Arenaviruses

A special issue of Viruses (ISSN 1999-4915). This special issue belongs to the section "Animal Viruses".

Deadline for manuscript submissions: closed (30 November 2012) | Viewed by 203103

Special Issue Editor

Institute of Microbiology, University Hospital Center and University of Lausanne, Rue du Bugnon 48, 1011 Lausanne, Switzerland

Special Issue Information

Dear Colleagues,

The arenaviruses are a large and diverse family of viruses that merit significant attention as powerful models for experimental virology and as important human pathogens. Over the past decades several arenaviruses have emerged as causative agents of severe viral hemorrhagic fevers that belong to the most devastating human diseases and cause considerable suffering in many countries of the Developing World. The Old World arenavirus Lassa virus is the most prevalent human pathogen among the arenaviruses with several hundred thousand infections per year in Africa with thousands of deaths. The South American hemorrhagic fever viruses Junin, Machupo, Guanarito, and Sabia have emerged as etiological agents of severe hemorrhagic fevers in Latin America. There is no licensed vaccine available and therapeutic options are restricted, resulting in 15-30% mortality in hospitalized patients. New pathogenic arenaviruses emerge with on average one new species being discovered every three years, representing a serious concern for public health.The past years have seen rapid progress in many aspects of arenavirus research. The present Special Issue covers major recent developments in molecular arenavirus virology, fundamental mechanisms of arenavirus-host cell interaction, and the development of novel anti-viral strategies against these important pathogens.

Prof. Dr. Stefan Kunz
Guest Editor

Keywords

  • arenaviruses
  • viral hemorrhagic fevers
  • emerging viruses
  • virus-host cell interaction
  • anti-viral drugs

Published Papers (18 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

1920 KiB  
Article
D471G Mutation in LCMV-NP Affects Its Ability to Self-associate and Results in a Dominant Negative Effect in Viral RNA Synthesis
by Emilio Ortiz-Riaño, Benson Y.H. Cheng, Juan C. de la Torre and Luis Martínez-Sobrido
Viruses 2012, 4(10), 2137-2161; https://doi.org/10.3390/v4102137 - 16 Oct 2012
Cited by 14 | Viewed by 7512
Abstract
Arenaviruses merit significant interest because several family members are etiological agents of severe hemorrhagic fevers, representing a major burden to public health. Currently, there are no FDA-licensed vaccines against arenaviruses and the only available antiviral therapy is limited to the use of ribavirin [...] Read more.
Arenaviruses merit significant interest because several family members are etiological agents of severe hemorrhagic fevers, representing a major burden to public health. Currently, there are no FDA-licensed vaccines against arenaviruses and the only available antiviral therapy is limited to the use of ribavirin that is partially effective. Arenavirus nucleoprotein (NP) is found associated with the genomic RNA forming the viral ribonucleoproteins (vRNPs) that together with the polymerase (L) direct viral replication and transcription. Virion formation requires the recruitment of vRNPs into budding sites, a process in which the arenavirus matrix-like protein (Z) plays a major role. Therefore, proper NP-NP and NP-Z interactions are required for the generation of infectious progeny. In this work we demonstrate the role of the amino acid residue D471 in the self-association of lymphocytic choriomeningitis virus nucleoprotein (LCMV-NP). Amino acid substitutions at this position abrogate NP oligomerization, affecting its ability to mediate replication and transcription of a minigenome reporter plasmid. However, its ability to interact with the Z protein, counteract the cellular interferon response and bind to dsRNA analogs was retained. Additionally, we also document the dominant negative effect of D471G mutation on viral infection, suggesting that NP self-association is an excellent target for the development of new antivirals against arenaviruses. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

401 KiB  
Article
Animal Models, Prophylaxis, and Therapeutics for Arenavirus Infections
by Eric Vela
Viruses 2012, 4(9), 1802-1829; https://doi.org/10.3390/v4091802 - 24 Sep 2012
Cited by 26 | Viewed by 7758
Abstract
Arenaviruses are enveloped, bipartite negative single-stranded RNA viruses that can cause a wide spectrum of disease in humans and experimental animals including hemorrhagic fever. The majority of these viruses are rodent-borne and the arenavirus family can be divided into two groups: the Lassa-Lymphocytic [...] Read more.
Arenaviruses are enveloped, bipartite negative single-stranded RNA viruses that can cause a wide spectrum of disease in humans and experimental animals including hemorrhagic fever. The majority of these viruses are rodent-borne and the arenavirus family can be divided into two groups: the Lassa-Lymphocytic choriomeningitis serocomplex and the Tacaribe serocomplex. Arenavirus-induced disease may include characteristic symptoms ranging from fever, malaise, body aches, petechiae, dehydration, hemorrhage, organ failure, shock, and in severe cases death. Currently, there are few prophylactic and therapeutic treatments available for arenavirus-induced symptoms. Supportive care and ribavirin remain the predominant strategies for treating most of the arenavirus-induced diseases. Therefore, efficacy testing of novel therapeutic and prophylactic strategies in relevant animal models is necessary. Because of the potential for person-to-person spread, the ability to cause lethal or debilitating disease in humans, limited treatment options, and potential as a bio-weapon, the development of prophylactics and therapeutics is essential. This article reviews the current arenavirus animal models and prophylactic and therapeutic strategies under development to treat arenavirus infection. Full article
(This article belongs to the Special Issue Arenaviruses)

Review

Jump to: Research

1457 KiB  
Review
Arenavirus Budding: A Common Pathway with Mechanistic Differences
by Svenja Wolff, Hideki Ebihara and Allison Groseth
Viruses 2013, 5(2), 528-549; https://doi.org/10.3390/v5020528 - 31 Jan 2013
Cited by 25 | Viewed by 9044
Abstract
The Arenaviridae is a diverse and growing family of viruses that includes several agents responsible for important human diseases. Despite the importance of this family for public health, particularly in Africa and South America, much of its biology remains poorly understood. However, in [...] Read more.
The Arenaviridae is a diverse and growing family of viruses that includes several agents responsible for important human diseases. Despite the importance of this family for public health, particularly in Africa and South America, much of its biology remains poorly understood. However, in recent years significant progress has been made in this regard, particularly relating to the formation and release of new enveloped virions, which is an essential step in the viral lifecycle. While this process is mediated chiefly by the viral matrix protein Z, recent evidence suggests that for some viruses the nucleoprotein (NP) is also required to enhance the budding process. Here we highlight and compare the distinct budding mechanisms of different arenaviruses, concentrating on the role of the matrix protein Z, its known late domain sequences, and the involvement of cellular endosomal sorting complex required for transport (ESCRT) pathway components. Finally we address the recently described roles for the nucleoprotein NP in budding and ribonucleoprotein complex (RNP) incorporation, as well as discussing possible mechanisms related to its involvement. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

476 KiB  
Review
Pathogenic Mechanisms Involved in the Hematological Alterations of Arenavirus-induced Hemorrhagic Fevers
by Mirta Schattner, Leonardo Rivadeneyra, Roberto G. Pozner and Ricardo M. Gómez
Viruses 2013, 5(1), 340-351; https://doi.org/10.3390/v5010340 - 21 Jan 2013
Cited by 17 | Viewed by 6994
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, [...] Read more.
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. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

862 KiB  
Review
Arenavirus Variations Due to Host-Specific Adaptation
by Juan C. Zapata and Maria S. Salvato
Viruses 2013, 5(1), 241-278; https://doi.org/10.3390/v5010241 - 17 Jan 2013
Cited by 47 | Viewed by 11127
Abstract
Arenavirus particles are enveloped and contain two single-strand RNA genomic segments with ambisense coding. Genetic plasticity of the arenaviruses comes from transcription errors, segment reassortment, and permissive genomic packaging, and results in their remarkable ability, as a group, to infect a wide variety [...] Read more.
Arenavirus particles are enveloped and contain two single-strand RNA genomic segments with ambisense coding. Genetic plasticity of the arenaviruses comes from transcription errors, segment reassortment, and permissive genomic packaging, and results in their remarkable ability, as a group, to infect a wide variety of hosts. In this review, we discuss some in vitro studies of virus genetic and phenotypic variation after exposure to selective pressures such as high viral dose, mutagens and antivirals. Additionally, we discuss the variation in vivo of selected isolates of Old World arenaviruses, particularly after infection of different animal species. We also discuss the recent emergence of new arenaviruses in the context of our observations of sequence variations that appear to be host-specific. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

802 KiB  
Review
A Systems Biology Starter Kit for Arenaviruses
by Magali E. Droniou-Bonzom and Paula M. Cannon
Viruses 2012, 4(12), 3625-3646; https://doi.org/10.3390/v4123625 - 11 Dec 2012
Cited by 33 | Viewed by 6497
Abstract
Systems biology approaches in virology aim to integrate viral and host biological networks, and thus model the infection process. The growing availability of high-throughput “-omics” techniques and datasets, as well as the ever-increasing sophistication of in silico modeling tools, has resulted in a [...] Read more.
Systems biology approaches in virology aim to integrate viral and host biological networks, and thus model the infection process. The growing availability of high-throughput “-omics” techniques and datasets, as well as the ever-increasing sophistication of in silico modeling tools, has resulted in a corresponding rise in the complexity of the analyses that can be performed. The present study seeks to review and organize published evidence regarding virus-host interactions for the arenaviruses, from alterations in the host proteome during infection, to reported protein-protein interactions. In this way, we hope to provide an overview of the interplay between arenaviruses and the host cell, and lay the foundations for complementing current arenavirus research with a systems-level approach. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Graphical abstract

2571 KiB  
Review
Multifunctional Nature of the Arenavirus RING Finger Protein Z
by Sarah Katharina Fehling, Frank Lennartz and Thomas Strecker
Viruses 2012, 4(11), 2973-3011; https://doi.org/10.3390/v4112973 - 09 Nov 2012
Cited by 59 | Viewed by 14538
Abstract
Arenaviruses are a family of enveloped negative-stranded RNA viruses that can cause severe human disease ranging from encephalitis symptoms to fulminant hemorrhagic fever. The bi‑segmented RNA genome encodes four polypeptides: the nucleoprotein NP, the surface glycoprotein GP, the polymerase L, and the RING [...] Read more.
Arenaviruses are a family of enveloped negative-stranded RNA viruses that can cause severe human disease ranging from encephalitis symptoms to fulminant hemorrhagic fever. The bi‑segmented RNA genome encodes four polypeptides: the nucleoprotein NP, the surface glycoprotein GP, the polymerase L, and the RING finger protein Z. Although it is the smallest arenavirus protein with a length of 90 to 99 amino acids and a molecular weight of approx. 11 kDa, the Z protein has multiple functions in the viral life cycle including (i) regulation of viral RNA synthesis, (ii) orchestration of viral assembly and budding, (iii) interaction with host cell proteins, and (iv) interferon antagonism. In this review, we summarize our current understanding of the structural and functional role of the Z protein in the arenavirus replication cycle. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

979 KiB  
Review
Arenaviruses and Lethal Mutagenesis. Prospects for New Ribavirin-based Interventions
by Héctor Moreno, Ana Grande-Pérez, Esteban Domingo and Verónica Martín
Viruses 2012, 4(11), 2786-2805; https://doi.org/10.3390/v4112786 - 06 Nov 2012
Cited by 23 | Viewed by 7395
Abstract
Lymphocytic choriomeningitis virus (LCMV) has contributed to unveil some of the molecular mechanisms of lethal mutagenesis, or loss of virus infectivity due to increased mutation rates. Here we review these developments, and provide additional evidence that ribavirin displays a dual mutagenic and inhibitory [...] Read more.
Lymphocytic choriomeningitis virus (LCMV) has contributed to unveil some of the molecular mechanisms of lethal mutagenesis, or loss of virus infectivity due to increased mutation rates. Here we review these developments, and provide additional evidence that ribavirin displays a dual mutagenic and inhibitory activity on LCMV that can be relevant to treatment designs. Using 5-fluorouracil as mutagenic agent and ribavirin either as inhibitor or mutagen, we document an advantage of a sequential inhibitor-mutagen administration over the corresponding combination treatment to achieve a low LCMV load in cell culture. This advantage is accentuated in the concentration range in which ribavirin acts mainly as an inhibitor, rather than as mutagen. This observation reinforces previous theoretical and experimental studies in supporting a sequential inhibitor-mutagen administration as a possible antiviral design. Given recent progress in the development of new inhibitors of arenavirus replication, our results suggest new options of ribavirin-based anti-arenavirus treatments. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

1175 KiB  
Review
Immune Responses and Lassa Virus Infection
by Marion Russier, Delphine Pannetier and Sylvain Baize
Viruses 2012, 4(11), 2766-2785; https://doi.org/10.3390/v4112766 - 05 Nov 2012
Cited by 38 | Viewed by 11320
Abstract
Lassa fever is a hemorrhagic fever endemic to West Africa and caused by Lassa virus, an Old World arenavirus. It may be fatal, but most patients recover from acute disease and some experience asymptomatic infection. The immune mechanisms associated with these different outcomes [...] Read more.
Lassa fever is a hemorrhagic fever endemic to West Africa and caused by Lassa virus, an Old World arenavirus. It may be fatal, but most patients recover from acute disease and some experience asymptomatic infection. The immune mechanisms associated with these different outcomes have not yet been fully elucidated, but considerable progress has recently been made, through the use of in vitro human models and nonhuman primates, the only relevant animal model that mimics the pathophysiology and immune responses induced in patients. We discuss here the roles of the various components of the innate and adaptive immune systems in Lassa virus infection and in the control of viral replication and pathogenesis. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

726 KiB  
Review
Role of Lymphocytic Choriomeningitis Virus (LCMV) in Understanding Viral Immunology: Past, Present and Future
by Xin Zhou, Srividya Ramachandran, Margaret Mann and Daniel L. Popkin
Viruses 2012, 4(11), 2650-2669; https://doi.org/10.3390/v4112650 - 29 Oct 2012
Cited by 90 | Viewed by 21329
Abstract
Lymphocytic choriomeningitis virus (LCMV) is a common infection of rodents first identified over eighty years ago in St. Louis, MO, U.S.A. It is best known for its application in immunological studies. The history of LCMV closely correlates with the development of modern immunology. [...] Read more.
Lymphocytic choriomeningitis virus (LCMV) is a common infection of rodents first identified over eighty years ago in St. Louis, MO, U.S.A. It is best known for its application in immunological studies. The history of LCMV closely correlates with the development of modern immunology. With the use of LCMV as a model pathogen several key concepts have emerged: Major Histocompatibility Complex (MHC) restriction, T cell memory, persistent infections, T cell exhaustion and the key role of immune pathology in disease. Given the phenomenal infrastructure within this field (e.g., defined immunodominant and subdominant epitopes to all T cell receptor specificities as well as the cognate tetramers for enumeration in vivo) the study of LCMV remains an active and productive platform for biological research across the globe to this day. Here we present a historical primer that highlights several breakthroughs since the discovery of LCMV. Next, we highlight current research in the field and conclude with our predictions for future directions in the remarkable field of LCMV research. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

1116 KiB  
Review
Advanced Vaccine Candidates for Lassa Fever
by Igor S. Lukashevich
Viruses 2012, 4(11), 2514-2557; https://doi.org/10.3390/v4112514 - 29 Oct 2012
Cited by 42 | Viewed by 11810
Abstract
Lassa virus (LASV) is the most prominent human pathogen of the Arenaviridae. The virus is transmitted to humans by a rodent reservoir, Mastomys natalensis, and is capable of causing lethal Lassa Fever (LF). LASV has the highest human impact of any of the [...] Read more.
Lassa virus (LASV) is the most prominent human pathogen of the Arenaviridae. The virus is transmitted to humans by a rodent reservoir, Mastomys natalensis, and is capable of causing lethal Lassa Fever (LF). LASV has the highest human impact of any of the viral hemorrhagic fevers (with the exception of Dengue Fever) with an estimated several hundred thousand infections annually, resulting in thousands of deaths in Western Africa. The sizeable disease burden, numerous imported cases of LF in non-endemic countries, and the possibility that LASV can be used as an agent of biological warfare make a strong case for vaccine development. Presently there is no licensed vaccine against LF or approved treatment. Recently, several promising vaccine candidates have been developed which can potentially target different groups at risk. The purpose of this manuscript is to review the LASV pathogenesis and immune mechanisms involved in protection. The current status of pre-clinical development of the advanced vaccine candidates that have been tested in non-human primates will be discussed. Major scientific, manufacturing, and regulatory challenges will also be considered. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

917 KiB  
Review
Junín Virus Pathogenesis and Virus Replication
by Ashley Grant, Alexey Seregin, Cheng Huang, Olga Kolokoltsova, Allan Brasier, Clarence Peters and Slobodan Paessler
Viruses 2012, 4(10), 2317-2339; https://doi.org/10.3390/v4102317 - 19 Oct 2012
Cited by 65 | Viewed by 13052
Abstract
Junín virus, the etiological agent of Argentine hemorrhagic fever, causes significant morbidity and mortality. The virus is spread through the aerosolization of host rodent excreta and endemic to the humid pampas of Argentina. Recently, significant progress has been achieved with the development of [...] Read more.
Junín virus, the etiological agent of Argentine hemorrhagic fever, causes significant morbidity and mortality. The virus is spread through the aerosolization of host rodent excreta and endemic to the humid pampas of Argentina. Recently, significant progress has been achieved with the development of new technologies (e.g. reverse genetics) that have expanded knowledge about the pathogenesis and viral replication of Junín virus. We will review the pathogenesis of Junín virus in various animal models and the role of innate and adaptive immunity during infection. We will highlight current research regarding the role of molecular biology of Junín virus in elucidating virus attenuation. We will also summarize current knowledge on Junín virus pathogenesis focusing on the recent development of vaccines and potential therapeutics. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

564 KiB  
Review
Arenavirus Evasion of Host Anti-Viral Responses
by Melissa Hayes and Maria Salvato
Viruses 2012, 4(10), 2182-2196; https://doi.org/10.3390/v4102182 - 17 Oct 2012
Cited by 11 | Viewed by 7160
Abstract
The innate response to infection by an Old World arenavirus is initiated and mediated by extracellular and intracellular receptors, and effector molecules. In response, the invading virus has evolved to inhibit these responses and create the best environment possible for replication and spread. [...] Read more.
The innate response to infection by an Old World arenavirus is initiated and mediated by extracellular and intracellular receptors, and effector molecules. In response, the invading virus has evolved to inhibit these responses and create the best environment possible for replication and spread. Here, we will discuss both the host’s response to infection with data from human infection and lessons learned from animal models, as well as the multitude of ways the virus combats the resulting immune response. Finally, we will highlight recent work identifying TLR2 as an innate sensor for arenaviruses and how the TLR2-dependent response differs depending on the pathogenicity of the strain. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

808 KiB  
Review
Envelope Glycoprotein of Arenaviruses
by Dominique J. Burri, Joel Ramos Da Palma, Stefan Kunz and Antonella Pasquato
Viruses 2012, 4(10), 2162-2181; https://doi.org/10.3390/v4102162 - 17 Oct 2012
Cited by 72 | Viewed by 16426
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. [...] Read more.
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. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

416 KiB  
Review
Serological Assays Based on Recombinant Viral Proteins for the Diagnosis of Arenavirus Hemorrhagic Fevers
by Shuetsu Fukushi, Hideki Tani, Tomoki Yoshikawa, Masayuki Saijo and Shigeru Morikawa
Viruses 2012, 4(10), 2097-2114; https://doi.org/10.3390/v4102097 - 12 Oct 2012
Cited by 22 | Viewed by 8602
Abstract
The family Arenaviridae, genus Arenavirus, consists of two phylogenetically independent groups: Old World (OW) and New World (NW) complexes. The Lassa and Lujo viruses in the OW complex and the Guanarito, Junin, Machupo, Sabia, and Chapare viruses in the NW complex cause [...] Read more.
The family Arenaviridae, genus Arenavirus, consists of two phylogenetically independent groups: Old World (OW) and New World (NW) complexes. The Lassa and Lujo viruses in the OW complex and the Guanarito, Junin, Machupo, Sabia, and Chapare viruses in the NW complex cause viral hemorrhagic fever (VHF) in humans, leading to serious public health concerns. These viruses are also considered potential bioterrorism agents. Therefore, it is of great importance to detect these pathogens rapidly and specifically in order to minimize the risk and scale of arenavirus outbreaks. However, these arenaviruses are classified as BSL-4 pathogens, thus making it difficult to develop diagnostic techniques for these virus infections in institutes without BSL-4 facilities. To overcome these difficulties, antibody detection systems in the form of an enzyme-linked immunosorbent assay (ELISA) and an indirect immunofluorescence assay were developed using recombinant nucleoproteins (rNPs) derived from these viruses. Furthermore, several antigen-detection assays were developed. For example, novel monoclonal antibodies (mAbs) to the rNPs of Lassa and Junin viruses were generated. Sandwich antigen-capture (Ag-capture) ELISAs using these mAbs as capture antibodies were developed and confirmed to be sensitive and specific for detecting the respective arenavirus NPs. These rNP-based assays were proposed to be useful not only for an etiological diagnosis of VHFs, but also for seroepidemiological studies on VHFs. We recently developed arenavirus neutralization assays using vesicular stomatitis virus (VSV)-based pseudotypes bearing arenavirus recombinant glycoproteins. The goal of this article is to review the recent advances in developing laboratory diagnostic assays based on recombinant viral proteins for the diagnosis of VHFs and epidemiological studies on the VHFs caused by arenaviruses. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

2136 KiB  
Review
Molecular Mechanism of Arenavirus Assembly and Budding
by Shuzo Urata and Jiro Yasuda
Viruses 2012, 4(10), 2049-2079; https://doi.org/10.3390/v4102049 - 10 Oct 2012
Cited by 28 | Viewed by 10308
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 [...] Read more.
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. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

319 KiB  
Review
Pathogenesis of Lassa Fever
by Nadezhda E. Yun and David H. Walker
Viruses 2012, 4(10), 2031-2048; https://doi.org/10.3390/v4102031 - 09 Oct 2012
Cited by 144 | Viewed by 21749
Abstract
Lassa virus, an Old World arenavirus (family Arenaviridae), is the etiological agent of Lassa fever, a severe human disease that is reported in more than 100,000 patients annually in the endemic regions of West Africa with mortality rates for hospitalized patients varying between [...] Read more.
Lassa virus, an Old World arenavirus (family Arenaviridae), is the etiological agent of Lassa fever, a severe human disease that is reported in more than 100,000 patients annually in the endemic regions of West Africa with mortality rates for hospitalized patients varying between 5-10%. Currently, there are no approved vaccines against Lassa fever for use in humans. Here, we review the published literature on the life cycle of Lassa virus with the specific focus put on Lassa fever pathogenesis in humans and relevant animal models. Advancing knowledge significantly improves our understanding of Lassa virus biology, as well as of the mechanisms that allow the virus to evade the host’s immune system. However, further investigations are required in order to design improved diagnostic tools, an effective vaccine, and therapeutic agents. Full article
(This article belongs to the Special Issue Arenaviruses)
760 KiB  
Review
Host Cell Factors as Antiviral Targets in Arenavirus Infection
by Florencia N. Linero, Claudia S. Sepúlveda, Federico Giovannoni, Viviana Castilla, Cybele C. García, Luis A. Scolaro and Elsa B. Damonte
Viruses 2012, 4(9), 1569-1591; https://doi.org/10.3390/v4091569 - 13 Sep 2012
Cited by 23 | Viewed by 8641
Abstract
Among the members of the Arenaviridae family, Lassa virus and Junin virus generate periodic annual outbreaks of severe human hemorrhagic fever (HF) in endemic areas of West Africa and Argentina, respectively. Given the human health threat that arenaviruses represent and the lack of [...] Read more.
Among the members of the Arenaviridae family, Lassa virus and Junin virus generate periodic annual outbreaks of severe human hemorrhagic fever (HF) in endemic areas of West Africa and Argentina, respectively. Given the human health threat that arenaviruses represent and the lack of a specific and safe chemotherapy, the search for effective antiviral compounds is a continuous demanding effort. Since diverse host cell pathways and enzymes are used by RNA viruses to fulfill their replicative cycle, the targeting of a host process has turned an attractive antiviral approach in the last years for many unrelated virus types. This strategy has the additional benefit to reduce the serious challenge for therapy of RNA viruses to escape from drug effects through selection of resistant variants triggered by their high mutation rate. This article focuses on novel strategies to identify inhibitors for arenavirus therapy, analyzing the potential for antiviral developments of diverse host factors essential for virus infection. Full article
(This article belongs to the Special Issue Arenaviruses)
Show Figures

Figure 1

Back to TopTop