Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 245
Filter
1.
Emerg Microbes Infect ; 13(1): 2356149, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38747061

ABSTRACT

Lassa virus (LASV), a risk-group 4 pathogen, must be handled in biosafety level-4 (BSL-4) conditions, thereby limiting its research and antiviral development. Here, we developed a novel LASV reverse genetics system which, to our knowledge, is the first to study the complete LASV life cycle under BSL-2 conditions. Viral particles can be produced efficiently when LASV minigenomic RNA harbouring minimal viral cis-elements and reporter genes is transfected into a helper cell line stably expressing viral NP, GP, Z and L proteins. The resulting defective virions, named LASVmg, can propagate only in the helper cell line, providing a BSL-2 model to study the complete LASV life cycle. Using this model, we found that a previously reported cellular receptor α-dystroglycan is dispensable for LASVmg infection. Furthermore, we showed that ribavirin can inhibit LASVmg infection by inducing viral mutations. This new BSL-2 system should facilitate studying the LASV life cycle and screening antivirals.


Subject(s)
Lassa virus , Reverse Genetics , Lassa virus/genetics , Lassa virus/physiology , Reverse Genetics/methods , Humans , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Cell Line , Virus Replication , Lassa Fever/virology , Ribavirin/pharmacology , Vero Cells , Containment of Biohazards , Genome, Viral , Virion/genetics , Virion/metabolism
2.
J Virol ; 98(6): e0057824, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38767352

ABSTRACT

The mammarenavirus Lassa virus (LASV) causes the life-threatening hemorrhagic fever disease, Lassa fever. The lack of licensed medical countermeasures against LASV underscores the urgent need for the development of novel LASV vaccines, which has been hampered by the requirement for a biosafety level 4 facility to handle live LASV. Here, we investigated the efficacy of mRNA-lipid nanoparticle (mRNA-LNP)-based vaccines expressing the LASV glycoprotein precursor (LASgpc) or nucleoprotein (LCMnp) of the prototypic mammarenavirus, lymphocytic choriomeningitis virus (LCMV), in mice. Two doses of LASgpc- or LCMnp-mRNA-LNP administered intravenously (i.v.) protected C57BL/6 mice from a lethal challenge with a recombinant (r) LCMV expressing a modified LASgpc (rLCMV/LASgpc2m) inoculated intracranially. Intramuscular (i.m.) immunization with two doses of LASgpc- or LCMnp-mRNA-LNP significantly reduced the viral load in C57BL/6 mice inoculated i.v. with rLCMV/LASgpc2m. High levels of viremia and lethality were observed in CBA mice inoculated i.v. with rLCMV/LASgpc2m, which were abrogated by i.m. immunization with two doses of LASgpc-mRNA-LNP. The protective efficacy of two i.m. doses of LCMnp-mRNA-LNP was confirmed in a lethal hemorrhagic disease model of FVB mice i.v. inoculated with wild-type rLCMV. In all conditions tested, negligible and high levels of LASgpc- and LCMnp-specific antibodies were detected in mRNA-LNP-immunized mice, respectively, but robust LASgpc- and LCMnp-specific CD8+ T cell responses were induced. Accordingly, plasma from LASgpc-mRNA-LNP-immunized mice did not exhibit neutralizing activity. Our findings and surrogate mouse models of LASV infection, which can be studied at a reduced biocontainment level, provide a critical foundation for the rapid development of mRNA-LNP-based LASV vaccines.IMPORTANCELassa virus (LASV) is a highly pathogenic mammarenavirus responsible for several hundred thousand infections annually in West African countries, causing a high number of lethal Lassa fever (LF) cases. Despite its significant impact on human health, clinically approved, safe, and effective medical countermeasures against LF are not available. The requirement of a biosafety level 4 facility to handle live LASV has been one of the main obstacles to the research and development of LASV countermeasures. Here, we report that two doses of mRNA-lipid nanoparticle-based vaccines expressing the LASV glycoprotein precursor (LASgpc) or nucleoprotein (LCMnp) of lymphocytic choriomeningitis virus (LCMV), a mammarenavirus genetically closely related to LASV, conferred protection to recombinant LCMV-based surrogate mouse models of lethal LASV infection. Notably, robust LASgpc- and LCMnp-specific CD8+ T cell responses were detected in mRNA-LNP-immunized mice, whereas no virus-neutralizing activity was observed.


Subject(s)
Lassa Fever , Lassa virus , Lymphocytic choriomeningitis virus , Nanoparticles , Viral Vaccines , Animals , Female , Mice , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Disease Models, Animal , Glycoproteins/immunology , Glycoproteins/genetics , Lassa Fever/prevention & control , Lassa Fever/immunology , Lassa virus/immunology , Lassa virus/genetics , Liposomes , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/genetics , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nucleoproteins/immunology , Nucleoproteins/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , Viral Load , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
3.
Emerg Microbes Infect ; 13(1): 2341141, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38597241

ABSTRACT

The Natal multimammate mouse (Mastomys natalensis) is the host of Lassa mammarenavirus, causing Lassa haemorrhagic fever in West Africa. As there is currently no operational vaccine and therapeutic drugs are limited, we explored rodent control as an alternative to prevent Lassa virus spillover in Upper Guinea, where the disease is highly endemic in rural areas. In a seven-year experiment, we distributed rodenticides for 10-30 days once a year and, in the last year, added intensive snap trapping for three months in all the houses of one village. We also captured rodents both before and after the intervention period to assess their effectiveness by examining alterations in trapping success and infection rates (Lassa virus RNA and IgG antibodies). We found that both interventions reduced the rodent population by 74-92% but swiftly rebounded to pre-treatment levels, even already six months after the last snap-trapping control. Furthermore, while we observed that chemical control modestly decreased Lassa virus infection rates annually (a reduction of 5% in seroprevalence per year), the intensive trapping unexpectedly led to a significantly higher infection rate (from a seroprevalence of 28% before to 67% after snap trapping control). After seven years, we conclude that annual chemical control, alone or with intensive trapping, is ineffective and sometimes counterproductive in preventing Lassa virus spillover in rural villages. These unexpected findings may result from density-dependent breeding compensation following culling and the survival of a small percentage of chronically infected rodents that may spread the virus to a new susceptible generation of mice.


Subject(s)
Lassa Fever , Lassa virus , Mice , Animals , Lassa virus/genetics , Guinea/epidemiology , Rodent Control , Seroepidemiologic Studies , Disease Reservoirs , Lassa Fever/epidemiology , Lassa Fever/prevention & control , Murinae , Africa, Western/epidemiology
4.
BMC Infect Dis ; 24(1): 314, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38486143

ABSTRACT

BACKGROUND: Lassa fever is a hemorrhagic disease caused by Lassa virus (LASV), which has been classified by the World Health Organization as one of the top infectious diseases requiring prioritized research. Previous studies have provided insights into the classification and geographic characteristics of LASV lineages. However, the factor of the distribution and evolution characteristics and phylodynamics of the virus was still limited. METHODS: To enhance comprehensive understanding of LASV, we employed phylogenetic analysis, reassortment and recombination detection, and variation evaluation utilizing publicly available viral genome sequences. RESULTS: The results showed the estimated the root of time of the most recent common ancestor (TMRCA) for large (L) segment was approximately 634 (95% HPD: [385879]), whereas the TMRCA for small (S) segment was around 1224 (95% HPD: [10301401]). LASV primarily spread from east to west in West Africa through two routes, and in route 2, the virus independently spread to surrounding countries through Liberia, resulting in a wider spread of LASV. From 1969 to 2018, the effective population size experienced two significant increased, indicating the enhanced genetic diversity of LASV. We also found the evolution rate of L segment was faster than S segment, further results showed zinc-binding protein had the fastest evolution rate. Reassortment events were detected in multiple lineages including sub-lineage IIg, while recombination events were observed within lineage V. Significant amino acid changes in the glycoprotein precursor of LASV were identified, demonstrating sequence diversity among lineages in LASV. CONCLUSION: This study comprehensively elucidated the transmission and evolution of LASV in West Africa, providing detailed insights into reassortment events, recombination events, and amino acid variations.


Subject(s)
Lassa Fever , Lassa virus , Humans , Lassa virus/genetics , Phylogeny , Lassa Fever/epidemiology , Amino Acids , Liberia
5.
PLoS Negl Trop Dis ; 18(2): e0011984, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38421939

ABSTRACT

West African Mastomys rodents are the primary reservoir of the zoonotic Lassa virus (LASV). The virus causes haemorrhagic Lassa fever and considerable mortality in humans. To date, the role of Mastomys immunogenetics in resistance to, and persistence of, LASV infections is largely unknown. Here, we investigated the role of Major Histocompatibility Complex class I (MHC-I) on LASV infection status (i.e., active vs. cleared infection, determined via PCR and an immunofluorescence assay on IgG antibodies, respectively) in Mastomys natalensis and M. erythroleucus sampled within southwestern Nigeria. We identified more than 190 and 90 MHC-I alleles by Illumina high throughput-sequencing in M. natalensis and M. erythroleucus, respectively, with different MHC allele compositions and frequencies between LASV endemic and non-endemic sites. In M. natalensis, the MHC allele ManaMHC-I*006 was negatively associated with active infections (PCR-positive) and positively associated with cleared infections (IgG-positive) simultaneously, suggesting efficient immune responses that facilitate LASV clearance in animals carrying this allele. Contrarily, alleles ManaMHC-I*008 and ManaMHC-I*021 in M. natalensis, and MaerMHC-I*008 in M. erythroleucus, were positively associated with active infection, implying susceptibility. Alleles associated with susceptibility shared a glutamic acid at the positively selected codon 57, while ManaMHC-I*006 featured an arginine. There was no link between number of MHC alleles per Mastomys individual and LASV prevalence. Thus, specific alleles, but not MHC diversity per se, seem to mediate antibody responses to viremia. We conclude that co-evolution with LASV likely shaped the MHC-I diversity of the main LASV reservoirs in southwestern Nigeria, and that information on reservoir immunogenetics may hold insights into transmission dynamics and zoonotic spillover risks.


Subject(s)
Lassa Fever , Lassa virus , Animals , Humans , Lassa virus/genetics , Alleles , Antibody Formation , Kinetics , Lassa Fever/genetics , Lassa Fever/veterinary , Immunoglobulin G
6.
Nat Microbiol ; 9(3): 751-762, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38326571

ABSTRACT

Infection with Lassa virus (LASV) can cause Lassa fever, a haemorrhagic illness with an estimated fatality rate of 29.7%, but causes no or mild symptoms in many individuals. Here, to investigate whether human genetic variation underlies the heterogeneity of LASV infection, we carried out genome-wide association studies (GWAS) as well as seroprevalence surveys, human leukocyte antigen typing and high-throughput variant functional characterization assays. We analysed Lassa fever susceptibility and fatal outcomes in 533 cases of Lassa fever and 1,986 population controls recruited over a 7 year period in Nigeria and Sierra Leone. We detected genome-wide significant variant associations with Lassa fever fatal outcomes near GRM7 and LIF in the Nigerian cohort. We also show that a haplotype bearing signatures of positive selection and overlapping LARGE1, a required LASV entry factor, is associated with decreased risk of Lassa fever in the Nigerian cohort but not in the Sierra Leone cohort. Overall, we identified variants and genes that may impact the risk of severe Lassa fever, demonstrating how GWAS can provide insight into viral pathogenesis.


Subject(s)
Lassa Fever , Humans , Lassa Fever/genetics , Lassa Fever/diagnosis , Lassa Fever/epidemiology , Genome-Wide Association Study , Seroepidemiologic Studies , Lassa virus/genetics , Fever , Human Genetics
7.
Emerg Microbes Infect ; 13(1): 2294859, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38088796

ABSTRACT

Identification of the diverse animal hosts responsible for spill-over events from animals to humans is crucial for comprehending the transmission patterns of emerging infectious diseases, which pose significant public health risks. To better characterize potential animal hosts of Lassa virus (LASV), we assessed domestic and non-domestic animals from 2021-2022 in four locations in southern Nigeria with reported cases of Lassa fever (LF). Birds, lizards, and domestic mammals (dogs, pigs, cattle and goats) were screened using RT-qPCR, and whole genome sequencing was performed for lineage identification on selected LASV positive samples. Animals were also screened for exposure to LASV by enzyme-linked immunosorbent assay (ELISA). Among these animals, lizards had the highest positivity rate by PCR. Genomic sequencing of samples in most infected animals showed sub-lineage 2 g of LASV. Seropositivity was highest among cattle and lowest in pigs. Though the specific impact these additional hosts may have in the broader virus-host context are still unknown - specifically relating to pathogen diversity, evolution, and transmission - the detection of LASV in non-rodent hosts living in proximity to confirmed human LF cases suggests their involvement during transmission as potential reservoirs. Additional epidemiological data comparing viral genomes from humans and animals, as well as those circulating within the environment will be critical in understanding LASV transmission dynamics and will ultimately guide the development of countermeasures for this zoonotic health threat.


Subject(s)
Lassa Fever , Lassa virus , Humans , Animals , Cattle , Dogs , Swine , Lassa virus/genetics , Lassa Fever/epidemiology , Lassa Fever/veterinary , Lassa Fever/genetics , Nigeria/epidemiology , Genome, Viral , Public Health , Mammals
8.
Emerg Microbes Infect ; 13(1): 2290834, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38047354

ABSTRACT

The spread of Lassa virus (LASV) in Guinea, Liberia and Sierra Leone, which together are named the Mano River Union (MRU) area, was examined phylogeographically. To provide a reliable evolutionary scenario, new rodent-derived, whole LASV sequences were included. These were generated by metatranscriptomic next-generation sequencing from rodents sampled between 2003 and 2020 in 21 localities of Guinea and Sierra Leone. An analysis was performed using BEAST to perform continuous phylogeographic inference and EvoLaps v36 to visualize spatio-temporal spread. LASV was identified as expected in its primary host reservoir, the Natal multimammate mouse (Mastomys natalensis), and also in two Guinean multimammate mice (Mastomys erythroleucus) in northern Sierra Leone and two rusty-bellied brush-furred mice (Lophuromys sikapusi) in southern Sierra Leone. This finding is consistent with the latter two species being secondary host reservoirs. The strains in these three species were very closely related in LASV lineage IV. Phylogenetic analysis indicated that the most recent common ancestor of lineage IV existed 316-374 years ago and revealed distinct, well-supported clades from Sierra Leone (Bo, Kabala and Kenema), Guinea (Faranah, Kissidougou-Guekedou and Macenta) and Liberia (Phebe-Ganta). The phylogeographic scenario suggests southern Guinea as the point of origin of LASV in the MRU area, with subsequent spread to towards Mali, Liberia and Sierra Leone at a mean speed of 1.6 to 1.1 km/year.


Subject(s)
Lassa Fever , Lassa virus , Mice , Animals , Lassa virus/genetics , Lassa Fever/epidemiology , Phylogeny , Africa, Western/epidemiology , Murinae
9.
Methods Mol Biol ; 2733: 115-131, 2024.
Article in English | MEDLINE | ID: mdl-38064030

ABSTRACT

Several mammarenaviruses cause hemorrhagic fever (HF) disease in humans and pose a significant public health problem in their endemic regions. The Old World (OW) mammarenavirus Lassa virus (LASV) is estimated to infect several hundred thousand people yearly in West Africa, resulting in high numbers of Lassa fever (LF) cases, a disease associated with high morbidity and mortality. No licensed vaccines are available to combat LASV infection, and anti-LASV drug therapy is limited to the off-label use of ribavirin whose efficacy remains controversial. The development of reverse genetics approaches has provided investigators with a powerful approach for the investigation of the molecular, cell biology and pathogenesis of mammarenaviruses. The use of cell-based minigenome systems has allowed examining the cis- and trans-acting factors involved in viral genome replication and gene transcription, assembly, and budding, which has facilitated the identification of several anti-mammarenavirus candidate drugs. Likewise, it is possible now to rescue infectious recombinant mammarenaviruses from cloned cDNAs containing predetermined mutations in their genomes to investigate virus-host interactions and mechanisms of viral pathogenesis. Reverse genetics have also allowed the generation of mammarenaviruses expressing foreign genes to facilitate virus detection, to identify antiviral drugs, and to generate live-attenuated vaccine (LAV) candidates. Likewise, reverse genetics techniques have allowed the generation of single-cycle infectious, reporter-expressing mammarenaviruses to study some aspects of the biology of HF-causing human mammarenavirus without the need of high security biocontainment laboratories. In this chapter, we describe the experimental procedures to generate recombinant (r)LASV using state-of-the-art plasmid-based reverse genetics.


Subject(s)
Arenaviridae , Hemorrhagic Fevers, Viral , Lassa Fever , Humans , Lassa virus/genetics , Reverse Genetics/methods , Arenaviridae/genetics , Plasmids/genetics
10.
J Am Chem Soc ; 145(51): 27958-27974, 2023 12 27.
Article in English | MEDLINE | ID: mdl-38104324

ABSTRACT

Lassa virus is a negative-strand RNA virus with only four structural proteins that causes periodic outbreaks in West Africa. The nucleoprotein (NP) encapsidates the viral genome, forming ribonucleoprotein complexes (RNPs) together with the viral RNA and the L protein. RNPs must be continuously restructured during viral genome replication and transcription. The Z protein is important for membrane recruitment of RNPs, viral particle assembly, and budding and has also been shown to interact with the L protein. However, the interaction of NP, viral RNA, and Z is poorly understood. Here, we characterize the interactions between Lassa virus NP, Z, and RNA using structural mass spectrometry. We identify the presence of RNA as the driver for the disassembly of ring-like NP trimers, a storage form, into monomers to subsequently form higher order RNA-bound NP assemblies. We locate the interaction site of Z and NP and demonstrate that while NP binds Z independently of the presence of RNA, this interaction is pH-dependent. These data improve our understanding of RNP assembly, recruitment, and release in Lassa virus.


Subject(s)
Lassa virus , Ribonucleoproteins , Lassa virus/genetics , Lassa virus/metabolism , Ribonucleoproteins/chemistry , Nucleoproteins , Virus Assembly , RNA, Viral/genetics , RNA, Viral/metabolism
11.
Nat Commun ; 14(1): 5603, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37699929

ABSTRACT

Lassa virus is a member of the Arenaviridae family, which causes human infections ranging from asymptomatic to severe hemorrhagic disease with a high case fatality rate. We have designed and generated lipid nanoparticle encapsulated, modified mRNA vaccines that encode for the wild-type Lassa virus strain Josiah glycoprotein complex or the prefusion stabilized conformation of the Lassa virus glycoprotein complex. Hartley guinea pigs were vaccinated with two 10 µg doses, 28 days apart, of either construct. Vaccination induced strong binding antibody responses, specific to the prefusion conformation of glycoprotein complex, which were significantly higher in the prefusion stabilized glycoprotein complex construct group and displayed strong Fc-mediated effects. However, Lassa virus-neutralizing antibody activity was detected in some but not all animals. Following the challenge with a lethal dose of the Lassa virus, all vaccinated animals were protected from death and severe disease. Although the definitive mechanism of protection is still unknown, and assessment of the cell-mediated immune response was not investigated in this study, these data demonstrate the promise of mRNA as a vaccine platform against the Lassa virus and that protection against Lassa virus can be achieved in the absence of virus-neutralizing antibodies.


Subject(s)
Arenaviridae , Lassa virus , Humans , Guinea Pigs , Animals , Lassa virus/genetics , Antibodies, Neutralizing , mRNA Vaccines , Glycoproteins
12.
Nat Commun ; 14(1): 4693, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37542071

ABSTRACT

Effective infectious disease surveillance in high-risk regions is critical for clinical care and pandemic preemption; however, few clinical diagnostics are available for the wide range of potential human pathogens. Here, we conduct unbiased metagenomic sequencing of 593 samples from febrile Nigerian patients collected in three settings: i) population-level surveillance of individuals presenting with symptoms consistent with Lassa Fever (LF); ii) real-time investigations of outbreaks with suspected infectious etiologies; and iii) undiagnosed clinically challenging cases. We identify 13 distinct viruses, including the second and third documented cases of human blood-associated dicistrovirus, and a highly divergent, unclassified dicistrovirus that we name human blood-associated dicistrovirus 2. We show that pegivirus C is a common co-infection in individuals with LF and is associated with lower Lassa viral loads and favorable outcomes. We help uncover the causes of three outbreaks as yellow fever virus, monkeypox virus, and a noninfectious cause, the latter ultimately determined to be pesticide poisoning. We demonstrate that a local, Nigerian-driven metagenomics response to complex public health scenarios generates accurate, real-time differential diagnoses, yielding insights that inform policy.


Subject(s)
Lassa Fever , Viruses , Humans , Nigeria/epidemiology , Metagenomics , Lassa Fever/diagnosis , Lassa Fever/epidemiology , Lassa virus/genetics , Viruses/genetics
13.
West Afr J Med ; 40(7): 684-688, 2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37515581

ABSTRACT

BACKGROUND: Lassa fever is an acute hemorrhagic viral disease caused by the Lassa virus. The Lassa virus belongs to the Arenaviridae family of RNA viruses. On 05/04/2016; two cases of Lassa fever were reported from Katsina State with the date of presentation of the first case on 23/03/2016 and 27/03/ 2016 for the second case. We investigated the outbreak to identify the agent and the source and propose recommendations as well as to assess the practice of infection, prevention and control (IPC). METHODS: We used descriptive study to describe contact tracing and facility assessment. We described the outbreak by time, place, and person. We defined a case using established guidelines and line-listed the contacts. We conducted IPC facility check in the state. Blood specimens were collected for Lassa fever detection. Microsoft Excel and Epi-info version 7.1.6 were used for data analysis. RESULTS: The index case of Lassa fever in Katsina State was seen on 23/03/2016 with a travel history from Kaduna State. The second case had contact with a positive Lassa fever case from Gwagwalada, Federal Capital Territory (FCT). A total of 82 contacts were line listed (9 developed Lassa fever). The case fatality rate was 27.3%. IPC checklist revealed 37.5% of the health facilities lacked personal protective equipment and safety boxes, 25% lacked isolation wards, and none had chlorine solution. Overall, 61% of personnel had poor knowledge of Lassa fever, 31% had fair knowledge and 8% had good knowledge. CONCLUSION: A multiple-source epidemic with sources of primary infection from outside Katsina state was noted. Most of the health facilities assessed lack basic IPC materials and knowledge on Lassa fever which should be addressed.


CONTEXTE: La fièvre de Lassa est une maladie virale hémorragique aiguë causée par le virus de Lassa. Le virus Lassa appartient à la famille des Arenaviridae, des virus à ARN. Le 05/04/2016 ; deux cas de fièvre de Lassa ont été signalés dans l'État de Katsina avec la date de présentation du premier cas le 23/03/2016 et le 27/03/2016 pour le second cas. Nous avons enquêté sur cette épidémie pour identifier l'agent et la source et proposer des recommandations ainsi que pour évaluer la pratique de l'infection, de la prévention et du contrôle (IPC). MÉTHODES: Nous avons utilisé une étude descriptive pour décrire la recherche des contacts et l'évaluation des installations. Nous avons décrit l'épidémie en fonction de la date, du lieu et de la personne. Nous avons défini un cas à l'aide de lignes directrices établies et dressé une liste des contacts. Nous avons vérifié les installations de CIP dans l'État. Des échantillons de sang ont été prélevés pour la détection de la fièvre de Lassa. Microsoft Excel et Epi-info version 7.1.6 ont été utilisés pour l'analyse des données. RÉSULTATS: Le cas index de fièvre de Lassa dans l'État de Katsina a été observé le 23/03/2016 avec des antécédents de voyage en provenance de l'État de Kaduna. Le deuxième cas a été en contact avec un cas positif de fièvre de Lassa à Gwagwalada, dans le Territoire de la capitale fédérale (FCT). Au total, 82 contacts ont été répertoriés (9 ont développé une fièvre de Lassa). Le taux de létalité était de 27,3%. La liste de contrôle IPC a révélé que 37,5 % des établissements de santé manquaient d'équipements de protection individuelle et de boîtes de sécurité, que 25 % n'avaient pas de salles d'isolement et qu'aucun n'avait de solution chlorée. Dans l'ensemble, 61 % du personnel avait une mauvaise connaissance de la fièvre de Lassa, 31 % une connaissance moyenne et 8 % une bonne connaissance. CONCLUSION: Une épidémie à sources multiples avec des sources d'infection primaire en dehors de l'État de Katsina a été observée. La plupart des établissements de santé évalués manquent de matériel IPC de base et de connaissances sur la fièvre de Lassa, ce qui devrait être corrigé. Mots clés: Épidémiologie, Contrôle des infections, Katsina, Épidémie, Fièvre de Lassa.


Subject(s)
Epidemics , Lassa Fever , Humans , Lassa Fever/epidemiology , Lassa Fever/prevention & control , Nigeria/epidemiology , Disease Outbreaks/prevention & control , Lassa virus/genetics
14.
Emerg Microbes Infect ; 12(1): 2219350, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37288752

ABSTRACT

We phylogenetically compared sequences of the zoonotic Lassa virus (LASV) obtained from Mastomys rodents in seven localities across the highly endemic Edo and Ondo States within Nigeria. Sequencing 1641 nt from the S segment of the virus genome, we resolved clades within lineage II that were either limited to Ebudin and Okhuesan in Edo state (2g-beta) or along Owo-Okeluse-Ifon in Ondo state (2g-gamma). We also found clades within Ekpoma, a relatively large cosmopolitan town in Edo state, that extended into other localities within Edo (2g-alpha) and Ondo (2g-delta). LASV variants from M. natalensis within Ebudin and Ekpoma in Edo State (dated approximately 1961) were more ancient compared to those from Ondo state (approximately 1977), suggesting a broadly east-west virus migration across south-western Nigeria; a pattern not always consistent with LASV sequences derived from humans in the same localities. Additionally, in Ebudin and Ekpoma, LASV sequences between M. natalensis and M. erythroleucus were interspersed on the phylogenetic tree, but those from M. erythroleucus were estimated to emerge more recently (approximately 2005). Overall, our results show that LASV amplification in certain localities (reaching a prevalence as high as 76% in Okeluse), anthropogenically-aided spread of rodent-borne variants amidst the larger towns (involving communal accommodation such as student hostels), and virus-exchange between syntopic M. natalensis and M. erythroleucus rodents (as the latter, a savanna species, encroaches southward into the degraded forest) pose perpetual zoonotic hazard across the Edo-Ondo Lassa fever belt, threatening to accelerate the dissemination of the virus into non endemic areas.


Subject(s)
Lassa Fever , Lassa virus , Humans , Mice , Animals , Lassa virus/genetics , Nigeria/epidemiology , Phylogeny , Lassa Fever/epidemiology , Lassa Fever/veterinary , Murinae
15.
Curr Top Microbiol Immunol ; 440: 165-192, 2023.
Article in English | MEDLINE | ID: mdl-37106159

ABSTRACT

Lassa fever is caused by Lassa virus (LASV), an Old World Mammarenavirus that is carried by Mastomys natalensis and other rodents. It is endemic in Sierra Leone, Nigeria, and other countries in West Africa. The clinical presentation of LASV infection is heterogenous varying from an inapparent or mild illness to a fatal hemorrhagic fever. Exposure to LASV is usually through contact with rodent excreta. After an incubation period of 1-3 weeks, initial symptoms such as fever, headache, and fatigue develop that may progress to sore throat, retrosternal chest pain, conjunctival injection, vomiting, diarrhea, and abdominal pain. Severe illness, including hypotension, shock, and multiorgan failure, develops in a minority of patients. Patient demographics and case fatality rates are distinctly different in Sierra Leone and Nigeria. Laboratory diagnosis relies on the detection of LASV antigens or genomic RNA. LASV-specific immunoglobulin G and M assays can also contribute to clinical management. The mainstay of treatment for Lassa fever is supportive care. The nucleoside analog ribavirin is commonly used to treat acute Lassa fever but is considered useful only if treatment is begun early in the disease course. Drugs in development, including a monoclonal antibody cocktail, have the potential to impact the management of Lassa fever.


Subject(s)
Lassa Fever , Humans , Lassa Fever/diagnosis , Lassa Fever/drug therapy , Lassa Fever/epidemiology , Lassa virus/genetics , Africa, Western , Sierra Leone/epidemiology , Antibodies, Viral
16.
Sci Rep ; 13(1): 6506, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37081092

ABSTRACT

The design of minimum CRISPR RNA (crRNA) sets for detection of diverse RNA targets using sequence degeneracy has not been systematically addressed. We tested candidate degenerate Cas13a crRNA sets designed for detection of diverse RNA targets (Lassa virus). A decision tree machine learning (ML) algorithm (RuleFit) was applied to define the top attributes that determine the specificity of degenerate crRNAs to elicit collateral nuclease activity. Although the total number of mismatches (0-4) is important, the specificity depends as well on the spacing of mismatches, and their proximity to the 5' end of the spacer. We developed a predictive algorithm for design of candidate degenerate crRNA sets, allowing improved discrimination between "included" and "excluded" groups of related target sequences. A single degenerate crRNA set adhering to these rules detected representatives of all Lassa lineages. Our general ML approach may be applied to the design of degenerate crRNA sets for any CRISPR/Cas system.


Subject(s)
Lassa virus , RNA , RNA/metabolism , Lassa virus/genetics , RNA Processing, Post-Transcriptional , CRISPR-Cas Systems/genetics
17.
Curr Top Microbiol Immunol ; 440: 147-164, 2023.
Article in English | MEDLINE | ID: mdl-37100973

ABSTRACT

Lassa virus (LASV) is the causative agent of Lassa fever, an often-fatal hemorrhagic fever that is endemic in West Africa. LASV virions are enveloped and contain two single-stranded RNA genome segments. Both segments are ambisense and encode two proteins. The nucleoprotein associates with viral RNAs forming ribonucleoprotein complexes. The glycoprotein complex mediates viral attachment and entry. The Zinc protein serves as the matrix protein. Large is a polymerase that catalyzes viral RNA transcription and replication. LASV virion entry occurs via a clathrin-independent endocytic pathway usually involving alpha-dystroglycan and lysosomal associated membrane protein 1 as surface and intracellular receptors, respectively. Advances in understanding LASV structural biology and replication have facilitated development of promising vaccine and drug candidates.


Subject(s)
Lassa Fever , Lassa virus , Humans , Lassa virus/genetics , Lassa virus/metabolism , Lassa Fever/prevention & control , Biology , Africa, Western
18.
Virol Sin ; 38(3): 380-386, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37059226

ABSTRACT

The Lassa virus (LASV) is endemic in West Africa and causes severe hemorrhagic Lassa fever in humans. The glycoprotein complex (GPC) of LASV is highly glycosylation-modified, with 11 â€‹N-glycosylation sites. All 11 N-linked glycan chains play critical roles in GPC cleavage, folding, receptor binding, membrane fusion, and immune evasion. In this study, we focused on the first glycosylation site because its deletion mutant (N79Q) results in an unexpected enhanced membrane fusion, whereas it exerts little effect on GPC expression, cleavage, and receptor binding. Meanwhile, the pseudotype virus bearing GPCN79Q was more sensitive to the neutralizing antibody 37.7H and was attenuated in virulence. Exploring the biological functions of the key glycosylation site on LASV GPC will help elucidate the mechanism of LASV infection and provide strategies for the development of attenuated vaccines against LASV infection.


Subject(s)
Lassa Fever , Lassa virus , Humans , Lassa virus/genetics , Glycosylation , Membrane Fusion , Glycoproteins/genetics , Lassa Fever/prevention & control
19.
Adv Exp Med Biol ; 1407: 279-297, 2023.
Article in English | MEDLINE | ID: mdl-36920703

ABSTRACT

Mammarenaviruses are classified into New World arenaviruses (NW) and Old World arenaviruses (OW). The OW arenaviruses include the first discovered mammarenavirus-lymphocytic choriomeningitis virus (LCMV) and the highly lethal Lassa virus (LASV). Mammarenaviruses are transmitted to human by rodents, resulting in severe acute infections and hemorrhagic fever. Pseudotyped viruses have been widely used as a tool in the study of mammarenaviruses. HIV-1, SIV, FIV-based lentiviral vectors, VSV-based vectors, MLV-based vectors, and reverse genetic approaches have been applied in the construction of pseudotyped mammarenaviruses. Pseudotyped mammarenaviruses are commonly used in receptor research, neutralizing antibody detection, inhibitor screening, viral virulence studies, functional analysis of N-linked glycans, and studies of viral infection, endocytosis, and fusion mechanisms.


Subject(s)
Arenaviridae , Arenaviruses, New World , Humans , Arenaviridae/genetics , Viral Pseudotyping , Lymphocytic choriomeningitis virus/genetics , Arenaviruses, New World/genetics , Lassa virus/genetics
20.
Curr Top Microbiol Immunol ; 440: 87-109, 2023.
Article in English | MEDLINE | ID: mdl-33861373

ABSTRACT

Lassa fever (LF) is a lethal hemorrhagic disease primarily concentrated in the tropical savannah regions of Nigeria and the Mano River Union countries of Sierra Leone, Liberia, and Guinea. Endemic hotspots within these countries have had recurrent exposure to Lassa virus (LASV) via continual spillover from the host reservoir Mastomys natalensis. Increased trade and travel throughout the region have spread the virus to previously unexposed countries, including Ghana, Benin, Mali, and Côte d'Ivoire. In the absence of effective treatment or vaccines to LASV, preventative measures against Lassa fever rely heavily on reducing or eliminating rodent exposure, increasing the knowledge base surrounding the virus and disease in communities, and diminishing the stigmas faced by Lassa fever survivors.


Subject(s)
Lassa Fever , Animals , Lassa Fever/epidemiology , Lassa virus/genetics , Murinae
SELECTION OF CITATIONS
SEARCH DETAIL
...