Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
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
2.
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
3.
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
4.
Curr Top Microbiol Immunol ; 440: 23-65, 2023.
Article in English | MEDLINE | ID: mdl-32418034

ABSTRACT

In a pattern repeated across a range of ecological niches, arenaviruses have evolved a compact four-gene genome to orchestrate a complex life cycle in a narrow range of susceptible hosts. A number of mammalian arenaviruses cross-infect humans, often causing a life-threatening viral hemorrhagic fever. Among this group of geographically bound zoonoses, Lassa virus has evolved a unique niche that leads to significant and sustained human morbidity and mortality. As a biosafety level 4 pathogen, direct study of the pathogenesis of Lassa virus is limited by the sparse availability, high operating costs, and technical restrictions of the high-level biocontainment laboratories required for safe experimentation. In this chapter, we introduce the relationship between genome structure and the life cycle of Lassa virus and outline reverse genetic approaches used to probe and describe functional elements of the Lassa virus genome. We then review the tools used to obtain viral genomic sequences used for phylogeny and molecular diagnostics, before shifting to a population perspective to assess the contributions of phylogenetic analysis in understanding the evolution and ecology of Lassa virus in West Africa. We finally consider the future outlook and clinical applications for genetic study of Lassa virus.


Subject(s)
Lassa Fever , Lassa virus , Animals , Humans , Lassa virus/genetics , Lassa Fever/epidemiology , Lassa Fever/genetics , Phylogeny , Africa, Western/epidemiology , Zoonoses , Mammals
5.
PLoS Pathog ; 17(10): e1009966, 2021 10.
Article in English | MEDLINE | ID: mdl-34634087

ABSTRACT

Nigeria continues to experience ever increasing annual outbreaks of Lassa fever (LF). The World Health Organization has recently declared Lassa virus (LASV) as a priority pathogen for accelerated research leading to a renewed international effort to develop relevant animal models of disease and effective countermeasures to reduce LF morbidity and mortality in endemic West African countries. A limiting factor in evaluating medical countermeasures against LF is a lack of well characterized animal models outside of those based on infection with LASV strain Josiah originating form Sierra Leone, circa 1976. Here we genetically characterize five recent LASV isolates collected from the 2018 outbreak in Nigeria. Three isolates were further evaluated in vivo and despite being closely related and from the same spatial / geographic region of Nigeria, only one of the three isolates proved lethal in strain 13 guinea pigs and non-human primates (NHP). Additionally, this isolate exhibited atypical pathogenesis characteristics in the NHP model, most notably respiratory failure, not commonly described in hemorrhagic cases of LF. These results suggest that there is considerable phenotypic heterogeneity in LASV infections in Nigeria, which leads to a multitude of pathogenesis characteristics that could account for differences between subclinical and lethal LF infections. Most importantly, the development of disease models using currently circulating LASV strains in West Africa are critical for the evaluation of potential vaccines and medical countermeasures.


Subject(s)
Disease Models, Animal , Lassa Fever/genetics , Lassa virus/genetics , Animals , Disease Outbreaks , Female , Guinea Pigs , Humans , Macaca fascicularis , Male , Nigeria , Phylogeny
6.
Viruses ; 13(8)2021 08 13.
Article in English | MEDLINE | ID: mdl-34452470

ABSTRACT

While investigating a signal of adaptive evolution in humans at the gene LARGE, we encountered an intriguing finding by Dr. Stefan Kunz that the gene plays a critical role in Lassa virus binding and entry. This led us to pursue field work to test our hypothesis that natural selection acting on LARGE-detected in the Yoruba population of Nigeria-conferred resistance to Lassa Fever in some West African populations. As we delved further, we conjectured that the "emerging" nature of recently discovered diseases like Lassa fever is related to a newfound capacity for detection, rather than a novel viral presence, and that humans have in fact been exposed to the viruses that cause such diseases for much longer than previously suspected. Dr. Stefan Kunz's critical efforts not only laid the groundwork for this discovery, but also inspired and catalyzed a series of events that birthed Sentinel, an ambitious and large-scale pandemic prevention effort in West Africa. Sentinel aims to detect and characterize deadly pathogens before they spread across the globe, through implementation of its three fundamental pillars: Detect, Connect, and Empower. More specifically, Sentinel is designed to detect known and novel infections rapidly, connect and share information in real time to identify emerging threats, and empower the public health community to improve pandemic preparedness and response anywhere in the world. We are proud to dedicate this work to Stefan Kunz, and eagerly invite new collaborators, experts, and others to join us in our efforts.


Subject(s)
Disaster Planning , Lassa Fever/epidemiology , Lassa virus/physiology , Africa, Western/epidemiology , Disaster Planning/methods , Humans , Lassa Fever/genetics , Lassa Fever/prevention & control , Lassa Fever/virology , Lassa virus/genetics , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/immunology , Nigeria/epidemiology , Pandemics , Polymorphism, Genetic , Receptors, Virus/genetics , Receptors, Virus/immunology
7.
Viruses ; 13(2)2021 01 30.
Article in English | MEDLINE | ID: mdl-33573250

ABSTRACT

Defective interfering particles (DIPs) are naturally occurring products during virus replication in infected cells. DIPs contain defective viral genomes (DVGs) and interfere with replication and propagation of their corresponding standard viral genomes by competing for viral and cellular resources, as well as promoting innate immune antiviral responses. Consequently, for many different viruses, including mammarenaviruses, DIPs play key roles in the outcome of infection. Due to their ability to broadly interfere with viral replication, DIPs are attractive tools for the development of a new generation of biologics to target genetically diverse and rapidly evolving viruses. Here, we provide evidence that in cells infected with the Lassa fever (LF) vaccine candidate ML29, a reassortant that carries the nucleoprotein (NP) and glycoprotein (GP) dominant antigens of the pathogenic Lassa virus (LASV) together with the L polymerase and Z matrix protein of the non-pathogenic genetically related Mopeia virus (MOPV), L-derived truncated RNA species are readily detected following infection at low multiplicity of infection (MOI) or in persistently-infected cells originally infected at high MOI. In the present study, we show that expression of green fluorescent protein (GFP) driven by a tri-segmented form of the mammarenavirus lymphocytic choriomeningitis virus (r3LCMV-GFP/GFP) was strongly inhibited in ML29-persistently infected cells, and that the magnitude of GFP suppression was dependent on the passage history of the ML29-persistently infected cells. In addition, we found that DIP-enriched ML29 was highly attenuated in immunocompetent CBA/J mice and in Hartley guinea pigs. Likewise, STAT-1-/- mice, a validated small animal model for human LF associated hearing loss sequelae, infected with DIP-enriched ML29 did not exhibit any hearing abnormalities throughout the observation period (62 days).


Subject(s)
Lassa Fever/prevention & control , Lassa virus/immunology , Viral Vaccines/immunology , Animals , Female , Genome, Viral , Guinea Pigs , Humans , Lassa Fever/genetics , Lassa Fever/immunology , Lassa Fever/virology , Lassa virus/genetics , Lassa virus/physiology , Mice , Mice, Inbred CBA , RNA, Viral/genetics , RNA, Viral/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virus Replication
8.
Neurosci Lett ; 740: 135427, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33069812

ABSTRACT

The STAT1 knock-out (KO) mouse is a frequently used transgenic immunodeficient strain to model human viral and bacterial diseases. The Lassa fever model was established in the STAT1 KO mice mimicking phenotypes seen in human patients including deafness in survivors. This model develops hearing loss at high prevalence and is a valuable tool to investigate viral infection-induced hearing loss. However, Lassa virus is a highly contagious and regulated agent requiring the unique logistics of the biosafety level 4 posing limitations for experimental work. Therefore, we did a detailed auditory analysis of the STAT1 KO mice to assess baseline auditory function in preparation for further auditory behavioral studies. Auditory brainstem response and distortion product otoacoustic emission tests were performed on males and females of the STAT1 KO mice and was compared to 129S6/SvEv wild type (WT) mice. The male WT mice had the best auditory performance and the female WT mice had the worst hearing performance. The male and female STAT1 KO mice had similar auditory performance to each other, which was intermediate between WT males and females. We conclude that both male and female STAT1 KO mice are suitable for studying viral infection-induced hearing loss.


Subject(s)
Hearing Loss/genetics , Lassa Fever/genetics , STAT1 Transcription Factor/genetics , Animals , Behavior, Animal , Disease Models, Animal , Disease Progression , Evoked Potentials, Auditory, Brain Stem , Female , Hearing Loss/etiology , Hearing Loss/physiopathology , Hearing Tests , Lassa Fever/physiopathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Otoacoustic Emissions, Spontaneous , Sex Characteristics
9.
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
10.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32817220

ABSTRACT

Lassa fever (LF) is a zoonotic viral hemorrhagic fever caused by Lassa virus (LASV), which is endemic to West African countries. Previous studies have suggested an important role for T-cell-mediated immunopathology in LF pathogenesis, but the mechanisms by which T cells influence disease severity and outcome are not well understood. Here, we present a multiparametric analysis of clinical immunology data collected during the 2017-2018 Lassa fever outbreak in Nigeria. During the acute phase of LF, we observed robust activation of the polyclonal T-cell repertoire, which included LASV-specific and antigenically unrelated T cells. However, severe and fatal LF cases were characterized by poor LASV-specific effector T-cell responses. Severe LF was also characterized by the presence of circulating T cells with homing capacity to inflamed tissues, including the gut mucosa. These findings in LF patients were recapitulated in a mouse model of LASV infection, in which mucosal exposure resulted in remarkably high lethality compared to skin exposure. Taken together, our findings indicate that poor LASV-specific T-cell responses and activation of nonspecific T cells with homing capacity to inflamed tissues are associated with severe LF.IMPORTANCE Lassa fever may cause severe disease in humans, in particular in areas of endemicity like Sierra Leone and Nigeria. Despite its public health importance, the pathophysiology of Lassa fever in humans is poorly understood. Here, we present clinical immunology data obtained in the field during the 2018 Lassa fever outbreak in Nigeria indicating that severe Lassa fever is associated with activation of T cells antigenically unrelated to Lassa virus and poor Lassa virus-specific effector T-cell responses. Mechanistically, we show that these bystander T cells express defined tissue homing signatures that suggest their recruitment to inflamed tissues and a putative role of these T cells in immunopathology. These findings open a window of opportunity to consider T-cell targeting as a potential postexposure therapeutic strategy against severe Lassa fever, a hypothesis that could be tested in relevant animal models, such as nonhuman primates.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Outbreaks , Intestinal Mucosa/immunology , Lassa Fever/immunology , Lassa virus/pathogenicity , Lymphocyte Activation , Adolescent , Adult , Aged , Animals , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Child , Child, Preschool , Female , Gene Expression Regulation , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Infant , Infant, Newborn , Integrin beta1/genetics , Integrin beta1/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Lassa Fever/genetics , Lassa Fever/mortality , Lassa Fever/virology , Lassa virus/growth & development , Lassa virus/immunology , Lysosomal-Associated Membrane Protein 1/genetics , Lysosomal-Associated Membrane Protein 1/immunology , Male , Mice , Middle Aged , Nigeria/epidemiology , Retrospective Studies , Severity of Illness Index , Skin/immunology , Skin/pathology , Skin/virology , Survival Analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
11.
J Virol ; 94(12)2020 06 01.
Article in English | MEDLINE | ID: mdl-32269122

ABSTRACT

Early and robust T cell responses have been associated with survival from Lassa fever (LF), but the Lassa virus-specific memory responses have not been well characterized. Regions within the virus surface glycoprotein (GPC) and nucleoprotein (NP) are the main targets of the Lassa virus-specific T cell responses, but, to date, only a few T cell epitopes within these proteins have been identified. We identified GPC and NP regions containing T cell epitopes and HLA haplotypes from LF survivors and used predictive HLA-binding algorithms to identify putative epitopes, which were then experimentally tested using autologous survivor samples. We identified 12 CD8-positive (CD8+) T cell epitopes, including epitopes common to both Nigerian and Sierra Leonean survivors. These data should be useful for the identification of dominant Lassa virus-specific T cell responses in Lassa fever survivors and vaccinated individuals as well as for designing vaccines that elicit cell-mediated immunity.IMPORTANCE The high morbidity and mortality associated with clinical cases of Lassa fever, together with the lack of licensed vaccines and limited and partially effective interventions, make Lassa virus (LASV) an important health concern in its regions of endemicity in West Africa. Previous infection with LASV protects from disease after subsequent exposure, providing a framework for designing vaccines to elicit similar protective immunity. Multiple major lineages of LASV circulate in West Africa, and therefore, ideal vaccine candidates should elicit immunity to all lineages. We therefore sought to identify common T cell epitopes between Lassa fever survivors from Sierra Leone and Nigeria, where distinct lineages circulate. We identified three such epitopes derived from highly conserved regions within LASV proteins. In this process, we also identified nine other T cell epitopes. These data should help in the design of an effective pan-LASV vaccine.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/chemistry , Lassa Fever/immunology , Lassa virus/immunology , Nucleoproteins/immunology , Viral Envelope Proteins/immunology , Adolescent , Amino Acid Sequence , Animals , Antibodies, Viral/biosynthesis , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/virology , Child , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , Haplotypes , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune Sera/analysis , Immunologic Memory , Lassa Fever/genetics , Lassa Fever/pathology , Lassa virus/pathogenicity , Male , Nigeria , Nucleoproteins/genetics , Sierra Leone , Survivors , Viral Envelope Proteins/genetics , Young Adult
12.
J Clin Invest ; 130(1): 539-551, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31820871

ABSTRACT

Recent occurrences of filoviruses and the arenavirus Lassa virus (LASV) in overlapping endemic areas of Africa highlight the need for a prophylactic vaccine that would confer protection against all of these viruses that cause lethal hemorrhagic fever (HF). We developed a quadrivalent formulation of VesiculoVax that contains recombinant vesicular stomatitis virus (rVSV) vectors expressing filovirus glycoproteins and that also contains a rVSV vector expressing the glycoprotein of a lineage IV strain of LASV. Cynomolgus macaques were vaccinated twice with the quadrivalent formulation, followed by challenge 28 days after the boost vaccination with each of the 3 corresponding filoviruses (Ebola, Sudan, Marburg) or a heterologous contemporary lineage II strain of LASV. Serum IgG and neutralizing antibody responses specific for all 4 glycoproteins were detected in all vaccinated animals. A modest and balanced cell-mediated immune response specific for the glycoproteins was also detected in most of the vaccinated macaques. Regardless of the level of total glycoprotein-specific immune response detected after vaccination, all immunized animals were protected from disease and death following lethal challenges. These findings indicate that vaccination with attenuated rVSV vectors each expressing a single HF virus glycoprotein may provide protection against those filoviruses and LASV most commonly responsible for outbreaks of severe HF in Africa.


Subject(s)
Antibodies, Viral/immunology , Genetic Vectors , Immunoglobulin G/immunology , Lassa Fever/prevention & control , Lassa virus/immunology , Vesiculovirus , Viral Vaccines/immunology , Animals , Humans , Lassa Fever/genetics , Lassa Fever/immunology , Lassa virus/genetics , Macaca fascicularis , Viral Vaccines/genetics
13.
Emerg Microbes Infect ; 8(1): 1511-1523, 2019.
Article in English | MEDLINE | ID: mdl-31631785

ABSTRACT

Interferons (IFNs) control viral infections by inducing expression of IFN-stimulated genes (ISGs) that restrict distinct steps of viral replication. We report herein that gamma-interferon-inducible lysosomal thiol reductase (GILT), a lysosome-associated ISG, restricts the infectious entry of selected enveloped RNA viruses. Specifically, we demonstrated that GILT was constitutively expressed in lung epithelial cells and fibroblasts and its expression could be further induced by type II interferon. While overexpression of GILT inhibited the entry mediated by envelope glycoproteins of SARS coronavirus (SARS-CoV), Ebola virus (EBOV) and Lassa fever virus (LASV), depletion of GILT enhanced the entry mediated by these viral envelope glycoproteins. Furthermore, mutations that impaired the thiol reductase activity or disrupted the N-linked glycosylation, a posttranslational modification essential for its lysosomal localization, largely compromised GILT restriction of viral entry. We also found that the induction of GILT expression reduced the level and activity of cathepsin L, which is required for the entry of these RNA viruses in lysosomes. Our data indicate that GILT is a novel antiviral ISG that specifically inhibits the entry of selected enveloped RNA viruses in lysosomes via disruption of cathepsin L metabolism and function and may play a role in immune control and pathogenesis of these viruses.


Subject(s)
Ebolavirus/physiology , Hemorrhagic Fever, Ebola/immunology , Lassa Fever/immunology , Lassa virus/physiology , Oxidoreductases Acting on Sulfur Group Donors/immunology , Severe Acute Respiratory Syndrome/immunology , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Envelope Proteins/metabolism , Virus Internalization , Cathepsin L/genetics , Cathepsin L/immunology , Cell Line , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/virology , Humans , Lassa Fever/genetics , Lassa Fever/virology , Lassa virus/genetics , Lysosomes/genetics , Lysosomes/immunology , Lysosomes/virology , Oxidoreductases Acting on Sulfur Group Donors/genetics , Severe acute respiratory syndrome-related coronavirus/genetics , Severe Acute Respiratory Syndrome/genetics , Severe Acute Respiratory Syndrome/virology , Viral Envelope Proteins/genetics , Virus Replication
14.
EBioMedicine ; 40: 605-613, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30711514

ABSTRACT

BACKGROUND: Lassa virus (LASV) is the etiologic agent of an acute hemorrhagic fever endemic in West Africa. Natural killer (NK) cells control viral infections in part through the interaction between killer cell immunoglobulin-like receptors (KIRs) and their ligands. LASV infection is associated with defective immune responses, including inhibition of NK cell activity in the presence of MHC-class 1+-infected target cells. METHODS: We compared individual KIR and HLA-class 1 genotypes of 68 healthy volunteers to 51 patients infected with LASV in Sierra Leone, including 37 survivors and 14 fatalities. Next, potential HLA-C1, HLA-C2, and HLA-Bw4 binding epitopes were in silico screened among LASV nucleoprotein (NP) and envelope glycoprotein (GP). Selected 10-mer peptides were then tested in peptide-HLA stabilization, KIR binding and polyfunction assays. FINDINGS: LASV-infected patients were similar to healthy controls, except for the inhibitory KIR2DL2 gene. We found a specific increase in the HLA-C1:KIR2DL2 interaction in fatalities (10/11) as compared to survivors (12/19) and controls (19/29). We also identified that strong of NP and GP viral epitopes was only observed with HLA-C molecules, and associated with strong inhibition of degranulation in the presence of KIR2DL+ NK cells. This inhibitory effect significantly increased in the presence of the vGP420 variant, detected in 28.1% of LASV sequences. INTERPRETATION: Our finding suggests that presentation of specific LASV epitopes by HLA-C alleles to the inhibitory KIR2DL2 receptor on NK cells could potentially prevent the killing of infected cells and provides insights into the mechanisms by which LASV can escape NK-cell-mediated immune pressure.


Subject(s)
Epitopes/immunology , HLA-C Antigens/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lassa Fever/immunology , Lassa Fever/metabolism , Lassa virus/immunology , Receptors, KIR2DL2/metabolism , Antigens, Viral/immunology , Cell Line , Cytotoxicity, Immunologic , Epitope Mapping/methods , Genotype , HLA-C Antigens/genetics , Humans , Immune Tolerance , Immunomodulation , Immunophenotyping , Lassa Fever/genetics , Lassa Fever/virology , Protein Binding , Receptors, KIR2DL2/genetics
15.
Methods Mol Biol ; 1604: 79-88, 2018.
Article in English | MEDLINE | ID: mdl-28986826

ABSTRACT

There are two types of viral diagnostics: (1) those that detect components of the pathogen (like viral RNA or proteins) and (2) those that detect host molecules that rise or fall as a consequence of pathogen infection (like anti-viral antibodies or virus-induced inflammatory cytokines). Quantitative PCR to detect Lassa RNA, and clinical chemistry to detect high liver enzymes (AST/ALT) are commonly used to diagnose Lassa fever. Here, we discuss the various types of diagnostics for Lassa fever and the urgent need for early diagnosis. We also describe a protocol for using the attenuated Lassa vaccine candidate, ML29 , as an antigen for detecting Lassa-specific antibodies. Since antibodies are developed late in the progression of Lassa fever disease, this is not an early diagnostic, but is more useful in surveillance of the population to determine the sero-prevalence of antibodies to Lassa virus (LASV ), and to define treatment options for people in close contact with a Lassa-infected person.


Subject(s)
Lassa Fever/diagnosis , Lassa Fever/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/genetics , Antibody Formation/immunology , Early Diagnosis , Humans , Lassa Fever/genetics , Lassa virus/genetics , Lassa virus/immunology , Lassa virus/pathogenicity , Polymerase Chain Reaction , Viral Vaccines/immunology
16.
J Virol ; 89(15): 7584-92, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972533

ABSTRACT

UNLABELLED: Lassa virus is a notorious human pathogen that infects many thousands of people each year in West Africa, causing severe viral hemorrhagic fevers and significant mortality. The surface glycoprotein of Lassa virus mediates receptor recognition through its GP1 subunit. Here we report the crystal structure of GP1 from Lassa virus, which is the first representative GP1 structure for Old World arenaviruses. We identify a unique triad of histidines that forms a binding site for LAMP1, a known lysosomal protein recently discovered to be a critical receptor for internalized Lassa virus at acidic pH. We demonstrate that mutation of this histidine triad, which is highly conserved among Old World arenaviruses, impairs LAMP1 recognition. Our biochemical and structural data further suggest that GP1 from Lassa virus may undergo irreversible conformational changes that could serve as an immunological decoy mechanism. Together with a variable region that we identify on the surface of GP1, those could be two distinct mechanisms that Lassa virus utilizes to avoid antibody-based immune response. IMPORTANCE: Structural data at atomic resolution for viral proteins is key for understanding their function at the molecular level and can facilitate novel avenues for combating viral infections. Here we used X-ray protein crystallography to decipher the crystal structure of the receptor-binding domain (GP1) from Lassa virus. This is a pathogenic virus that causes significant illness and mortality in West Africa. This structure reveals the overall architecture of GP1 domains from the group of viruses known as the Old World arenaviruses. Using this structural information, we elucidated the mechanisms for pH switch and binding of Lassa virus to LAMP1, a recently identified host receptor that is critical for successful infection. Lastly, our structural analysis suggests two novel immune evasion mechanisms that Lassa virus may utilize to escape antibody-based immune response.


Subject(s)
Lassa Fever/metabolism , Lassa virus/metabolism , Lysosomal Membrane Proteins/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Cell Line , Humans , Lassa Fever/genetics , Lassa Fever/virology , Lassa virus/chemistry , Lassa virus/genetics , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Protein Binding , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Sequence Alignment , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
17.
Genome Biol ; 15(11): 519, 2014.
Article in English | MEDLINE | ID: mdl-25403361

ABSTRACT

We have developed a robust RNA sequencing method for generating complete de novo assemblies with intra-host variant calls of Lassa and Ebola virus genomes in clinical and biological samples. Our method uses targeted RNase H-based digestion to remove contaminating poly(rA) carrier and ribosomal RNA. This depletion step improves both the quality of data and quantity of informative reads in unbiased total RNA sequencing libraries. We have also developed a hybrid-selection protocol to further enrich the viral content of sequencing libraries. These protocols have enabled rapid deep sequencing of both Lassa and Ebola virus and are broadly applicable to other viral genomics studies.


Subject(s)
Ebolavirus/genetics , High-Throughput Nucleotide Sequencing/methods , Lassa virus/genetics , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/virology , Humans , Lassa Fever/genetics , Lassa Fever/virology , RNA, Viral
18.
J Virol ; 87(19): 10908-11, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903830

ABSTRACT

Lassa fever (LF) is a potentially lethal human disease that is caused by the arenavirus Lassa virus (LASV). Annually, around 300,000 infections with up to 10,000 deaths occur in regions of Lassa fever endemicity in West Africa. Here we demonstrate that mice lacking a functional STAT1 pathway are highly susceptible to infection with LASV and develop lethal disease with pathology similar to that reported in humans.


Subject(s)
Lassa Fever/virology , Lassa virus/pathogenicity , STAT1 Transcription Factor/physiology , Africa, Western , Animals , Cells, Cultured , Chlorocebus aethiops , Humans , Kidney/metabolism , Kidney/virology , Lassa Fever/genetics , Lassa Fever/mortality , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/physiology , Survival Rate , Vero Cells
19.
PLoS Negl Trop Dis ; 7(4): e2171, 2013.
Article in English | MEDLINE | ID: mdl-23638192

ABSTRACT

Lassa virus (LASV) is a significant human pathogen that is endemic to several countries in West Africa. Infection with LASV leads to the development of hemorrhagic fever in a significant number of cases, and it is estimated that thousands die each year from the disease. Little is known about the complex immune mechanisms governing the response to LASV or the genetic determinants of susceptibility and resistance to infection. In the study presented here, we have used a whole-genome, microarray-based approach to determine the temporal host response in the peripheral blood mononuclear cells (PBMCs) of non-human primates (NHP) following aerosol exposure to LASV. Sequential sampling over the entire disease course showed that there are strong transcriptional changes of the immune response to LASV exposure, including the early induction of interferon-responsive genes and Toll-like receptor signaling pathways. However, this increase in early innate responses was coupled with a lack of pro-inflammatory cytokine response in LASV exposed NHPs. There was a distinct lack of cytokines such as IL1ß and IL23α, while immunosuppressive cytokines such as IL27 and IL6 were upregulated. Comparison of IRF/STAT1-stimulated gene expression with the viral load in LASV exposed NHPs suggests that mRNA expression significantly precedes viremia, and thus might be used for early diagnostics of the disease. Our results provide a transcriptomic survey of the circulating immune response to hemorrhagic LASV exposure and provide a foundation for biomarker identification to allow clinical diagnosis of LASV infection through analysis of the host response.


Subject(s)
Gene Expression Profiling/methods , Lassa Fever/genetics , Lassa virus/immunology , Leukocytes, Mononuclear/metabolism , Macaca/immunology , Macaca/virology , Animals , Female , Immunity, Innate/immunology , Lassa Fever/virology , Lassa virus/pathogenicity , Male , Toll-Like Receptors/metabolism
20.
J Biol Chem ; 288(23): 16949-16959, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23615902

ABSTRACT

A hallmark of severe Lassa fever is the generalized immune suppression, the mechanism of which is poorly understood. Lassa virus (LASV) nucleoprotein (NP) is the only known 3'-5' exoribonuclease that can suppress type I interferon (IFN) production possibly by degrading immune-stimulatory RNAs. How this unique enzymatic activity of LASV NP recognizes and processes RNA substrates is unknown. We provide an atomic view of a catalytically active exoribonuclease domain of LASV NP (LASV NP-C) in the process of degrading a 5' triphosphate double-stranded (ds) RNA substrate, a typical pathogen-associated molecular pattern molecule, to induce type I IFN production. Additionally, we provide for the first time a high-resolution crystal structure of an active exoribonuclease domain of Tacaribe arenavirus (TCRV) NP. Coupled with the in vitro enzymatic and cell-based interferon suppression assays, these structural analyses strongly support a unified model of an exoribonuclease-dependent IFN suppression mechanism shared by all known arenaviruses. New knowledge learned from these studies should aid the development of therapeutics against pathogenic arenaviruses that can infect hundreds of thousands of individuals and kill thousands annually.


Subject(s)
Arenaviruses, New World , Exoribonucleases , Immune Tolerance , Interferon Type I , Lassa Fever , Lassa virus , Nucleoproteins , RNA, Double-Stranded , RNA, Viral , Viral Proteins , Arenaviruses, New World/enzymology , Arenaviruses, New World/genetics , Arenaviruses, New World/immunology , Cell Line , Crystallography, X-Ray , Exoribonucleases/chemistry , Exoribonucleases/genetics , Exoribonucleases/immunology , Exoribonucleases/metabolism , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Lassa Fever/genetics , Lassa Fever/immunology , Lassa Fever/metabolism , Lassa virus/enzymology , Lassa virus/genetics , Lassa virus/immunology , Nucleoproteins/chemistry , Nucleoproteins/genetics , Nucleoproteins/immunology , Nucleoproteins/metabolism , Protein Structure, Tertiary , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/immunology , RNA, Viral/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL