidemiology , Phylogeny , Phylogeography
7.
Curr Opin Virol ; 37: 91-96, 2019 08.
Article in English | MEDLINE | ID: mdl-31357141

ABSTRACT

Lassa virus is endemic in a large area of sub-Saharan Africa, and exhibits a large amount of genetic diversity. Of the four currently recognized lineages, lineages I-III circulate in Nigeria, and lineage IV circulates in Sierra Leone, Guinea, and Liberia. However, several newly detected lineages have been proposed. LASV genetic diversity may result in differences in pathogenicity or response to medical countermeasures, necessitating the testing of multiple lineages during the development of countermeasures and diagnostics. Logistical and biosafety concerns can make it difficult to obtain representative collections of divergent LASV clades for comparison studies. For example, lack of a cold chain in remote areas, or shipping restrictions on live viruses can prevent the dissemination of natural virus isolates to researchers. Reverse genetics systems that have been developed for LASV can facilitate acquisition of hard-to-obtain LASV strains and enable comprehensive development of medical countermeasures.


Subject(s)
Genetic Variation , Lassa virus/genetics , Reverse Genetics , Animals , Humans , Lassa virus/classification , Mice , RNA, Viral/genetics
8.
Emerg Infect Dis ; 25(5): 999-1002, 2019 05.
Article in English | MEDLINE | ID: mdl-31002054

ABSTRACT

We report detection of Lassa virus and Crimean-Congo hemorrhagic fever virus infections in the area of Bamako, the capital of Mali. Our investigation found 2 cases of infection with each of these viruses. These results show the potential for both of these viruses to be endemic to Mali.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean/epidemiology , Hemorrhagic Fever, Crimean/virology , Lassa Fever/epidemiology , Lassa Fever/virology , Lassa virus , Hemorrhagic Fever Virus, Crimean-Congo/classification , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Humans , Lassa virus/classification , Lassa virus/genetics , Mali/epidemiology , Public Health Surveillance
9.
F1000Res ; 82019.
Article in English | MEDLINE | ID: mdl-30774934

ABSTRACT

Lassa virus (LASV) is a highly prevalent mammarenavirus in West Africa and is maintained in nature in a persistently infected rodent host, Mastomys natalensis, which is widely spread in sub-Saharan Africa. LASV infection of humans can cause Lassa fever (LF), a disease associated with high morbidity and significant mortality. Recent evidence indicates an LASV expansion outside its traditional endemic areas. In 2017, the World Health Organization (WHO) included LASV in top-priority pathogens and released a Target Product Profile (TPP) for vaccine development. Likewise, in 2018, the US Food and Drug Administration added LF to a priority review voucher program to encourage the development of preventive and therapeutics measures. In this article, we review recent progress in LASV vaccine research and development with a focus on the impact of LASV genetic and biological diversity on the design and development of vaccine candidates meeting the WHO's TPP for an LASV vaccine.


Subject(s)
Lassa Fever/prevention & control , Lassa Fever/virology , Lassa virus/classification , Africa, Western , Animals , Humans , Murinae , Viral Vaccines
10.
Emerg Infect Dis ; 25(5): 1026-1027, 2019 05.
Article in English | MEDLINE | ID: mdl-30807268

ABSTRACT

We reviewed data pertaining to the massive wave of Lassa fever cases that occurred in Nigeria in 2018. No new virus strains were detected, but in 2018, the outbreak response was intensified, additional diagnostic support was available, and surveillance sensitivity increased. These factors probably contributed to the high case count.


Subject(s)
Disease Outbreaks , Lassa Fever/epidemiology , Animals , History, 21st Century , Humans , Incidence , Lassa Fever/diagnosis , Lassa Fever/history , Lassa Fever/virology , Lassa virus/classification , Lassa virus/genetics , Lassa virus/isolation & purification , Nigeria/epidemiology , Public Health Surveillance , Seasons
11.
PLoS Negl Trop Dis ; 12(11): e0006971, 2018 11.
Article in English | MEDLINE | ID: mdl-30500827

ABSTRACT

Lassa virus (LASV) is endemic in parts of West Africa where it causes Lassa fever (LF), a viral hemorrhagic fever with frequent fatal outcomes. The diverse LASV strains are grouped into six major lineages based on the geographical location of the isolated strains. In this study, we have focused on the lineage II strains from southern Nigeria. We determined the viral sequences from positive cases of LF reported at tertiary hospitals in Ebonyi and Enugu between 2012 and 2016. Reverse transcription-polymerase chain reaction (RT-PCR) showed that 29 out of 123 suspected cases were positive for the virus among which 11 viral gene sequences were determined. Phylogenetic analysis of the complete coding sequences of the four viral proteins revealed that lineage II strains are broadly divided into two genetic clades that diverged from a common ancestor 195 years ago. One clade, consisting of strains from Ebonyi and Enugu, was more conserved than the other from Irrua, although the four viral proteins were evolving at similar rates in both clades. These results suggested that the viruses of these clades have been distinctively evolving in geographically separate parts of southern Nigeria. Furthermore, the epidemiological data of the 2014 outbreak highlighted the role of human-to-human transmission in this outbreak, which was supported by phylogenetic analysis showing that 13 of the 16 sequences clustered together. These results provide new insights into the evolution of LASV in southern Nigeria and have important implications for vaccine development, diagnostic assay design, and LF outbreak management.


Subject(s)
Lassa Fever/virology , Lassa virus/genetics , Lassa virus/isolation & purification , Evolution, Molecular , Genetic Variation , Humans , Lassa Fever/epidemiology , Lassa virus/classification , Nigeria/epidemiology , Phylogeny , Viral Proteins/genetics
12.
Emerg Infect Dis ; 24(6): 978-987, 2018 06.
Article in English | MEDLINE | ID: mdl-29775178

ABSTRACT

Nosocomial transmission of Lassa virus (LASV) is reported to be low when care for the index patient includes proper barrier nursing methods. We investigated whether asymptomatic LASV infection occurred in healthcare workers who used standard barrier nursing methods during the first 15 days of caring for a patient with Lassa fever in Sweden. Of 76 persons who were defined as having been potentially exposed to LASV, 53 provided blood samples for detection of LASV IgG. These persons also responded to a detailed questionnaire to evaluate exposure to different body fluids from the index patient. LASV-specific IgG was not detected in any of the 53 persons. Five of 53 persons had not been using proper barrier nursing methods. Our results strengthen the argument for a low risk of secondary transmission of LASV in humans when standard barrier nursing methods are used and the patient has only mild symptoms.


Subject(s)
Communicable Diseases, Imported/epidemiology , Communicable Diseases, Imported/virology , Cross Infection/epidemiology , Cross Infection/virology , Lassa Fever/epidemiology , Lassa Fever/virology , Nursing Care , Adult , Aged , Communicable Diseases, Imported/transmission , Cross Infection/transmission , Female , Health Personnel , Humans , Lassa Fever/transmission , Lassa virus/classification , Lassa virus/genetics , Lassa virus/immunology , Male , Middle Aged , Nursing Care/methods , Sentinel Surveillance , Sweden/epidemiology
14.
Sci Rep ; 6: 25280, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27140942

ABSTRACT

Lassa virus (LASV) causes a deadly haemorrhagic fever in humans, killing several thousand people in West Africa annually. For 40 years, the Natal multimammate rat, Mastomys natalensis, has been assumed to be the sole host of LASV. We found evidence that LASV is also hosted by other rodent species: the African wood mouse Hylomyscus pamfi in Nigeria, and the Guinea multimammate mouse Mastomys erythroleucus in both Nigeria and Guinea. Virus strains from these animals were isolated in the BSL-4 laboratory and fully sequenced. Phylogenetic analyses of viral genes coding for glycoprotein, nucleoprotein, polymerase and matrix protein show that Lassa strains detected in M. erythroleucus belong to lineages III and IV. The strain from H. pamfi clusters close to lineage I (for S gene) and between II &III (for L gene). Discovery of new rodent hosts has implications for LASV evolution and its spread into new areas within West Africa.


Subject(s)
Host Specificity , Lassa virus/isolation & purification , Lassa virus/physiology , Murinae/virology , Africa , Animals , Genotype , Guinea , Lassa virus/classification , Lassa virus/genetics , Nigeria , Phylogeny , Sequence Analysis, DNA
16.
Sci Rep ; 6: 21977, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26911443

ABSTRACT

This study aimed at reconstructing the spatial and temporal evolution of Lassa virus (LASV) in the natural host population. To this end, we generated 132 partial nucleoprotein sequences of LASV from M. natalensis trapped in 12 villages around Faranah, Upper Guinea, over a period of 12 years. This study reveals two main features of LASV evolution in M. natalensis. First, the virus evolves in the reservoir with a molecular clock rate of 9 (7-11) × 10(-4) position(-1) year(-1) implying that contemporary LASV lineages circulate in the Faranah area since less than 100 years. Second, viruses circulating in a specific village are diverse and polyphyletic. We observed, however, there are monophyletic clusters at village and sub-village level at specific points in time. In conclusion, our data indicate that the temporal and spatial pattern of LASV evolution in the natural reservoir is characterized by a combination of stationary circulation within a village and virus movement between villages. The latter feature is relevant for rodent control strategies, as it implies that recurrence of the virus from neighbouring villages may occur in villages where the virus has previously been eradicated.


Subject(s)
Lassa Fever/epidemiology , Lassa Fever/virology , Lassa virus/physiology , Spatio-Temporal Analysis , Viral Tropism , Animals , Genotype , Geography , Guinea , Lassa virus/classification , Murinae , Nucleocapsid Proteins/genetics , Phylogeny , Sequence Analysis, DNA
17.
Cell ; 162(4): 738-50, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26276630

ABSTRACT

The 2013-2015 West African epidemic of Ebola virus disease (EVD) reminds us of how little is known about biosafety level 4 viruses. Like Ebola virus, Lassa virus (LASV) can cause hemorrhagic fever with high case fatality rates. We generated a genomic catalog of almost 200 LASV sequences from clinical and rodent reservoir samples. We show that whereas the 2013-2015 EVD epidemic is fueled by human-to-human transmissions, LASV infections mainly result from reservoir-to-human infections. We elucidated the spread of LASV across West Africa and show that this migration was accompanied by changes in LASV genome abundance, fatality rates, codon adaptation, and translational efficiency. By investigating intrahost evolution, we found that mutations accumulate in epitopes of viral surface proteins, suggesting selection for immune escape. This catalog will serve as a foundation for the development of vaccines and diagnostics. VIDEO ABSTRACT.


Subject(s)
Genome, Viral , Lassa Fever/virology , Lassa virus/genetics , RNA, Viral/genetics , Africa, Western/epidemiology , Animals , Biological Evolution , Disease Reservoirs , Ebolavirus/genetics , Genetic Variation , Glycoproteins/genetics , Hemorrhagic Fever, Ebola/virology , Humans , Lassa Fever/epidemiology , Lassa Fever/transmission , Lassa virus/classification , Lassa virus/physiology , Murinae/genetics , Mutation , Nigeria/epidemiology , Viral Proteins/genetics , Zoonoses/epidemiology , Zoonoses/virology
18.
Emerg Infect Dis ; 21(4): 609-18, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25811712

ABSTRACT

Lassa virus (LASV) is endemic to parts of West Africa and causes highly fatal hemorrhagic fever. The multimammate rat (Mastomys natalensis) is the only known reservoir of LASV. Most human infections result from zoonotic transmission. The very diverse LASV genome has 4 major lineages associated with different geographic locations. We used reverse transcription PCR and resequencing microarrays to detect LASV in 41 of 214 samples from rodents captured at 8 locations in Sierra Leone. Phylogenetic analysis of partial sequences of nucleoprotein (NP), glycoprotein precursor (GPC), and polymerase (L) genes showed 5 separate clades within lineage IV of LASV in this country. The sequence diversity was higher than previously observed; mean diversity was 7.01% for nucleoprotein gene at the nucleotide level. These results may have major implications for designing diagnostic tests and therapeutic agents for LASV infections in Sierra Leone.


Subject(s)
Genetic Variation , Lassa Fever/epidemiology , Lassa Fever/virology , Lassa virus/classification , Lassa virus/genetics , Phylogeography , Animals , Genes, Viral , Genome, Viral , Genotype , Geography , Lassa Fever/transmission , Oligonucleotide Array Sequence Analysis , Phylogeny , Rats , Sierra Leone/epidemiology
19.
Virology ; 476: 249-256, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25559385

ABSTRACT

Despite its near pan-African range, the Natal multimammate mouse, Mastomys natalensis, carries the human pathogen Lassa virus only in West Africa, while the seemingly non-pathogenic arenaviruses Mopeia, Morogoro, and Luna have been detected in this semi-commensal rodent in Mozambique/Zimbabwe, Tanzania and Zambia, respectively. Here, we describe a novel arenavirus in M. natalensis from Gairo district of central Tanzania, for which we propose the name "Gairo virus". Surprisingly, the virus is not closely related with Morogoro virus that infects M. natalensis only 90km south of Gairo, but clusters phylogenetically with Mobala-like viruses that infect non-M. natalensis host species in Central African Republic and Ethiopia. Despite the evolutionary distance, Gairo virus shares basic ecological features with the other M. natalensis-borne viruses Lassa and Morogoro. Our data show that M. natalensis, carrying distantly related viruses even in the same geographical area, is a potent reservoir host for a variety of arenaviruses.


Subject(s)
Arenaviridae Infections/veterinary , Arenavirus/isolation & purification , Genetic Variation , Murinae/virology , Rodent Diseases/virology , Animals , Antibodies, Viral/immunology , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Arenavirus/classification , Arenavirus/genetics , Disease Reservoirs/virology , Female , Lassa virus/classification , Lassa virus/genetics , Lassa virus/isolation & purification , Male , Molecular Sequence Data , Murinae/immunology , Phylogeny , Rodent Diseases/immunology , Tanzania
20.
PLoS Negl Trop Dis ; 7(12): e2582, 2013.
Article in English | MEDLINE | ID: mdl-24340119

ABSTRACT

BACKGROUND: Lassa fever is an acute viral illness characterized by multi-organ failure and hemorrhagic manifestations. Lassa fever is most frequently diagnosed in Nigeria, Sierra Leone, Liberia, and Guinea, although sporadic cases have been recorded in other West African countries, including Mali. The etiological agent of Lassa fever is Lassa virus (LASV), an Arenavirus which is maintained in nature and frequently transmitted to humans by Mastomys natalensis. The purpose of this study was to better define the geographic distribution of LASV-infected rodents in sub-Saharan Mali. METHODOLOGIES/PRINCIPAL FINDINGS: Small mammals were live-trapped at various locations across Mali for the purpose of identifying potential zoonotic pathogens. Serological and molecular assays were employed and determined LASV infected rodents were exclusively found in the southern Mali near the border of Côte d'Ivoire. Overall, 19.4% of Mastomys natalensis sampled in this region had evidence of LASV infection, with prevalence rates for individual villages ranging from 0 to 52%. Full-length genomic sequences were determined using high throughput sequencing methodologies for LASV isolates generated from tissue samples of rodents collected in four villages and confirmed the phylogenetic clustering of Malian LASV with strain AV. CONCLUSIONS/SIGNIFICANCE: The risk of human infections with LASV is greatest in villages in southern Mali. Lassa fever should be considered in the differential diagnosis for febrile individuals and appropriate diagnostic techniques need to be established to determine the incidence of infection and disease in these regions.


Subject(s)
Lassa Fever/veterinary , Lassa virus/classification , Lassa virus/genetics , Phylogeography , Rodent Diseases/epidemiology , Rodent Diseases/virology , Topography, Medical , Animals , Cluster Analysis , Genome, Viral , High-Throughput Nucleotide Sequencing , Lassa Fever/epidemiology , Lassa Fever/virology , Lassa virus/isolation & purification , Mali/epidemiology , Molecular Sequence Data , Murinae , Prevalence , RNA, Viral/genetics , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...