Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Viruses ; 13(6)2021 05 25.
Article in English | MEDLINE | ID: mdl-34070281

ABSTRACT

Arenaviruses cause chronic and asymptomatic infections in their natural host, rodents, and several arenaviruses cause severe hemorrhagic fever that has a high mortality in infected humans, seriously threatening public health. There are currently no FDA-licensed drugs available against arenaviruses; therefore, it is important to develop novel antiviral strategies to combat them, which would be facilitated by a detailed understanding of the interactions between the viruses and their hosts. To this end, we performed a transcriptomic analysis on cells infected with arenavirus lymphocytic choriomeningitis virus (LCMV), a neglected human pathogen with clinical significance, and found that the signal transducer and activator of transcription 3 (STAT3) signaling pathway was activated. A further investigation indicated that STAT3 could be activated by the RNA-dependent RNA polymerase L protein (Lp) of LCMV. Our functional analysis found that STAT3 cannot affect LCMV multiplication in A549 cells. We also found that STAT3 was activated by the Lp of Mopeia virus and Junin virus, suggesting that this activation may be conserved across certain arenaviruses. Our study explored the interactions between arenaviruses and STAT3, which may help us to better understand the molecular and cell biology of arenaviruses.


Subject(s)
Arenavirus/enzymology , Arenavirus/metabolism , Host-Pathogen Interactions , RNA-Dependent RNA Polymerase/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , A549 Cells , Arenavirus/genetics , Arenavirus/pathogenicity , Cell Line , HEK293 Cells , HeLa Cells , Humans , RNA-Dependent RNA Polymerase/metabolism , Signal Transduction/physiology , Virus Replication
2.
Viruses ; 13(4)2021 04 18.
Article in English | MEDLINE | ID: mdl-33919632

ABSTRACT

Rodent-borne arenaviruses have been traditionally predominantly associated with certain muroid species from Mastomys/Praomys genera (African arenaviruses) or with species that belong to murid subfamily Cricetidae (New World arenaviruses) [...].


Subject(s)
Arenaviridae Infections/veterinary , Arenavirus/genetics , Arenavirus/pathogenicity , Amino Acid Sequence , Animals , Arenaviridae Infections/transmission , Arenavirus/classification , Fishes/virology , Humans , Rodentia/virology , Snakes/virology
3.
Internet resource in Portuguese | LIS -Health Information Locator, LIS-bvsms | ID: lis-46946

ABSTRACT

Portal do Ministério da Saúde- Publicado: Segunda, 20 de Janeiro de 2020, 21h28 Apresenta um vídeo com esclarecimentos sobre o Arenavírus, informa sobre a situação atual do virús em São Paulo e disponibiliza o Boletim epidemiológico


Subject(s)
Arenavirus/pathogenicity , Hemorrhagic Fever, American/virology , Fatal Outcome
4.
J Virol ; 94(4)2020 01 31.
Article in English | MEDLINE | ID: mdl-31748396

ABSTRACT

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.


Subject(s)
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
5.
Viruses ; 11(3)2019 03 23.
Article in English | MEDLINE | ID: mdl-30909570

ABSTRACT

Lassa virus (LASV) and Mopeia virus (MOPV) are two closely related Old-World mammarenaviruses. LASV causes severe hemorrhagic fever with high mortality in humans, whereas no case of MOPV infection has been reported. Comparing MOPV and LASV is a powerful strategy to unravel pathogenic mechanisms that occur during the course of pathogenic arenavirus infection. We used a yeast two-hybrid approach to identify cell partners of MOPV and LASV Z matrix protein in which two autophagy adaptors were identified, NDP52 and TAX1BP1. Autophagy has emerged as an important cellular defense mechanism against viral infections but its role during arenavirus infection has not been shown. Here, we demonstrate that autophagy is transiently induced by MOPV, but not LASV, in infected cells two days after infection. Impairment of the early steps of autophagy significantly decreased the production of MOPV and LASV infectious particles, whereas a blockade of the degradative steps impaired only MOPV infectious particle production. Our study provides insights into the role played by autophagy during MOPV and LASV infection and suggests that this process could partially explain their different pathogenicity.


Subject(s)
Arenavirus/physiology , Autophagy , Lassa virus/physiology , Animals , Arenavirus/pathogenicity , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lassa virus/pathogenicity , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Two-Hybrid System Techniques , Vero Cells
6.
J Virol ; 92(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29669840

ABSTRACT

RIG-I is a major cytoplasmic sensor of viral pathogen-associated molecular pattern (PAMP) RNA and induces type I interferon (IFN) production upon viral infection. A double-stranded RNA (dsRNA)-binding protein, PACT, plays an important role in potentiating RIG-I function. We have shown previously that arenaviral nucleoproteins (NPs) suppress type I IFN production via their RNase activity to degrade PAMP RNA. We report here that NPs of arenaviruses block the PACT-induced enhancement of RIG-I function to mediate type I IFN production and that this inhibition is dependent on the RNase function of NPs, which is different from that of a known mechanism of other viral proteins to abolish the interaction between PACT and RIG-I. To understand the biological roles of PACT and RIG-I in authentic arenavirus infection, we analyze growth kinetics of recombinant Pichinde virus (PICV), a prototypical arenavirus, in RIG-I knockout (KO) and PACT KO mouse embryonic fibroblast (MEF) cells. Wild-type (WT) PICV grew at higher titers in both KO MEF lines than in normal MEFs, suggesting the important roles of these cellular proteins in restricting virus replication. PICV carrying the NP RNase catalytically inactive mutation could not grow in normal MEFs but could replicate to some extent in both KO MEF lines. The level of virus growth was inversely correlated with the amount of type I IFNs produced. These results suggest that PACT plays an important role in potentiating RIG-I function to produce type I IFNs in order to restrict arenavirus replication and that viral NP RNase activity is essential for optimal viral replication by suppressing PACT-induced RIG-I activation.IMPORTANCE We report here a new role of the nucleoproteins of arenaviruses that can block type I IFN production via their specific inhibition of the cellular protein sensors of virus infection (RIG-I and PACT). Our results suggest that PACT plays an important role in potentiating RIG-I function to produce type I IFNs in order to restrict arenavirus replication. This new knowledge can be exploited for the development of novel antiviral treatments and/or vaccines against some arenaviruses that can cause severe and lethal hemorrhagic fever diseases in humans.


Subject(s)
Arenavirus/pathogenicity , Host-Pathogen Interactions , Interferon Type I/metabolism , Nucleoproteins/metabolism , RNA-Binding Proteins/metabolism , Receptors, Retinoic Acid/metabolism , Viral Proteins/metabolism , Arenaviridae Infections/genetics , Arenaviridae Infections/metabolism , Arenaviridae Infections/virology , HEK293 Cells , Humans , Nucleoproteins/genetics , Pichinde virus/physiology , RNA-Binding Proteins/genetics , Receptors, Retinoic Acid/genetics , Viral Proteins/genetics , Virus Replication
7.
Methods Mol Biol ; 1604: 55-78, 2018.
Article in English | MEDLINE | ID: mdl-28986825

ABSTRACT

This article will outline surveillance approaches for viral hemorrhagic fevers. Specific methods for surveillance of clinical samples will be emphasized. Separate articles will describe methods for surveillance of rodent-borne viruses (roboviruses) and arthropod-borne viruses (arboviruses). Since the appearance of hantaviruses and arenaviruses in the Americas, more than 30 different species in each group have been established, and therefore they have become the most frequently emerging viruses. Flaviviruses such as yellow fever and dengue viruses, although easier to recognize, are also more widely spread and therefore considered a very important public health issue, particularly for under-developed countries. On the other hand, marburgviruses and ebolaviruses, previously thought to be restricted to the African continent, have recently been shown to be more global. For many of these agents virus isolation has been a challenging task: trapping the specific vectors (mosquitoes and ticks), and reservoirs (rodents and bats), or obtaining the samples from suspected clinical human cases demands special protective gear, uncommon devices (respirators), special facilities (BSL-3 and 4), and particular skills to recognize the slow and inapparent cytopathic effects in cell culture. Alternatively, serological and molecular approaches have been very helpful in discovering and describing newly emerging viruses in many areas where the previous resources are unavailable. Unfortunately, in many cases, detailed studies have been performed only after outbreaks occur, and then active surveillance is needed to prevent viral dissemination in human populations.


Subject(s)
Hemorrhagic Fever with Renal Syndrome/epidemiology , Hemorrhagic Fever with Renal Syndrome/genetics , Hemorrhagic Fevers, Viral/epidemiology , Hemorrhagic Fevers, Viral/genetics , Animals , Arboviruses/genetics , Arboviruses/pathogenicity , Arenavirus/genetics , Arenavirus/pathogenicity , Orthohantavirus/genetics , Orthohantavirus/pathogenicity , Hemorrhagic Fever with Renal Syndrome/virology , Hemorrhagic Fevers, Viral/virology , Humans , Public Health
8.
Methods Mol Biol ; 1604: 257-267, 2018.
Article in English | MEDLINE | ID: mdl-28986841

ABSTRACT

Hemorrhagic fever (HF) viruses, such as Lassa, Ebola, and dengue viruses, represent major human health risks due to their highly contagious nature, the severity of the clinical manifestations induced, the lack of vaccines, and the very limited therapeutic options currently available. Appropriate animal models are obviously critical to study disease pathogenesis and develop efficient therapies. We recently reported that the clone 13 (Cl13) variant of the lymphocytic choriomeningitis virus (LCMV-Cl13), a prototype arenavirus closely related to Lassa virus, causes in some mouse strains endothelial damage, vascular leakage, platelet loss, and death, mimicking pathological aspects typically observed in Lassa and other HF syndromes. This model has the advantage that the mice used are fully immunocompetent, allowing studies on the contribution of the immune response to disease progression. Moreover, LCMV is very well characterized and exhibits limited pathogenicity in humans, allowing handling in convenient BSL-2 facilities. In this chapter we outline protocols for the induction and analysis of arenavirus-mediated pathogenesis in the NZB/LCMV model, including mouse infection, virus titer determination, platelet counting, phenotypic analysis of virus-specific T cells, and assessment of vascular permeability.


Subject(s)
Hemorrhagic Fevers, Viral/virology , Lymphocytic choriomeningitis virus/pathogenicity , Animals , Arenavirus/genetics , Arenavirus/pathogenicity , Bronchoalveolar Lavage , Disease Models, Animal , Lassa virus/genetics , Lassa virus/pathogenicity , Lymphocytic choriomeningitis virus/genetics , Mice , Platelet Count , Viral Vaccines
9.
Methods Mol Biol ; 1604: 291-302, 2018.
Article in English | MEDLINE | ID: mdl-28986844

ABSTRACT

Viral hemorrhagic fevers affect liver functions such as important metabolic processes and the replacement of new blood cells, coagulation factors, and growth factors. Typically, multi-organ diseases such as viral hemorrhagic fevers are studied in an organism, but it is also possible to derive information about the molecular events involved in disease processes by focusing on liver cell culture. Here we describe a multi-cell culture system that is capable of replicating the arenavirus LCMV-WE, a virus that can cause hemorrhagic fever in primates, as a model for liver infection by a hemorrhagic fever virus.


Subject(s)
Hemorrhagic Fevers, Viral/virology , Liver Diseases/virology , Liver/virology , Arenavirus/genetics , Arenavirus/pathogenicity , Hemorrhagic Fevers, Viral/genetics , Humans , Primary Cell Culture
10.
Uirusu ; 68(1): 51-62, 2018.
Article in Japanese | MEDLINE | ID: mdl-31105135

ABSTRACT

Arenavirus is a genetic term for viruses belonging to the family Arenaviridae and is presented from lymphocytic choriomeningitis virus (LCMV), which shows almost no pathogenicity to humans, to Lassa virus, Junin virus, Machupo virus, Chapare virus, Lujo virus, Sabia virus, and Guanarito virus, which shows high pathogenicity to humans. These viruses except for LCMV are risk group 4 pathogens specified by World Health Organization. Based on this designation, it is designated as Class I pathogens in Japan. Although there have been no reports excluding one imported case of the Lassa fever patient, it is not surprising whenever imported cases occur in our country. Considering the disease severity and mortality rate, it is an urgent matter to develop vaccines and therapeutic drugs in endemic areas, and maintenances of these are also important in countries other than endemic areas. However, basic research on highly pathogenic arenavirus infections and development of therapeutic drugs are not easily progressed, because handling in highly safe research facilities is indispensable. In this article, we will outline the current knowledge from the recent basic research on arenavirus to the development situation of antivirals against arenaviruses.


Subject(s)
Antiviral Agents , Arenaviridae Infections/drug therapy , Arenaviridae Infections/virology , Arenavirus/classification , Arenavirus/pathogenicity , Drug Discovery , Africa, Western/epidemiology , Arenaviridae Infections/epidemiology , Arenaviridae Infections/prevention & control , Arenavirus/genetics , Arenavirus/physiology , Disease Outbreaks , Drug Discovery/trends , Genome, Viral/genetics , Humans , Research/trends , Transcription, Genetic , Viral Vaccines , Virion
11.
Ecohealth ; 14(3): 463-473, 2017 09.
Article in English | MEDLINE | ID: mdl-28616660

ABSTRACT

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.


Subject(s)
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
12.
J Virol ; 90(8): 3810-3818, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26865707

ABSTRACT

Mammalian arenaviruses are zoonotic viruses that cause asymptomatic, persistent infections in their rodent hosts but can lead to severe and lethal hemorrhagic fever with bleeding and multiorgan failure in human patients. Lassa virus (LASV), for example, is endemic in several West African countries, where it is responsible for an estimated 500,000 infections and 5,000 deaths annually. There are currently no FDA-licensed therapeutics or vaccines available to combat arenavirus infection. A hallmark of arenavirus infection (e.g., LASV) is general immunosuppression that contributes to high viremia. Here, we discuss the early host immune responses to arenavirus infection and the recently discovered molecular mechanisms that enable pathogenic viruses to suppress host immune recognition and to contribute to the high degree of virulence. We also directly compare the innate immune evasion mechanisms between arenaviruses and other hemorrhagic fever-causing viruses, such as Ebola, Marburg, Dengue, and hantaviruses. A better understanding of the immunosuppression and immune evasion strategies of these deadly viruses may guide the development of novel preventative and therapeutic options.


Subject(s)
Arenaviridae Infections/immunology , Arenavirus/immunology , Arenavirus/pathogenicity , Immunity, Innate , Animals , Hemorrhagic Fevers, Viral/immunology , Hemorrhagic Fevers, Viral/virology , Humans , Immune Evasion , Interferons/antagonists & inhibitors , Zinc Fingers
13.
Biomed Res Int ; 2015: 793257, 2015.
Article in English | MEDLINE | ID: mdl-26266264

ABSTRACT

Human pathogenic hantaviruses and arenaviruses are maintained in nature by persistent infection of rodent carrier populations. Several members of these virus groups can cause significant disease in humans that is generically termed viral hemorrhagic fever (HF) and is characterized as a febrile illness with an increased propensity to cause acute inflammation. Human interaction with rodent carrier populations leads to infection. Arenaviruses are also viewed as potential biological weapons threat agents. There is an increased interest in studying these viruses in animal models to gain a deeper understating not only of viral pathogenesis, but also for the evaluation of medical countermeasures (MCM) to mitigate disease threats. In this review, we examine current knowledge regarding animal models employed in the study of these viruses. We include analysis of infection models in natural reservoirs and also discuss the impact of strain heterogeneity on the susceptibility of animals to infection. This information should provide a comprehensive reference for those interested in the study of arenaviruses and hantaviruses not only for MCM development but also in the study of viral pathogenesis and the biology of these viruses in their natural reservoirs.


Subject(s)
Arenavirus/pathogenicity , Hantavirus Infections/virology , Hemorrhagic Fevers, Viral/virology , Orthohantavirus/pathogenicity , Animals , Disease Reservoirs , Hantavirus Infections/epidemiology , Hantavirus Infections/pathology , Hemorrhagic Fevers, Viral/epidemiology , Hemorrhagic Fevers, Viral/pathology , Humans , Models, Animal , Rodentia/virology
14.
Sci Rep ; 5: 10445, 2015 May 29.
Article in English | MEDLINE | ID: mdl-26022445

ABSTRACT

Arenaviruses can cause mild to severe hemorrhagic fevers. Humans mainly get infected through contact with infected rodents or their excretions, yet little is known about transmission dynamics within rodent populations. Morogoro virus (MORV) is an Old World arenavirus closely related to Lassa virus with which it shares the same host species Mastomys natalensis. We injected MORV in its host, and sampled blood and excretions at frequent intervals. Infection in adults was acute; viral RNA disappeared from blood after 18 days post infection (dpi) and from excretions after 39 dpi. Antibodies were present from 7 dpi and never disappeared. Neonatally infected animals acquired a chronic infection with RNA and antibodies in blood for at least 3 months. The quantified excretion and antibody patterns can be used to inform mathematical transmission models, and are essential for understanding and controlling transmission in the natural rodent host populations.


Subject(s)
Arenaviridae Infections/transmission , Arenavirus/pathogenicity , Lassa virus/pathogenicity , Animals , Antibodies, Viral/blood , Arenaviridae Infections/pathology , Arenaviridae Infections/virology , Arenavirus/genetics , Disease Reservoirs/virology , Humans , Lassa virus/genetics , Murinae/virology , RNA, Viral/blood , Tanzania
15.
J Clin Virol ; 64: 120-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25549822

ABSTRACT

Significant progress has been made in the past 10 years in unraveling the molecular biology of highly pathogenic arenaviruses that are endemic in several West African countries (Lassa fever virus) and in some regions of South America (Argentine and Bolivian hemorrhagic fever viruses). While this has resulted in proof-of-concept studies of novel vaccine candidates in non-human primates and in the discovery of several novel antiviral small molecule drug candidates, none of them has been tested in the clinic to date. The recent Ebola outbreak in West Africa has demonstrated very clearly that there is an urgent need to develop the prophylactic and therapeutic armamentarium against viral hemorrhagic fever viruses as part of a global preparedness for future epidemics. As it pertains to this goal, the present article summarizes the current knowledge of highly pathogenic arenaviruses and identifies opportunities for translational research.


Subject(s)
Antiviral Agents/therapeutic use , Arenaviridae Infections/epidemiology , Arenaviridae Infections/therapy , Biomedical Research , Lassa Fever/epidemiology , Lassa Fever/therapy , Viral Vaccines , Africa, Western/epidemiology , Animals , Arenavirus/pathogenicity , Argentina/epidemiology , Bolivia/epidemiology , Epidemics/prevention & control , Hemorrhagic Fevers, Viral/diagnosis , Hemorrhagic Fevers, Viral/epidemiology , Hemorrhagic Fevers, Viral/therapy , Humans , Lassa Fever/diagnosis , Lassa virus/pathogenicity
16.
PLoS One ; 8(8): e72290, 2013.
Article in English | MEDLINE | ID: mdl-23991083

ABSTRACT

Lassa virus causes hemorrhagic Lassa fever in humans, while the related Old World arenaviruses Mopeia, Morogoro, and Mobala are supposedly apathogenic to humans and cause only inapparent infection in non-human primates. Here, we studied whether the virulence of Old World arenaviruses in humans and non-human primates is reflected in type I interferon receptor deficient (IFNAR(-/-)) mice by testing several strains of Lassa virus vs. the apathogenic viruses Mopeia, Morogoro, and Mobala. All Lassa virus strains tested-Josiah, AV, BA366, and Nig04-10-replicated to high titers in blood, lung, kidney, heart, spleen, brain, and liver and caused disease as evidenced by weight loss and elevation of aspartate and alanine aminotransferase (AST and ALT) levels with a high AST/ALT ratio. Lassa fever-like pathology included acute hepatitis, interstitial pneumonia, and pronounced disturbance of splenic cytoarchitecture. Infiltrations of activated monocytes/macrophages expressing inducible nitric oxide synthase and T cells were found in liver and lung. In contrast, Mopeia, Morogoro, and Mobala virus replicated poorly in the animals and acute inflammatory alterations were not noted. Depletion of CD4(+) and CD8(+) T cells strongly enhanced susceptibility of IFNAR(-/-) mice to the apathogenic viruses. In conclusion, the virulence of Old World arenaviruses in IFNAR(-/-) mice correlates with their virulence in humans and non-human primates. In addition to the type I interferon system, T cells seem to regulate whether or not an arenavirus can productively infect non-host rodent species. The observation that Lassa virus overcomes the species barrier without artificial depletion of T cells suggests it is able to impair T cell functionality in a way that corresponds to depletion.


Subject(s)
Arenaviridae Infections/virology , Arenavirus/pathogenicity , Receptor, Interferon alpha-beta/physiology , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/pathology , Host-Pathogen Interactions , Lymphocyte Depletion , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , T-Lymphocytes/immunology , Virulence
17.
Viruses ; 5(1): 340-51, 2013 Jan 21.
Article in English | MEDLINE | ID: mdl-23337384

ABSTRACT

Viral hemorrhagic fevers (VHFs) caused by arenaviruses are acute diseases characterized by fever, headache, general malaise, impaired cellular immunity, eventual neurologic involvement, and hemostatic alterations that may ultimately lead to shock and death. The causes of the bleeding are still poorly understood. However, it is generally accepted that these causes are associated to some degree with impaired hemostasis, endothelial cell dysfunction and low platelet counts or function. In this article, we present the current knowledge about the hematological alterations present in VHF induced by arenaviruses, including new aspects on the underlying pathogenic mechanisms.


Subject(s)
Arenaviridae Infections/virology , Arenavirus/pathogenicity , Hemorrhagic Fevers, Viral/virology , Animals , Arenavirus/genetics , Arenavirus/physiology , Blood Coagulation Factors/metabolism , Hemorrhagic Fevers, Viral/blood , Hemorrhagic Fevers, Viral/metabolism , Hemorrhagic Fevers, Viral/physiopathology , Humans , Platelet Count
18.
Antiviral Res ; 97(2): 81-92, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23261843

ABSTRACT

A number of arenaviruses are pathogenic for humans, but they differ significantly in virulence. Lassa virus, found in West Africa, causes severe hemorrhagic fever (HF), while the other principal Old World arenavirus, lymphocytic choriomeningitis virus, causes mild illness in persons with normal immune function, and poses a threat only to immunocompromised individuals. The New World agents, including Junin, Machupo and Sabia virus, are highly pathogenic for humans. Arenaviral HF is characterized by high viremia and general immune suppression, the mechanism of which is unknown. Studies using viral reverse genetics, cell-based assays, animal models and human genome-wide association analysis have revealed potential mechanisms by which arenaviruses cause severe disease in humans. Each of the four viral gene products (GPC, L polymerase, NP, and Z matrix protein) and several host-cell factors (e.g., α-dystroglycan) are responsible for mediating viral entry, genome replication, and the inhibition of apoptosis, translation and interferon-beta (IFNß) production. This review summarizes current knowledge of the role of each viral protein and host factor in the pathogenesis of arenaviral HF. Insights from recent studies are being exploited for the development of novel therapies.


Subject(s)
Arenavirus/pathogenicity , Hemorrhagic Fever, American/pathology , Hemorrhagic Fever, American/virology , Host-Pathogen Interactions , Viral Proteins/metabolism , Virulence Factors/metabolism , Hemorrhagic Fever, American/prevention & control , Hemorrhagic Fever, American/therapy , Humans , Viral Proteins/antagonists & inhibitors , Virulence Factors/antagonists & inhibitors
19.
Viruses ; 4(10): 2049-79, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-23202453

ABSTRACT

Arenaviruses have a bisegmented negative-strand RNA genome, which encodes four viral proteins: GP and NP by the S segment and L and Z by the L segment. These four viral proteins possess multiple functions in infection, replication and release of progeny viruses from infected cells. The small RING finger protein, Z protein is a matrix protein that plays a central role in viral assembly and budding. Although all arenaviruses encode Z protein, amino acid sequence alignment showed a huge variety among the species, especially at the C-terminus where the L-domain is located. Recent publications have demonstrated the interactions between viral protein and viral protein, and viral protein and host cellular protein, which facilitate transportation and assembly of viral components to sites of virus egress. This review presents a summary of current knowledge regarding arenavirus assembly and budding, in comparison with other enveloped viruses. We also refer to the restriction of arenavirus production by the antiviral cellular factor, Tetherin/BST-2.


Subject(s)
Arenavirus/physiology , Genome, Viral , Virus Assembly , Virus Release , Amino Acid Sequence , Animals , Arenaviridae Infections/pathology , Arenaviridae Infections/virology , Arenavirus/genetics , Arenavirus/metabolism , Arenavirus/pathogenicity , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cell Membrane/virology , Endosomal Sorting Complexes Required for Transport/metabolism , Host-Pathogen Interactions , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Protein Structure, Tertiary , Protein Transport , Structure-Activity Relationship
20.
Viruses ; 4(10): 2162-81, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-23202458

ABSTRACT

Arenaviruses include lethal human pathogens which pose serious public health threats. So far, no FDA approved vaccines are available against arenavirus infections, and therapeutic options are limited, making the identification of novel drug targets for the development of efficacious therapeutics an urgent need. Arenaviruses are comprised of two RNA genome segments and four proteins, the polymerase L, the envelope glycoprotein GP, the matrix protein Z, and the nucleoprotein NP. A crucial step in the arenavirus life-cycle is the biosynthesis and maturation of the GP precursor (GPC) by cellular signal peptidases and the cellular enzyme Subtilisin Kexin Isozyme-1 (SKI-1)/Site-1 Protease (S1P) yielding a tripartite mature GP complex formed by GP1/GP2 and a stable signal peptide (SSP). GPC cleavage by SKI-1/S1P is crucial for fusion competence and incorporation of mature GP into nascent budding virion particles. In a first part of our review, we cover basic aspects and newer developments in the biosynthesis of arenavirus GP and its molecular interaction with SKI-1/S1P. A second part will then highlight the potential of SKI-1/S1P-mediated processing of arenavirus GPC as a novel target for therapeutic intervention to combat human pathogenic arenaviruses.


Subject(s)
Arenavirus/metabolism , Proprotein Convertases/metabolism , Serine Endopeptidases/metabolism , Viral Envelope Proteins/biosynthesis , Amino Acid Sequence , Antiviral Agents/pharmacology , Arenaviridae Infections/drug therapy , Arenaviridae Infections/metabolism , Arenaviridae Infections/virology , Arenavirus/drug effects , Arenavirus/pathogenicity , Glycosylation , Golgi Apparatus/metabolism , Golgi Apparatus/virology , Humans , Proprotein Convertases/antagonists & inhibitors , Protein Sorting Signals , Proteolysis , Pyrrolidines/pharmacology , Receptors, Cell Surface/metabolism , Viral Envelope Proteins/chemistry , Virus Assembly , Virus Attachment
SELECTION OF CITATIONS
SEARCH DETAIL
...