Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Viruses ; 14(9)2022 09 12.
Article in English | MEDLINE | ID: mdl-36146824

ABSTRACT

Arenaviruses include important zoonotic pathogens that cause hemorrhagic fever (e.g., Junín virus; JUNV) as well as other viruses that are closely related but apathogenic (e.g., Tacaribe virus; TCRV). We have found that, while TCRV and JUNV differ in their ability to induce apoptosis in infected cells, due to active inhibition of caspase activation by the JUNV nucleoprotein, both viruses trigger similar upstream pro-apoptotic signaling events, including the activation/phosphorylation of p53. In the case of TCRV, the pro-apoptotic factor Bad is also phosphorylated (leading to its inactivation). These events clearly implicate upstream kinases in regulating the induction of apoptosis. Consistent with this, here we show activation in TCRV-infected cells of the stress-activated protein kinases p38 and JNK, which are known to regulate p53 activation, as well as the downstream kinase MK2 and transcription factor c-Jun. We also observed the early transient activation of Akt, but not Erk. Importantly, the chemical inhibition of Akt, p38, JNK and c-Jun all dramatically reduced viral growth, even though we have shown that inhibition of apoptosis itself does not. This indicates that kinase activation is crucial for viral infection, independent of its downstream role in apoptosis regulation, a finding that has the potential to shed further light on the determinants of arenavirus pathogenesis, as well as to inform future therapeutic approaches.


Subject(s)
Arenaviridae Infections , Arenaviruses, New World , Apoptosis , Arenaviruses, New World/physiology , Caspases , Humans , Nucleoproteins , Proto-Oncogene Proteins c-akt , Transcription Factors , Tumor Suppressor Protein p53 , Virus Replication , p38 Mitogen-Activated Protein Kinases
2.
J Virol ; 95(17): e0186820, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34132574

ABSTRACT

Pathogenic clade B New World mammarenaviruses (NWM) can cause Argentine, Venezuelan, Brazilian, and Bolivian hemorrhagic fevers. Sequence variability among NWM glycoproteins (GP) poses a challenge to the development of broadly neutralizing therapeutics against the entire clade of viruses. However, blockade of their shared binding site on the apical domain of human transferrin receptor 1 (hTfR1/CD71) presents an opportunity for the development of effective and broadly neutralizing therapeutics. Here, we demonstrate that the murine monoclonal antibody OKT9, which targets the apical domain of hTfR1, can sterically block cellular entry by viral particles presenting clade B NWM glycoproteins (GP1-GP2). OKT9 blockade is also effective against viral particles pseudotyped with glycoproteins of a recently identified pathogenic Sabia-like virus. With nanomolar affinity for hTfR1, the OKT9 antigen binding fragment (OKT9-Fab) sterically blocks clade B NWM-GP1s and reduces infectivity of an attenuated strain of Junin virus. Binding of OKT9 to the hTfR1 ectodomain in its soluble, dimeric state produces stable assemblies that are observable by negative-stain electron microscopy. A model of the OKT9-sTfR1 complex, informed by the known crystallographic structure of sTfR1 and a newly determined structure of the OKT9 antigen binding fragment (Fab), suggests that OKT9 and the Machupo virus GP1 share a binding site on the hTfR1 apical domain. The structural basis for this interaction presents a framework for the design and development of high-affinity, broadly acting agents targeting clade B NWMs. IMPORTANCE Pathogenic clade B NWMs cause grave infectious diseases, the South American hemorrhagic fevers. Their etiological agents are Junin (JUNV), Guanarito (GTOV), Sabiá (SABV), Machupo (MACV), Chapare (CHAV), and a new Sabiá-like (SABV-L) virus recently identified in Brazil. These are priority A pathogens due to their high infectivity and mortality, their potential for person-to-person transmission, and the limited availability of effective therapeutics and vaccines to curb their effects. While low homology between surface glycoproteins of NWMs foils efforts to develop broadly neutralizing therapies targeting NWMs, this work provides structural evidence that OKT9, a monoclonal antibody targeting a single NWM glycoprotein binding site on hTfR1, can efficiently prevent their entry into cells.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/administration & dosage , Arenaviruses, New World/physiology , Glycoproteins/immunology , Hemorrhagic Fever, American/prevention & control , Receptors, Transferrin/immunology , A549 Cells , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/virology , Humans , Protein Structure, Tertiary , Receptors, Transferrin/chemistry , Receptors, Transferrin/genetics
3.
PLoS Pathog ; 16(10): e1008948, 2020 10.
Article in English | MEDLINE | ID: mdl-33045019

ABSTRACT

Pathogenicity often differs dramatically among even closely related arenavirus species. For instance, Junín virus (JUNV), the causative agent of Argentine hemorrhagic fever (AHF), is closely related to Tacaribe virus (TCRV), which is normally avirulent in humans. While little is known about how host cell pathways are regulated in response to arenavirus infection, or how this contributes to virulence, these two viruses have been found to differ markedly in their ability to induce apoptosis. However, details of the mechanism(s) governing the apoptotic response to arenavirus infections are unknown. Here we confirm that TCRV-induced apoptosis is mitochondria-regulated, with associated canonical hallmarks of the intrinsic apoptotic pathway, and go on to identify the pro- and anti-apoptotic Bcl-2 factors responsible for regulating this process. In particular, levels of the pro-apoptotic BH3-only proteins Noxa and Puma, as well as their canonical transcription factor p53, were strongly increased. Interestingly, TCRV infection also led to the accumulation of the inactive phosphorylated form of another pro-apoptotic BH3-only protein, Bad (i.e. as phospho-Bad). Knockout of Noxa or Puma suppressed apoptosis in response to TCRV infection, whereas silencing of Bad increased apoptosis, confirming that these factors are key regulators of apoptosis induction in response to TCRV infection. Further, we found that while the highly pathogenic JUNV does not induce caspase activation, it still activated upstream pro-apoptotic factors, consistent with current models suggesting that JUNV evades apoptosis by interfering with caspase activation through a nucleoprotein-mediated decoy function. This new mechanistic insight into the role that individual BH3-only proteins and their regulation play in controlling apoptotic fate in arenavirus-infected cells provides an important experimental framework for future studies aimed at dissecting differences in the apoptotic responses between arenaviruses, their connection to other cell signaling events and ultimately the relationship of these processes to pathogenesis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Arenaviridae Infections/pathology , Arenaviruses, New World/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Virus Replication , bcl-Associated Death Protein/metabolism , Apoptosis Regulatory Proteins/genetics , Arenaviridae Infections/genetics , Arenaviridae Infections/metabolism , Arenaviridae Infections/virology , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Protein Domains , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-Associated Death Protein/genetics
4.
Proc Natl Acad Sci U S A ; 117(32): 19497-19506, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32719120

ABSTRACT

Understanding the genetics of susceptibility to infectious agents is of great importance to our ability to combat disease. Here, we show that voltage-gated calcium channels (VGCCs) are critical for cellular binding and entry of the New World arenaviruses Junín and Tacaribe virus, suggesting that zoonosis via these receptors could occur. Moreover, we demonstrate that α1s haploinsufficiency renders cells and mice more resistant to infection by these viruses. In addition to being more resistant to infection, haploinsufficient cells and mice required a lower dosage of VGCC antagonists to block infection. These studies underscore the importance of genetic variation in susceptibility to both viruses and pharmaceutics.


Subject(s)
Arenaviridae Infections/genetics , Calcium Channels, L-Type/genetics , Disease Resistance/genetics , Animals , Arenaviridae Infections/drug therapy , Arenaviruses, New World/physiology , Calcium Channel Agonists/pharmacology , Calcium Channel Agonists/therapeutic use , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, L-Type/deficiency , Calcium Channels, L-Type/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Heterozygote , Humans , Mice , Mice, Mutant Strains , Mutation , Virus Attachment/drug effects , Virus Internalization/drug effects
5.
Viruses ; 10(4)2018 04 17.
Article in English | MEDLINE | ID: mdl-29673133

ABSTRACT

We have previously shown that the infection of cell cultures with the arenaviruses Junín (JUNV), Tacaribe (TCRV), and Pichindé promotes the phosphorylation of mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinases 1 and 2 (ERK1/2) and that this activation is required for the achievement of a productive infection. Here we examined the contribution of ERK1/2 in early steps of JUNV and TCRV multiplication. JUNV adsorption, internalization, and uncoating were not affected by treatment of cultured cells with U0126, an inhibitor of the ERK1/2 signaling pathway. In contrast, U0126 caused a marked reduction in viral protein expression and RNA synthesis, while JUNV RNA synthesis was significantly augmented in the presence of an activator of the ERK1/2 pathway. Moreover, U0126 impaired the expression of a reporter gene in a TCRV-based replicon system, confirming the ability of the compound to hinder arenavirus macromolecular synthesis. By using a cell-based assay, we determined that the inhibitor did not affect the translation of a synthetic TCRV-like mRNA. No changes in the phosphorylation pattern of the translation factor eIF2α were found in U0126-treated cells. Our results indicate that U0126 impairs viral RNA synthesis, thereby leading to a subsequent reduction in viral protein expression. Thus, we conclude that ERK1/2 signaling activation is required for an efficient arenavirus RNA synthesis.


Subject(s)
Arenaviruses, New World/physiology , Host-Pathogen Interactions , MAP Kinase Signaling System , Virus Replication , Animals , Butadienes/metabolism , Cell Line , Enzyme Inhibitors/metabolism , Nitriles/metabolism , RNA, Viral/biosynthesis , Viral Proteins/biosynthesis
6.
J Virol ; 91(20)2017 10 15.
Article in English | MEDLINE | ID: mdl-28794024

ABSTRACT

The arenavirus family consists of several highly pathogenic viruses, including the Old World (OW) arenavirus Lassa fever virus (LASV) and the New World (NW) Junin virus (JUNV) and Machupo virus (MACV). Host response to infection by these pathogenic arenaviruses is distinct in many aspects. JUNV and MACV infections readily induce an interferon (IFN) response in human cells, while LASV infection usually triggers an undetectable or weak IFN response. JUNV induces an IFN response through RIG-I, suggesting that the host non-self RNA sensor readily detects JUNV viral RNAs (vRNAs) during infection and activates IFN response. Double-stranded-RNA (dsRNA)-activated protein kinase R (PKR) is another host non-self RNA sensor classically known for its vRNA recognition activity. Here we report that infection with NW arenaviruses JUNV and MACV, but not OW LASV, activated PKR, concomitant with elevated phosphorylation of the translation initiation factor α subunit of eukaryotic initiation factor 2 (eIF2α). Host protein synthesis was substantially suppressed in MACV- and JUNV-infected cells but was only marginally reduced in LASV-infected cells. Despite the antiviral activity known for PKR against many other viruses, the replication of JUNV and MACV was not impaired but was slightly augmented in wild-type (wt) cells compared to that in PKR-deficient cells, suggesting that PKR or PKR activation did not negatively affect JUNV and MACV infection. Additionally, we found an enhanced IFN response in JUNV- or MACV-infected PKR-deficient cells, which was inversely correlated with virus replication.IMPORTANCE The detection of viral RNA by host non-self RNA sensors, including RIG-I and MDA5, is critical to the initiation of the innate immune response to RNA virus infection. Among pathogenic arenaviruses, the OW LASV usually does not elicit an interferon response. However, the NW arenaviruses JUNV and MACV readily trigger an IFN response in a RIG-I-dependent manner. Here, we demonstrate for the first time that pathogenic NW arenaviruses JUNV and MACV, but not the OW arenavirus LASV, activated the dsRNA-dependent PKR, another host non-self RNA sensor, during infection. Interestingly, the replication of JUNV and MACV was not restricted but was rather slightly augmented in the presence of PKR. Our data provide new evidence for a distinct interplay between host non-self RNA sensors and pathogenic arenaviruses, which also provides insights into the pathogenesis of arenaviruses and may facilitate the design of vaccines and treatments against arenavirus-caused diseases.


Subject(s)
Arenaviruses, New World/pathogenicity , Arenaviruses, Old World/pathogenicity , Immunity, Innate , Junin virus/pathogenicity , Receptors, Pattern Recognition/metabolism , Virus Replication , eIF-2 Kinase/metabolism , A549 Cells , Arenaviruses, New World/physiology , Arenaviruses, Old World/physiology , Host-Pathogen Interactions , Humans , Interferons/biosynthesis , Interferons/immunology , Junin virus/physiology , Phosphorylation , Receptors, Pattern Recognition/genetics , Transcription Factors/metabolism , eIF-2 Kinase/genetics
7.
Arch Virol ; 161(2): 353-63, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26559962

ABSTRACT

Tacaribe virus (TCRV) entry occurs by receptor-mediated endocytosis. To explore the entry mechanism used by TCRV, the inhibitory effects of drugs and dominant negative (DN) constructions affecting the main endocytic pathways were analyzed. In cells lacking the human transferrin receptor (hTfR), compounds and DN proteins that impair clathrin-mediated endocytosis were shown to reduce virus internalization without affecting virion binding. In contrast, in cells expressing the hTfR, compounds that affect clathrin-mediated endocytosis did not affect TCRV infection. Destabilization of cholesterol-rich plasma membrane microdomains by treatment with nystatin was not able to block virus entry in the presence of hTfR. However methyl-ß-cyclodextrin, which extracts cholesterol from cell membranes, reduced virus internalization in cells expressing the hTfR. Inhibition of dynamin and neutralization of the pH of intracellular vesicles reduced virus internalization in all cell lines tested. Taken together, these results demonstrate that in cells expressing the hTfR, TCRV internalization depends on the presence of cholesterol, dynamin and acidic intracellular vesicles, while in the rest of the cell lines analyzed, clathrin-mediated endocytosis is the main TCRV entry pathway and, as expected, depends on dynamin and acidic intracellular vesicles. These results represent an important contribution to the characterization of the arenavirus replication cycle.


Subject(s)
Antigens, CD/metabolism , Arenaviruses, New World/physiology , Host-Pathogen Interactions , Receptors, Transferrin/metabolism , Virus Internalization , Animals , CHO Cells , Cholesterol/metabolism , Cricetulus , Dynamins/metabolism , Humans , Hydrogen-Ion Concentration
8.
J Virol ; 89(22): 11643-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26355089

ABSTRACT

UNLABELLED: Animal viruses frequently cause zoonotic disease in humans. As these viruses are highly diverse, evaluating the threat that they pose remains a major challenge, and efficient approaches are needed to rapidly predict virus-host compatibility. Here, we develop a combined computational and experimental approach to assess the compatibility of New World arenaviruses, endemic in rodents, with the host TfR1 entry receptors of different potential new host species. Using signatures of positive selection, we identify a small motif on rodent TfR1 that conveys species specificity to the entry of viruses into cells. However, we show that mutations in this region affect the entry of each arenavirus differently. For example, a human single nucleotide polymorphism (SNP) in this region, L212V, makes human TfR1 a weaker receptor for one arenavirus, Machupo virus, but a stronger receptor for two other arenaviruses, Junin and Sabia viruses. Collectively, these findings set the stage for potential evolutionary trade-offs, where natural selection for resistance to one virus may make humans or rodents susceptible to other arenavirus species. Given the complexity of this host-virus interplay, we propose a computational method to predict these interactions, based on homology modeling and computational docking of the virus-receptor protein-protein interaction. We demonstrate the utility of this model for Machupo virus, for which a suitable cocrystal structural template exists. Our model effectively predicts whether the TfR1 receptors of different species will be functional receptors for Machupo virus entry. Approaches such at this could provide a first step toward computationally predicting the "host jumping" potential of a virus into a new host species. IMPORTANCE: We demonstrate how evolutionary trade-offs may exist in the dynamic evolutionary interplay between viruses and their hosts, where natural selection for resistance to one virus could make humans or rodents susceptible to other virus species. We present an algorithm that predicts which species have cell surface receptors that make them susceptible to Machupo virus, based on computational docking of protein structures. Few molecular models exist for predicting the risk of spillover of a particular animal virus into humans or new animal populations. Our results suggest that a combination of evolutionary analysis, structural modeling, and experimental verification may provide an efficient approach for screening and assessing the potential spillover risks of viruses circulating in animal populations.


Subject(s)
Antigens, CD/genetics , Arenaviruses, New World/physiology , Host Specificity , Receptors, Transferrin/genetics , Receptors, Virus/metabolism , Virus Attachment , Algorithms , Animals , Cell Line, Tumor , Computational Biology/methods , Disease Resistance/genetics , Dogs , HEK293 Cells , Humans , Molecular Docking Simulation , Receptors, Transferrin/metabolism , Receptors, Virus/ultrastructure , Virus Internalization
9.
J Virol ; 89(14): 7079-88, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25926656

ABSTRACT

UNLABELLED: The arenavirus family includes several important pathogens that cause severe and sometimes fatal diseases in humans. The highly pathogenic Old World (OW) arenavirus Lassa fever virus (LASV) is the causative agent of Lassa fever (LF) disease in humans. LASV infections in severe cases are generally immunosuppressive without stimulating interferon (IFN) induction, a proinflammatory response, or T cell activation. However, the host innate immune responses to highly pathogenic New World (NW) arenaviruses are not well understood. We have previously shown that the highly pathogenic NW arenavirus, Junin virus (JUNV), induced an IFN response in human A549 cells. Here, we report that Machupo virus (MACV), another highly pathogenic NW arenavirus, also induces an IFN response. Importantly, both pathogenic NW arenaviruses, in contrast to the OW highly pathogenic arenavirus LASV, readily elicited an IFN response in human primary dendritic cells and A549 cells. Coinfection experiments revealed that LASV could potently inhibit MACV-activated IFN responses even at 6 h after MACV infection, while the replication levels of MACV and LASV were not affected by virus coinfection. Our results clearly demonstrated that although all viruses studied herein are highly pathogenic to humans, the host IFN responses toward infections with the NW arenaviruses JUNV and MACV are quite different from responses to infections with the OW arenavirus LASV, a discovery that needs to be further investigated in relevant animal models. This finding might help us better understand various interplays between the host immune system and highly pathogenic arenaviruses as well as distinct mechanisms underlying viral pathogenesis. IMPORTANCE: Infections of humans with the highly pathogenic OW LASV are accompanied by potent suppression of interferon or proinflammatory cytokine production. In contrast, infections with the highly pathogenic NW arenavirus JUNV are associated with high levels of IFNs and cytokines in severe and fatal cases. Arenaviruses initially target macrophages and dendritic cells, which are potent IFN/cytokine-producers. In human macrophages, JUNV reportedly does not trigger IFN responses. We here demonstrated that JUNV activated IFN responses in human dendritic cells. MACV, another highly pathogenic NW arenavirus, also activated IFN responses. LASV did not induce detectable IFN responses, in spite of higher replication levels, and blocked the MACV-triggered IFN response in a coinfection assay. Although these viruses are highly pathogenic to humans, our study highlights distinct innate immune responses to infections with the NW arenaviruses JUNV and MACV and to infection with the OW arenavirus LASV and provides important insights into the virus-host interaction and pathogenesis.


Subject(s)
Arenaviruses, New World/immunology , Dendritic Cells/immunology , Epithelial Cells/immunology , Interferons/biosynthesis , Junin virus/immunology , Arenaviruses, New World/physiology , Cells, Cultured , Dendritic Cells/virology , Epithelial Cells/virology , Humans , Junin virus/physiology , Virus Replication
10.
Vet Pathol ; 52(1): 18-20, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25352204

ABSTRACT

The reports for Ebola virus Zaire (EBOV), Nipah virus, and Machupo virus (MACV) pathogenesis, in this issue of Veterinary Pathology, are timely considering recent events, both nationally and internationally. EBOV, Nipah virus, and MACV cause highly lethal infections for which no Food and Drug Administration (FDA) licensed vaccines or therapies exist. Not only are there concerns that these agents could be used by those with malicious intent, but shifts in ecological distribution of viral reservoirs due to climate change or globalization could lead to more frequent infections within remote regions than previously seen as well as outbreaks in more populous areas. The current EBOV epidemic shows no sign of abating across 3 West African nations (as of October 2014), including densely populated areas, far outpacing infection rates of previous outbreaks. A limited number of cases have also arisen in the United States and Europe. With few treatment options for these deadly viruses, development of animal models reflective of human disease is paramount to combat these diseases. As an example of this potential, a new treatment compound, ZMapp, that had demonstrated efficacy against EBOV infection in nonhuman primates (NHPs) received an emergency compassionate use exception from the FDA for the treatment of 2 American medical workers infected with EBOV, and they are currently virus free and recovering.


Subject(s)
Arenaviruses, New World/physiology , Disease Models, Animal , Ebolavirus/physiology , Hemorrhagic Fever, American/epidemiology , Hemorrhagic Fever, Ebola/epidemiology , Henipavirus Infections/epidemiology , Nipah Virus/physiology , Animals , Arenaviruses, New World/drug effects , Climate Change , Compassionate Use Trials , Disease Outbreaks , Ebolavirus/drug effects , Epidemics , Europe/epidemiology , Hemorrhagic Fever, American/drug therapy , Hemorrhagic Fever, American/virology , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Henipavirus Infections/drug therapy , Henipavirus Infections/virology , Humans , Internationality , Nipah Virus/drug effects , United States/epidemiology , United States Food and Drug Administration
11.
Vet Pathol ; 52(1): 26-37, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24990481

ABSTRACT

Machupo virus, the causative agent of Bolivian hemorrhagic fever (BHF), is a highly lethal viral hemorrhagic fever of which little is known and for which no Food and Drug Administration-approved vaccines or therapeutics are available. This study evaluated the cynomolgus macaque as an animal model using the Machupo virus, Chicava strain, via intramuscular and aerosol challenge. The incubation period was 6 to 10 days with initial signs of depression, anorexia, diarrhea, mild fever, and a petechial skin rash. These were often followed by neurologic signs and death within an average of 18 days. Complete blood counts revealed leukopenia as well as marked thrombocytopenia. Serum chemistry values identified a decrease in total protein, marked increases in alanine aminotransferase and aspartate aminotransferase, and moderate increases in alkaline phosphatase. Gross pathology findings included a macular rash extending across the axillary and inguinal regions beginning at approximately 10 days postexposure as well as enlarged lymph nodes and spleen, enlarged and friable liver, and sporadic hemorrhages along the gastrointestinal mucosa and serosa. Histologic lesions consisted of foci of degeneration and necrosis/apoptosis in the haired skin, liver, pancreas, adrenal glands, lymph nodes, tongue, esophagus, salivary glands, stomach, small intestine, and large intestine. Lymphohistiocytic interstitial pneumonia was also present. Inflammation within the central nervous system (nonsuppurative encephalitis) was histologically apparent approximately 16 days postexposure and was generally progressive. This study provides insight into the course of Machupo virus infection in cynomolgus macaques and supports the usefulness of cynomolgus macaques as a viable model of human Machupo virus infection.


Subject(s)
Arenaviruses, New World/physiology , Hemorrhagic Fever, American/pathology , Adrenal Glands/pathology , Aerosols/administration & dosage , Animals , Disease Models, Animal , Female , Hemorrhagic Fever, American/virology , Humans , Injections, Intramuscular , Liver/pathology , Lung/pathology , Lymph Nodes/pathology , Macaca fascicularis , Male , Spleen/pathology
12.
Curr Opin Virol ; 5: 82-90, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24636947

ABSTRACT

The etiologic agent of Bolivian hemorrhagic fever (BHF), Machupo virus (MACV) is reported to have a mortality rate of 25-35%. First identified in 1959, BHF was the cause of a localized outbreak in San Joaquin until rodent population controls were implemented in 1964. The rodent Calomys collosus was identified as the primary vector and reservoir for the virus. Multiple animal models were considered during the 1970s with the most human-like disease identified in Rhesus macaques but minimal characterization of the pathogenesis has been published since. A reemergence of reported BHF cases has been reported in recent years, which necessitates the further study and development of a vaccine to prevent future outbreaks.


Subject(s)
Arenaviruses, New World/pathogenicity , Hemorrhagic Fever, American/virology , Animals , Arenaviruses, New World/genetics , Arenaviruses, New World/physiology , Disease Models, Animal , Hemorrhagic Fever, American/epidemiology , Humans , Macaca mulatta , Virulence
13.
J Mol Biol ; 425(24): 4893-903, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24075870

ABSTRACT

Arenaviruses are enveloped, negative-stranded RNA viruses that belong to the family Arenaviridae. This diverse family can be further classified into OW (Old World) and NW (New World) arenaviruses based on their antigenicity, phylogeny, and geographical distribution. Many of the NW arenaviruses are highly pathogenic viruses that cause systemic human infections characterized by hemorrhagic fever and/or neurological manifestations, constituting public health problems in their endemic regions. NW arenavirus infection induces a variety of host innate immune responses, which could contribute to the viral pathogenesis and/or influence the final outcome of virus infection in vitro and in vivo. On the other hand, NW arenaviruses have also developed several strategies to counteract the host innate immune response. We will review current knowledge regarding the interplay between the host innate immune response and NW arenavirus infection in vitro and in vivo, with emphasis on viral-encoded proteins and their effect on the type I interferon response.


Subject(s)
Arenaviridae Infections/immunology , Arenaviruses, New World/immunology , Immunity, Innate/immunology , Interferon Type I/metabolism , Viral Proteins/metabolism , Animals , Arenaviridae Infections/virology , Arenaviruses, New World/genetics , Arenaviruses, New World/physiology , Arenaviruses, Old World/immunology , Host-Pathogen Interactions/immunology , Humans , Immune Evasion , Interferon Type I/genetics , Mice , Models, Molecular , Viral Proteins/genetics
14.
J Virol ; 87(24): 13930-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24109228

ABSTRACT

Ocozocoautla de Espinosa virus (OCEV) is a novel, uncultured arenavirus. We found that the OCEV glycoprotein mediates entry into grivet and bat cells through transferrin receptor 1 (TfR1) binding but that OCEV glycoprotein precursor (GPC)-pseudotyped retroviruses poorly entered 53 human cancer cell lines. Interestingly, OCEV and Tacaribe virus could use bat, but not human, TfR1. Replacing three human TfR1 amino acids with their bat ortholog counterparts transformed human TfR1 into an efficient OCEV and Tacaribe virus receptor.


Subject(s)
Arenaviridae Infections/metabolism , Arenaviridae Infections/veterinary , Arenaviruses, New World/physiology , Chiroptera/metabolism , Chlorocebus aethiops/metabolism , Receptors, Transferrin/metabolism , Receptors, Virus/metabolism , Virus Internalization , Amino Acid Sequence , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Arenaviridae Infections/genetics , Arenaviridae Infections/virology , Arenaviruses, New World/genetics , Cell Line , Chiroptera/genetics , Chiroptera/virology , Chlorocebus aethiops/genetics , Chlorocebus aethiops/virology , Humans , Molecular Sequence Data , Receptors, Transferrin/genetics , Receptors, Virus/genetics , Sequence Alignment , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
15.
Article in Russian | MEDLINE | ID: mdl-24000605

ABSTRACT

Analysis of data of the available literature on epidemiology of Bolivian hemorrhagic fever, manifestations of human disease, biological properties of the causative agent and development carried out abroad of means and methods of diagnostics, prophylaxis and therapy of this infection that presents a potential threat for the population and economy of the Russian Federation in case of introduction of the causative agent is presented.


Subject(s)
Arenaviruses, New World/physiology , Hemorrhagic Fever, American/epidemiology , Hemorrhagic Fever, American/physiopathology , Muridae/virology , Virion/physiology , Animals , Antibodies, Neutralizing/blood , Antiviral Agents/therapeutic use , Arenaviruses, New World/pathogenicity , Disease Management , Disease Outbreaks , Disease Vectors , Hemorrhagic Fever, American/therapy , Hemorrhagic Fever, American/virology , Humans , Russia/epidemiology , South America/epidemiology , Virion/pathogenicity , Virus Replication
16.
Bioorg Med Chem Lett ; 23(14): 4258-62, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23747225

ABSTRACT

Novel N-alkyldeoxynojirimycins (NADNJs) based on our previous lead 3 were designed, synthesized and tested in metabolic assays and in virus cultures. NADNJs containing terminal tertiary benzamide, sulfonamide, urea, and oxazolidinone moieties were discovered to have improved metabolic stability compared to 3, while maintaining submicromolar EC50 against BVDV and Tacaribe virus; and low cytotoxicity.


Subject(s)
Antiviral Agents/chemical synthesis , Arenaviruses, New World/physiology , Diarrhea Viruses, Bovine Viral/physiology , Drug Design , Glucosamine/analogs & derivatives , 1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/metabolism , 1-Deoxynojirimycin/pharmacology , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Glucosamine/chemistry , Glucosamine/metabolism , Glucosamine/pharmacology , Humans , Mice , Microsomes, Liver/metabolism , Oxazolidinones/chemistry , Rats , Sulfonamides/chemistry , Urea/chemistry , Virus Replication/drug effects
17.
J Biol Chem ; 288(12): 8702-8711, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23382389

ABSTRACT

The arenavirus nucleoprotein (NP) can suppress induction of type I interferon (IFN). This anti-IFN activity is thought to be shared by all arenaviruses with the exception of Tacaribe virus (TCRV). To identify the TCRV NP amino acid residues that prevent its IFN-countering ability, we created a series of NP chimeras between residues of TCRV NP and Pichinde virus (PICV) NP, an arenavirus NP with potent anti-IFN function. Chimera NP analysis revealed that a minimal four amino acid stretch derived from PICV NP could impart efficient anti-IFN activity to TCRV NP. Strikingly, the TCRV NP gene cloned and sequenced from viral stocks obtained through National Institutes of Health Biodefense and Emerging Infections (BEI) resources deviated from the reference sequence at this particular four-amino acid region, GPPT (GenBank KC329849) versus DLQL (GenBank NC004293), respectively at residues 389-392. When efficiently expressed in cells through codon-optimization, TCRV NP containing the GPPT residues rescued the antagonistic IFN function. Consistent with cell expression results, TCRV infection did not stimulate an IFNß response early in infection in multiple cells types (e.g. A549, P388D1), and IRF-3 was not translocated to the nucleus in TCRV-infected A549 cells. Collectively, these data suggest that certain TCRV strain variants contain the important NP amino acids necessary for anti-IFN activity.


Subject(s)
Arenaviruses, New World/physiology , Interferon-beta/metabolism , Nucleoproteins/chemistry , Recombinant Fusion Proteins/chemistry , Viral Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Arenaviruses, New World/immunology , Cell Nucleus/metabolism , Chlorocebus aethiops , HEK293 Cells , Host-Pathogen Interactions , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Mice , Molecular Sequence Data , Nucleoproteins/biosynthesis , Nucleoproteins/immunology , Promoter Regions, Genetic , Protein Transport , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/immunology , Transcriptional Activation , Vero Cells , Viral Proteins/biosynthesis , Viral Proteins/immunology
18.
Vector Borne Zoonotic Dis ; 13(1): 50-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23210637

ABSTRACT

A total of 3941 rodents were captured during a 46-month prospective (mark-recapture) study on the ecology of Catarina virus in southern Texas. Antibody reactive against Catarina virus was found in 73 (11.9%) of 611 southern plains woodrats (Neotoma micropus) and none of 3330 other rodents; strains of Catarina virus were isolated from 6 antibody-negative and 9 antibody-positive southern plains woodrats; and the infections in at least 3 southern plains woodrats were chronic. These results affirm the notion that the southern plains woodrat is the principal host of Catarina virus and suggest that Catarina virus infection is highly specific to N. micropus.


Subject(s)
Antibodies, Viral/blood , Arenaviridae Infections/veterinary , Arenaviruses, New World/physiology , Rodent Diseases/virology , Sigmodontinae/virology , Animals , Arenaviridae Infections/epidemiology , Arenaviridae Infections/virology , Arenaviruses, New World/genetics , Arenaviruses, New World/immunology , Arenaviruses, New World/isolation & purification , Ecology , Host Specificity , Immunoglobulin G/blood , Phylogeny , Prevalence , Prospective Studies , Rain , Rats , Rodent Diseases/epidemiology , Rodentia , Texas/epidemiology
19.
J Virol ; 86(20): 11301-10, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22875974

ABSTRACT

Arenaviruses are responsible for acute hemorrhagic fevers with high mortality and pose significant threats to public health and biodefense. These enveloped negative-sense RNA viruses replicate in the cell cytoplasm and express four proteins. To better understand how these proteins insinuate themselves into cellular processes to orchestrate productive viral replication, we have identified and characterized novel cytosolic structures involved in arenavirus replication and transcription. In cells infected with the nonpathogenic Tacaribe virus or the attenuated Candid#1 strain of Junín virus, we find that newly synthesized viral RNAs localize to cytosolic puncta containing the nucleoprotein (N) of the virus. Density gradient centrifugation studies reveal that these replication-transcription complexes (RTCs) are associated with cellular membranes and contain full-length genomic- and antigenomic-sense RNAs. Viral mRNAs segregate at a higher buoyant density and are likewise scant in immunopurified RTCs, consistent with their translation on bulk cellular ribosomes. In addition, confocal microscopy analysis reveals that RTCs contain the lipid phosphatidylinositol-4-phosphate and proteins involved in cellular mRNA metabolism, including the large and small ribosomal subunit proteins L10a and S6, the stress granule protein G3BP1, and a subset of translation initiation factors. Elucidating the structure and function of RTCs will enhance our understanding of virus-cell interactions that promote arenavirus replication and mitigate against host cell immunity. This knowledge may lead to novel intervention strategies to limit viral virulence and pathogenesis.


Subject(s)
Arenaviridae Infections/virology , Arenaviruses, New World/physiology , Cytosol/virology , Junin virus/physiology , RNA, Viral/biosynthesis , Virus Replication , Animals , Arenaviruses, New World/pathogenicity , Cell Membrane/virology , Centrifugation, Density Gradient , Chlorocebus aethiops , Cytosol/ultrastructure , Junin virus/pathogenicity , Nucleoproteins , Phosphatidylinositol Phosphates/analysis , RNA, Viral/genetics , Transcription, Genetic , Vero Cells
20.
J Virol ; 86(7): 4024-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22278244

ABSTRACT

Five New World (NW) arenaviruses cause human hemorrhagic fevers. Four of these arenaviruses are known to enter cells by binding human transferrin receptor 1 (hTfR1). Here we show that the fifth arenavirus, Chapare virus, similarly uses hTfR1. We also identify an anti-hTfR1 antibody, ch128.1, which efficiently inhibits entry mediated by the glycoproteins of all five viruses, as well as replication of infectious Junín virus. Our data indicate that all NW hemorrhagic fever arenaviruses utilize a common hTfR1 apical-domain epitope and suggest that therapeutic agents targeting this epitope, including ch128.1 itself, can be broadly effective in treating South American hemorrhagic fevers.


Subject(s)
Antibodies/immunology , Antigens, CD/chemistry , Antigens, CD/immunology , Arenaviruses, New World/physiology , Down-Regulation , Hemorrhagic Fevers, Viral/virology , Receptors, Transferrin/chemistry , Receptors, Transferrin/immunology , Virus Internalization , Amino Acid Sequence , Animals , Antigens, CD/genetics , Cell Line , Hemorrhagic Fevers, Viral/genetics , Hemorrhagic Fevers, Viral/immunology , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, Transferrin/genetics , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/immunology , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...