Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 65
1.
Cell Physiol Biochem ; 55(6): 726-738, 2021 Nov 24.
Article En | MEDLINE | ID: mdl-34816678

Viruses have been widely used to treat cancer for many years and they achieved tremendous success in clinical trials with outstanding results, which has led to the foundation of companies that develop recombinant viruses for a better tumor treatment. Even though there has been a great progress in the field of viral tumor immunotherapy, until now only one virus, the oncolytic virus talimogene laherparepvec (TVEC), a genetically modified herpes simplex virus type 1 (T-VEC), has been approved by the FDA for cancer treatment. Although oncolytic viruses showed progress in certain cancer types and patient populations but they have yet shown limited efficacy when it comes to solid tumors. Only recently it was demonstrated that the immune stimulatory aspect of oncolytic viruses can strongly contribute to their anti-tumoral activity. One specific example in this context are arenaviruses, which have been shown to be non-cytopathic in nature lead to the massive immune activation within the tumor resulting in strong anti-tumoral activity. This strong immune activation might be also linked to their noncytopathic features, as their immune stimulatory potential is not self-limiting as is the case for oncolytic viruses due to their fast eradication by anti-viral immune effects. Because of this strong immune activation, arenaviruses appear superior to oncolytic viruses when it comes to potent and long-lasting anti-tumor effects in a broad variety of tumor types. Currently one of the most promising therapeutics which has turned to be very much beneficial for the treatment of different cancer types is represented by antibodies targeting checkpoint inhibitors such as PD-1/PD-L-1. In this review, we will summarize anti-tumoral effects of arenaviruses, and will discuss their potential to be combined with checkpoint inhibitors for a more efficient tumor treatment, which further emphasizes that arenavirus therapy as a viroimmunotherapy can be an efficient tool for the better clearance of tumors.


Arenavirus/immunology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Biological Products/immunology , Biological Products/therapeutic use , Herpesvirus 1, Human/immunology , Humans , Neoplasms/immunology , Neoplasms/therapy
2.
PLoS Pathog ; 17(3): e1009356, 2021 03.
Article En | MEDLINE | ID: mdl-33647064

Several arenaviruses cause hemorrhagic fevers in humans with high case fatality rates. A vaccine named Candid#1 is available only against Junin virus (JUNV) in Argentina. Specific N-linked glycans on the arenavirus surface glycoprotein (GP) mask important epitopes and help the virus evade antibody responses. However the role of GPC glycans in arenavirus pathogenicity is largely unclear. In a lethal animal model of hemorrhagic fever-causing Machupo virus (MACV) infection, we found that a chimeric MACV with the ectodomain of GPC from Candid#1 vaccine was partially attenuated. Interestingly, mutations resulting in acquisition of N-linked glycans at GPC N83 and N166 frequently occurred in late stages of the infection. These glycosylation sites are conserved in the GPC of wild-type MACV, indicating that this is a phenotypic reversion for the chimeric MACV to gain those glycans crucial for infection in vivo. Further studies indicated that the GPC mutant viruses with additional glycans became more resistant to neutralizing antibodies and more virulent in animals. On the other hand, disruption of these glycosylation sites on wild-type MACV GPC rendered the virus substantially attenuated in vivo and also more susceptible to antibody neutralization, while loss of these glycans did not affect virus growth in cultured cells. We also found that MACV lacking specific GPC glycans elicited higher levels of neutralizing antibodies against wild-type MACV. Our findings revealed the critical role of specific glycans on GPC in arenavirus pathogenicity and have important implications for rational design of vaccines against this group of hemorrhagic fever-causing viruses.


Antibodies, Viral/immunology , Arenavirus/immunology , Hemorrhagic Fever, American/virology , Junin virus/pathogenicity , Animals , Antibodies, Neutralizing/immunology , Arenaviruses, New World/genetics , Arenaviruses, New World/immunology , Arenaviruses, New World/pathogenicity , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/prevention & control , Humans , Junin virus/immunology , Viral Vaccines/immunology
3.
Viruses ; 12(7)2020 07 21.
Article En | MEDLINE | ID: mdl-32708250

Endemic to West Africa and South America, mammalian arenaviruses can cross the species barrier from their natural rodent hosts to humans, resulting in illnesses ranging from mild flu-like syndromes to severe and fatal haemorrhagic zoonoses. The increased frequency of outbreaks and associated high fatality rates of the most prevalent arenavirus, Lassa, in West African countries, highlights the significant risk to public health and to the socio-economic development of affected countries. The devastating impact of these viruses is further exacerbated by the lack of approved vaccines and effective treatments. Differential immune responses to arenavirus infections that can lead to either clearance or rapid, widespread and uncontrolled viral dissemination are modulated by the arenavirus multifunctional proteins, NP and Z. These two proteins control the antiviral response to infection by targeting multiple cellular pathways; and thus, represent attractive targets for antiviral development to counteract infection. The interplay between the host immune responses and viral replication is a key determinant of virus pathogenicity and disease outcome. In this review, we examine the current understanding of host immune defenses against arenavirus infections and summarise the host protein interactions of NP and Z and the mechanisms that govern immune evasion strategies.


Arenaviridae Infections/immunology , Arenavirus/immunology , Nucleocapsid Proteins/immunology , Viral Matrix Proteins/immunology , Animals , Arenaviridae Infections/virology , Host-Pathogen Interactions/immunology , Humans , Immunity , Nucleocapsid Proteins/metabolism , Viral Matrix Proteins/metabolism
4.
J Virol ; 94(9)2020 04 16.
Article En | MEDLINE | ID: mdl-32051278

The arenaviruses Lassa virus (LASV), Junín virus (JUNV), and Machupo virus (MACV) can cause severe and fatal diseases in humans. Although these pathogens are closely related, the host immune responses to these virus infections differ remarkably, with direct implications for viral pathogenesis. LASV infection is immunosuppressive, with a very low-level interferon response. In contrast, JUNV and MACV infections stimulate a robust interferon (IFN) response in a retinoic acid-inducible gene I (RIG-I)-dependent manner and readily activate protein kinase R (PKR), a known host double-stranded RNA (dsRNA) sensor. In response to infection with RNA viruses, host nonself RNA sensors recognize virus-derived dsRNA as danger signals and initiate innate immune responses. Arenavirus nucleoproteins (NPs) contain a highly conserved exoribonuclease (ExoN) motif, through which LASV NP has been shown to degrade virus-derived immunostimulatory dsRNA in biochemical assays. In this study, we for the first time present evidence that LASV restricts dsRNA accumulation during infection. Although JUNV and MACV NPs also have the ExoN motif, dsRNA readily accumulated in infected cells and often colocalized with dsRNA sensors. Moreover, LASV coinfection diminished the accumulation of dsRNA and the IFN response in JUNV-infected cells. The disruption of LASV NP ExoN with a mutation led to dsRNA accumulation and impaired LASV replication in minigenome systems. Importantly, both LASV NP and RNA polymerase L protein were required to diminish the accumulation of dsRNA and the IFN response in JUNV infection. For the first time, we discovered a collaboration between LASV NP ExoN and L protein in limiting dsRNA accumulation. Our new findings provide mechanistic insights into the differential host innate immune responses to highly pathogenic arenavirus infections.IMPORTANCE Arenavirus NPs contain a highly conserved DEDDh ExoN motif, through which LASV NP degrades virus-derived, immunostimulatory dsRNA in biochemical assays to eliminate the danger signal and inhibit the innate immune response. Nevertheless, the function of NP ExoN in arenavirus infection remains to be defined. In this study, we discovered that LASV potently restricts dsRNA accumulation during infection and minigenome replication. In contrast, although the NPs of JUNV and MACV also harbor the ExoN motif, dsRNA readily formed during JUNV and MACV infections, accompanied by IFN and PKR responses. Interestingly, LASV NP alone was not sufficient to limit dsRNA accumulation. Instead, both LASV NP and L protein were required to restrict immunostimulatory dsRNA accumulation. Our findings provide novel and important insights into the mechanism for the distinct innate immune response to these highly pathogenic arenaviruses and open new directions for future studies.


Arenaviruses, New World/immunology , Junin virus/immunology , Lassa virus/immunology , Arenaviridae Infections/virology , Arenavirus/genetics , Arenavirus/immunology , Cell Line , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon Type I/metabolism , Lassa Fever/immunology , Lassa virus/metabolism , Nucleoproteins/metabolism , RNA, Double-Stranded/immunology , Virus Replication , eIF-2 Kinase/metabolism
5.
Nat Commun ; 11(1): 67, 2020 01 03.
Article En | MEDLINE | ID: mdl-31900422

Certain arenaviruses that circulate in rodent populations can cause life-threatening hemorrhagic fevers when they infect humans. Due to their efficient transmission, arenaviruses pose a severe risk for outbreaks and might be exploited as biological weapons. Effective countermeasures against these viruses are highly desired. Ideally, a single remedy would be effective against many or even all the pathogenic viruses in this family. However, despite the fact that all pathogenic arenaviruses from South America utilize transferrin receptor 1 (TfR1) as a cellular receptor, their viral glycoproteins are highly diversified, impeding efforts to isolate cross-neutralizing antibodies. Here we address this problem using a rational design approach to target TfR1-tropic arenaviruses with high potency and breadth. The pan-reactive molecule is highly effective against all arenaviruses that were tested, offering a universal therapeutic approach. Our design scheme avoids the shortcomings of previous immunoadhesins and can be used to combat other zoonotic pathogens.


Arenaviridae Infections/therapy , Arenavirus/immunology , Immunotherapy , Receptors, Transferrin/chemistry , Receptors, Transferrin/immunology , Receptors, Virus/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Arenavirus/chemistry , Arenavirus/genetics , Drug Design , Humans , Receptors, Transferrin/genetics , Receptors, Virus/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology
6.
J Anim Ecol ; 89(2): 506-518, 2020 02.
Article En | MEDLINE | ID: mdl-31545505

A key aim in wildlife disease ecology is to understand how host and parasite characteristics influence parasite transmission and persistence. Variation in host population density can have strong impacts on transmission and outbreaks, and theory predicts particular transmission-density patterns depending on how parasites are transmitted between individuals. Here, we present the results of a study on the dynamics of Morogoro arenavirus in a population of multimammate mice (Mastomys natalensis). This widespread African rodent, which is also the reservoir host of Lassa arenavirus in West Africa, is known for its strong seasonal density fluctuations driven by food availability. We investigated to what degree virus transmission changes with host population density and how the virus might be able to persist during periods of low host density. A seven-year capture-mark-recapture study was conducted in Tanzania where rodents were trapped monthly and screened for the presence of antibodies against Morogoro virus. Observed seasonal seroprevalence patterns were compared with those generated by mathematical transmission models to test different hypotheses regarding the degree of density dependence and the role of chronically infected individuals. We observed that Morogoro virus seroprevalence correlates positively with host density with a lag of 1-4 months. Model results suggest that the observed seasonal seroprevalence dynamics can be best explained by a combination of vertical and horizontal transmission and that a small number of animals need to be infected chronically to ensure viral persistence. Transmission dynamics and viral persistence were best explained by the existence of both acutely and chronically infected individuals and by seasonally changing transmission rates. Due to the presence of chronically infected rodents, rodent control is unlikely to be a feasible approach for eliminating arenaviruses such as Lassa virus from Mastomys populations.


Arenaviridae Infections/epidemiology , Arenavirus/immunology , Rodent Diseases/epidemiology , Animals , Antibodies, Viral , Disease Reservoirs/veterinary , Mice , Population Density , Seroepidemiologic Studies , Tanzania/epidemiology
7.
J Virol ; 94(4)2020 01 31.
Article En | MEDLINE | ID: mdl-31748396

Several clade B New World arenaviruses (NWAs) can cause severe and often fatal hemorrhagic fever, for which preventive and therapeutic measures are severely limited. These NWAs use human transferrin receptor 1 (hTfR1) as a host cell receptor for virus entry. The most prevalent of the pathogenic NWAs is Junín virus (JUNV), the etiological agent of Argentine hemorrhagic fever. Small animal models of JUNV infection are limited because most laboratory rodent species are refractory to disease. Only guinea pigs are known to develop disease following JUNV infection, but the underlying mechanisms are not well characterized. In the present study, we demonstrate marked susceptibility of Hartley guinea pigs to uniformly lethal disease when challenged with as few as 4 PFU of the Romero strain of JUNV. In vitro, we show that infection of primary guinea pig macrophages results in greater JUNV replication compared to infection of hamster or mouse macrophages. We provide evidence that the guinea pig TfR1 (gpTfR1) is the principal receptor for JUNV, while hamster and mouse orthologs fail to support viral entry/infection of pseudotyped murine leukemia viruses expressing pathogenic NWA glycoproteins or JUNV. Together, our results indicate that gpTfR1 serves as the primary receptor for pathogenic NWAs, enhancing viral infection in guinea pigs.IMPORTANCE JUNV is one of five known NWAs that cause viral hemorrhagic fever in humans. Countermeasures against JUNV infection are limited to immunization with the Candid#1 vaccine and immune plasma, which are available only in Argentina. The gold standard small animal model for JUNV infection is the guinea pig. Here, we demonstrate high sensitivity of this species to severe JUNV infection and identify gpTfR1 as the primary receptor. Use of hTfR1 for host cell entry is a feature shared by pathogenic NWAs. Our results show that expression of gpTfR1 or hTfR1 comparably enhances JUNV virus entry/infectivity. Our findings shed light on JUNV infection in guinea pigs as a model for human disease and suggest that similar pathophysiological mechanisms related to iron sequestration during infection and regulation of TfR1 expression may be shared between humans and guinea pigs. A better understanding of the underlying disease process will guide development of new therapeutic interventions.


Junin virus/immunology , Junin virus/pathogenicity , Receptors, Transferrin/metabolism , Animals , Arenavirus/immunology , Arenavirus/pathogenicity , CHO Cells , Chlorocebus aethiops , Cricetulus , Disease Models, Animal , Female , Glycoproteins/metabolism , Guinea Pigs/immunology , Guinea Pigs/metabolism , HEK293 Cells , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/virology , Hemorrhagic Fevers, Viral/immunology , Hemorrhagic Fevers, Viral/virology , Humans , Junin virus/metabolism , Macrophages/virology , Male , Receptors, Transferrin/immunology , Vero Cells , Virus Internalization , Virus Replication
8.
Rev. Soc. Bras. Med. Trop ; 53: e20190132, 2020. tab, graf
Article En | LILACS | ID: biblio-1057283

Abstract INTRODUCTION: In Colombia, there is insufficient epidemiological surveillance of zoonotic hemorrhagic viruses. METHODS: We performed a sero-epidemiological study in indigenous populations of Wayuü, Kankuamos, and Tuchin communities using Maciel hantavirus and Junin arenavirus antigens for IgG detection by ELISA. RESULTS IgG antibodies to hantavirus and arenavirus were found in 5/506 (1%) and 2/506 (0.4%) serum samples, respectively. CONCLUSIONS: Arenavirus and hantavirus circulate in indigenous populations from the Colombian Caribbean region, and the results indicate that the indigenous populations are exposed to these zoonotic agents, with unknown consequences on their health, despite low seroprevalence.


Humans , Male , Female , Adult , Immunoglobulin G/blood , Indians, South American , Orthohantavirus/immunology , Arenavirus/immunology , Arenaviridae Infections/epidemiology , Hantavirus Infections/epidemiology , Antibodies, Viral/blood , Enzyme-Linked Immunosorbent Assay , Seroepidemiologic Studies , Cross-Sectional Studies , Risk Factors , Colombia/epidemiology , Arenaviridae Infections/diagnosis , Hantavirus Infections/diagnosis
9.
Rev Soc Bras Med Trop ; 53: e20190132, 2019.
Article En | MEDLINE | ID: mdl-31859943

INTRODUCTION: In Colombia, there is insufficient epidemiological surveillance of zoonotic hemorrhagic viruses. METHODS: We performed a sero-epidemiological study in indigenous populations of Wayuü, Kankuamos, and Tuchin communities using Maciel hantavirus and Junin arenavirus antigens for IgG detection by ELISA. RESULTS: IgG antibodies to hantavirus and arenavirus were found in 5/506 (1%) and 2/506 (0.4%) serum samples, respectively. CONCLUSIONS: Arenavirus and hantavirus circulate in indigenous populations from the Colombian Caribbean region, and the results indicate that the indigenous populations are exposed to these zoonotic agents, with unknown consequences on their health, despite low seroprevalence.


Antibodies, Viral/blood , Arenaviridae Infections/epidemiology , Arenavirus/immunology , Hantavirus Infections/epidemiology , Immunoglobulin G/blood , Indians, South American , Orthohantavirus/immunology , Adult , Arenaviridae Infections/diagnosis , Colombia/epidemiology , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Female , Hantavirus Infections/diagnosis , Humans , Male , Risk Factors , Seroepidemiologic Studies
10.
Front Immunol ; 10: 372, 2019.
Article En | MEDLINE | ID: mdl-30918506

Hemorrhagic fevers (HF) resulting from pathogenic arenaviral infections have traditionally been neglected as tropical diseases primarily affecting African and South American regions. There are currently no FDA-approved vaccines for arenaviruses, and treatments have been limited to supportive therapy and use of non-specific nucleoside analogs, such as Ribavirin. Outbreaks of arenaviral infections have been limited to certain geographic areas that are endemic but known cases of exportation of arenaviruses from endemic regions and socioeconomic challenges for local control of rodent reservoirs raise serious concerns about the potential for larger outbreaks in the future. This review synthesizes current knowledge about arenaviral evolution, ecology, transmission patterns, life cycle, modulation of host immunity, disease pathogenesis, as well as discusses recent development of preventative and therapeutic pursuits against this group of deadly viral pathogens.


Arenaviridae Infections , Arenavirus/immunology , Disease Outbreaks , Hemorrhagic Fevers, Viral , Immune Tolerance , Ribavirin/therapeutic use , Africa/epidemiology , Arenaviridae Infections/drug therapy , Arenaviridae Infections/epidemiology , Arenaviridae Infections/immunology , Hemorrhagic Fevers, Viral/drug therapy , Hemorrhagic Fevers, Viral/epidemiology , Hemorrhagic Fevers, Viral/immunology , Humans , South America/epidemiology
11.
Sci Rep ; 8(1): 12179, 2018 08 15.
Article En | MEDLINE | ID: mdl-30111770

Immune responses are critical for defense against pathogens. However, prolonged viral infection can result in defective T cell immunity, leading to chronic viral infection. We studied immune activation in response to arenavirus infection during cholestasis using bile duct ligation (BDL). We monitored T cell responses, virus load and liver pathology markers after infection with lymphocytic choriomeningitis virus (LCMV). BDL mice failed to induce protective anti-viral immunity against LCMV and consequently exhibited chronic viral infection. BDL mice exhibited reduced anti-viral T cell immunity as well as reduced type 1 interferon production early after LCMV infection. Consistently, the presence of serum from BDL mice reduced the responsiveness of dendritic cell (DC) and T cell cultures when compared to Sham controls. Following fractionation and mass spectrometry analyses of sera, we identified several serum factors to be upregulated following BDL including bilirubin, bile acids, 78 kDa Glucose regulated protein (GRP78) and liver enzymes. Bilirubin and GRP78 were capable of inhibiting DC and T cell activation. In this work, we demonstrate that liver damage mediated by cholestasis results in defective immune induction following arenavirus infection.


Arenaviridae Infections/immunology , Cholestasis/immunology , Liver Diseases/immunology , Liver/immunology , Animals , Arenaviridae Infections/pathology , Arenavirus/immunology , Bile Ducts/immunology , Bile Ducts/pathology , CD4-Positive T-Lymphocytes/immunology , Cholestasis/pathology , Dendritic Cells/immunology , Endoplasmic Reticulum Chaperone BiP , Interferon Type I/immunology , Liver/pathology , Liver Diseases/pathology , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Signal Transduction
12.
Parasit Vectors ; 11(1): 416, 2018 Jul 13.
Article En | MEDLINE | ID: mdl-30005641

BACKGROUND: Lassa fever, killing thousands of people annually, is the most reported viral zoonotic disease in Nigeria. Recently, different rodent species carrying diverse lineages of the Lassa virus (LASV) in addition to a novel Mobala-like genetic sequence were detected within the country. Here, screening 906 small mammal specimens from 11 localities for IgG antibodies and incorporating previous PCR detection data involving the same populations, we further describe arenavirus prevalence across Nigeria in relation to host species and geographical location. METHODS: Small mammals were trapped during the period 2011-2015 according to geographical location (endemic and non-endemic zones for Lassa fever), season (rainy and dry seasons between 2011 and 2012 for certain localities) and habitat (indoors, peridomestic settings and sylvatic vegetation). Identification of animal specimens from genera such as Mastomys and Mus (Nannomys) was assisted by DNA sequencing. Small mammals were tested for LASV IgG antibody using an indirect immunofluorescence assay (IFA). RESULTS: Small mammals were infected in both the endemic and non-endemic zones for Lassa fever, with a wider range of species IgG-positive (n = 8) than those which had been previously detected to be PCR-positive (n = 3). IgG-positive species, according to number of infected individuals, were Mastomys natalensis (n = 40), Mastomys erythroleucus (n = 15), Praomys daltoni (n = 6), Mus baoulei (n = 5), Rattus rattus (n = 2), Crocidura spp. (n = 2), Mus minutoides (n = 1) and Praomys misonnei (n = 1). Multimammate mice (Mastomys natalensis and M. erythroleucus) were the most ubiquitously infected, with animals testing positive by either PCR or IgG in 7 out of the 11 localities sampled. IgG prevalence in M. natalensis ranged from 1% in Abagboro, 17-36 % in Eguare Egoro, Ekpoma and Ngel Nyaki, up to 52 % in Mayo Ranewo. Prevalence according to locality, season and age was not, however, statistically significant for M. natalensis in Eguare Egoro and Ekpoma, localities that were sampled longitudinally. CONCLUSIONS: Overall, our study demonstrates that arenavirus occurrence is probably more widely distributed geographically and in extent of host taxa than is currently realized. This expanded scope should be taken into consideration in Lassa fever control efforts. Further sampling should also be carried out to isolate and characterize potential arenaviruses present in small mammal populations we found to be seropositive.


Antibodies, Viral/blood , Arenaviridae Infections/blood , Arenaviridae Infections/veterinary , Arenavirus/physiology , Rodent Diseases/blood , Rodent Diseases/epidemiology , Animals , Arenaviridae Infections/epidemiology , Arenaviridae Infections/virology , Arenavirus/immunology , Disease Reservoirs/virology , Eulipotyphla/virology , Geography , Lassa virus/immunology , Lassa virus/physiology , Mice , Nigeria/epidemiology , Prevalence , RNA, Viral/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Rodent Diseases/virology , Rodentia/virology , Seroepidemiologic Studies
13.
Parasit Vectors ; 11(1): 90, 2018 02 08.
Article En | MEDLINE | ID: mdl-29422075

BACKGROUND: Parasite evolution is hypothesized to select for levels of parasite virulence that maximise transmission success. When host population densities fluctuate, low levels of virulence with limited impact on the host are expected, as this should increase the likelihood of surviving periods of low host density. We examined the effects of Morogoro arenavirus on the survival and recapture probability of multimammate mice (Mastomys natalensis) using a seven-year capture-mark-recapture time series. Mastomys natalensis is the natural host of Morogoro virus and is known for its strong seasonal density fluctuations. RESULTS: Antibody presence was negatively correlated with survival probability (effect size: 5-8% per month depending on season) but positively with recapture probability (effect size: 8%). CONCLUSIONS: The small negative correlation between host survival probability and antibody presence suggests that either the virus has a negative effect on host condition, or that hosts with lower survival probability are more likely to obtain Morogoro virus infection, for example due to particular behavioural or immunological traits. The latter hypothesis is supported by the positive correlation between antibody status and recapture probability which suggests that risky behaviour might increase the probability of becoming infected.


Arenaviridae Infections/veterinary , Arenavirus/isolation & purification , Murinae , Rodent Diseases/mortality , Animals , Antibodies, Viral/blood , Arenaviridae Infections/mortality , Arenavirus/immunology , Behavior, Animal , Rodent Diseases/virology , Survival Analysis
14.
J Gen Virol ; 99(2): 187-193, 2018 02.
Article En | MEDLINE | ID: mdl-29393022

We report the development of recombinant New World (Junín; JUNV) and Old World (lymphocytic choriomeningitis virus; LCMV) mammarenaviruses that encode an HA-tagged matrix protein (Z). These viruses permit the robust affinity purification of Z from infected cells or virions, as well as the detection of Z by immunofluorescent microscopy. Importantly, the HA-tagged viruses grow with wild-type kinetics in a multi-cycle growth assay. Using these viruses, we report a novel description of JUNV Z localization in infected cells, as well as the first description of colocalization between LCMV Z and the GTPase Rab5c. This latter result, when combined with our previous findings that LCMV genome and glycoprotein also colocalize with Rab5c, suggest that LCMV may target Rab5c-positive membranes for preassembly of virus particles prior to budding. The recombinant viruses reported here will provide the field with new tools to better study Z protein functionality and identify key Z protein interactions with host machinery.


Arenavirus/physiology , Carrier Proteins/metabolism , Epitopes/immunology , GTP Phosphohydrolases/metabolism , Host-Pathogen Interactions , Lymphocytic choriomeningitis virus/physiology , A549 Cells , Arenavirus/immunology , Carrier Proteins/genetics , Endosomes/metabolism , Endosomes/virology , GTP Phosphohydrolases/genetics , Genes, Reporter , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/virology , Intracellular Signaling Peptides and Proteins , Lymphocytic choriomeningitis virus/immunology , Microscopy, Fluorescence , Virus Assembly
15.
Ecohealth ; 14(3): 463-473, 2017 09.
Article En | MEDLINE | ID: mdl-28616660

Infectious diseases of wildlife are typically studied using data on antibody and pathogen levels. In order to interpret these data, it is necessary to know the course of antibodies and pathogen levels after infection. Such data are typically collected using experimental infection studies in which host individuals are inoculated in the laboratory and sampled over an extended period, but because laboratory conditions are controlled and much less variable than natural conditions, the immune response and pathogen dynamics may differ. Here, we compared Morogoro arenavirus infection patterns between naturally and experimentally infected multimammate mice (Mastomys natalensis). Longitudinal samples were collected during three months of bi-weekly trapping in Morogoro, Tanzania, and antibody titer and viral RNA presence were determined. The time of infection was estimated from these data using a recently developed Bayesian approach, which allowed us to assess whether the natural temporal patterns match the previously observed patterns in the laboratory. A good match was found for 52% of naturally infected individuals, while most of the mismatches can be explained by the presence of chronically infected individuals (35%), maternal antibodies (10%), and an antibody detection limit (25%). These results suggest that while laboratory data are useful for interpreting field samples, there can still be differences due to conditions that were not tested in the laboratory.


Animals, Wild/virology , Arenaviridae Infections/immunology , Arenaviridae Infections/pathology , Arenavirus/pathogenicity , Disease Reservoirs/virology , Mice/virology , Rodent Diseases/virology , Animals , Animals, Wild/immunology , Arenavirus/immunology , Rodent Diseases/immunology , Rodent Diseases/pathology , Tanzania
16.
Virology ; 501: 35-46, 2017 01 15.
Article En | MEDLINE | ID: mdl-27855284

Several arenaviruses, chiefly Lassa (LASV) in West Africa, cause hemorrhagic fever (HF) disease in humans and pose important public health problems in their endemic regions. To date, there are no FDA-approved arenavirus vaccines and current anti-arenaviral therapy is limited to the use of ribavirin that has very limited efficacy. In this work we document that a recombinant prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) with a codon deoptimized (CD) surface glycoprotein (GP), rLCMV/CD, exhibited wild type (WT)-like growth properties in cultured cells despite barely detectable GP expression levels in rLCMV/CD-infected cells. Importantly, rLCMV/CD was highly attenuated in vivo but able to induce complete protection against a subsequent lethal challenge with rLCMV/WT. Our findings support the feasibility of implementing an arenavirus GP CD-based approach for the development of safe and effective live-attenuated vaccines (LAVs) to combat diseases caused by human pathogenic arenaviruses.


Arenaviridae Infections/virology , Arenavirus/genetics , Codon/genetics , Glycoproteins/genetics , Vaccines, Attenuated/genetics , Viral Proteins/genetics , Viral Vaccines/genetics , Amino Acid Sequence , Animals , Arenaviridae Infections/immunology , Arenavirus/immunology , Codon/immunology , Glycoproteins/administration & dosage , Glycoproteins/immunology , Humans , Mice , Molecular Sequence Data , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Proteins/administration & dosage , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
17.
J Virol ; 90(22): 10259-10270, 2016 Nov 15.
Article En | MEDLINE | ID: mdl-27605671

The family Arenaviridae includes several important human pathogens that can cause severe hemorrhagic fever and greatly threaten public health. As a major component of the innate immune system, the RLR/MAVS signaling pathway is involved in recognizing viral components and initiating antiviral activity. It has been reported that arenavirus infection can suppress the innate immune response, and NP and Z proteins of pathogenic arenaviruses can disrupt RLR/MAVS signaling, thus inhibiting production of type I interferon (IFN-I). However, recent studies have shown elevated IFN-I levels in certain arenavirus-infected cells. The mechanism by which arenavirus infection induces IFN-I responses remains unclear. In this study, we determined that the L polymerase (Lp) of Mopeia virus (MOPV), an Old World (OW) arenavirus, can activate the RLR/MAVS pathway and thus induce the production of IFN-I. This activation is associated with the RNA-dependent RNA polymerase activity of Lp. This study provides a foundation for further studies of interactions between arenaviruses and the innate immune system and for the elucidation of arenavirus pathogenesis. IMPORTANCE: Distinct innate immune responses are observed when hosts are infected with different arenaviruses. It has been widely accepted that NP and certain Z proteins of arenaviruses inhibit the RLR/MAVS signaling pathway. The viral components responsible for the activation of the RLR/MAVS signaling pathway remain to be determined. In the current study, we demonstrate for the first time that the Lp of MOPV, an OW arenavirus, can activate the RLR/MAVS signaling pathway and thus induce the production of IFN-I. Based on our results, we proposed that dynamic interactions exist among Lp-produced RNA, NP, and the RLR/MAVS signaling pathway, and the outcome of these interactions may determine the final IFN-I response pattern: elevated or reduced. Our study provides a possible explanation for how IFN-I can become activated during arenavirus infection and may help us gain insights into the interactions that form between different arenavirus components and the innate immune system.


Adaptor Proteins, Signal Transducing/metabolism , Arenaviridae Infections/metabolism , Arenaviruses, Old World/metabolism , Signal Transduction/physiology , Viral Proteins/metabolism , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Arenavirus/immunology , Arenavirus/metabolism , Arenaviruses, Old World/immunology , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , DNA-Directed RNA Polymerases/metabolism , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Immunity, Innate/immunology , Interferon Type I/metabolism , Vero Cells
19.
Nat Commun ; 7: 11544, 2016 05 10.
Article En | MEDLINE | ID: mdl-27161536

Lassa fever is a severe multisystem disease that often has haemorrhagic manifestations. The epitopes of the Lassa virus (LASV) surface glycoproteins recognized by naturally infected human hosts have not been identified or characterized. Here we have cloned 113 human monoclonal antibodies (mAbs) specific for LASV glycoproteins from memory B cells of Lassa fever survivors from West Africa. One-half bind the GP2 fusion subunit, one-fourth recognize the GP1 receptor-binding subunit and the remaining fourth are specific for the assembled glycoprotein complex, requiring both GP1 and GP2 subunits for recognition. Notably, of the 16 mAbs that neutralize LASV, 13 require the assembled glycoprotein complex for binding, while the remaining 3 require GP1 only. Compared with non-neutralizing mAbs, neutralizing mAbs have higher binding affinities and greater divergence from germline progenitors. Some mAbs potently neutralize all four LASV lineages. These insights from LASV human mAb characterization will guide strategies for immunotherapeutic development and vaccine design.


Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Lassa virus/immunology , Antibody Specificity , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , Arenavirus/immunology , Cross Reactions , Epitope Mapping , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Humans , Lassa Fever/immunology , Lassa Fever/prevention & control , Lassa virus/genetics , Models, Molecular , Mutagenesis, Site-Directed , Sequence Deletion , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
20.
Methods Mol Biol ; 1403: 313-51, 2016.
Article En | MEDLINE | ID: mdl-27076139

Several arenavirus cause hemorrhagic fever disease in humans and pose a significant public health problem in their endemic regions. To date, no licensed vaccines are available to combat human arenavirus infections, and anti-arenaviral drug therapy is limited to an off-label use of ribavirin that is only partially effective. The development of arenavirus reverse genetics approaches provides investigators with a novel and powerful approach for the investigation of the arenavirus molecular and cell biology. The use of cell-based minigenome systems has allowed examining the cis- and trans-acting factors involved in arenavirus replication and transcription and the identification of novel anti-arenaviral drug targets without requiring the use of live forms of arenaviruses. Likewise, it is now feasible to rescue infectious arenaviruses entirely from cloned cDNAs containing predetermined mutations in their genomes to investigate virus-host interactions and mechanisms of pathogenesis, as well as to facilitate screens to identify anti-arenaviral drugs and development of novel live-attenuated arenavirus vaccines. Recently, reverse genetics have also allowed the generation of tri-segmented arenaviruses expressing foreign genes, facilitating virus detection and opening the possibility of implementing live-attenuated arenavirus-based vaccine vector approaches. Likewise, the development of single-cycle infectious, reporter-expressing, arenaviruses has provided a new experimental method to study some aspects of the biology of highly pathogenic arenaviruses without the requirement of high-security biocontainment required to study HF-causing arenaviruses. In this chapter we summarize the current knowledge on arenavirus reverse genetics and the implementation of plasmid-based reverse genetics techniques for the development of arenavirus vaccines and vaccine vectors.


Arenaviridae Infections/prevention & control , Arenavirus/immunology , Reverse Genetics/methods , Viral Vaccines/genetics , Viral Vaccines/immunology , Animals , Arenaviridae Infections/immunology , Humans
...