Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Virulence ; 11(1): 1131-1141, 2020 12.
Article in English | MEDLINE | ID: mdl-32799623

ABSTRACT

Arenaviruses, such as Lassa virus (LASV), can cause severe and fatal hemorrhagic fevers (e.g., Lassa fever, LF) in humans with no vaccines or therapeutics. Research on arenavirus-induced hemorrhagic fevers (AHFs) has been hampered by the highly virulent nature of these viral pathogens, which require high biocontainment laboratory, and the lack of an immune-competent small animal model that can recapitulate AHF disease and pathological features. Guinea pig infected with Pichinde virus (PICV), an arenavirus that does not cause disease in humans, has been established as a convenient surrogate animal model for AHFs as it can be handled in a conventional laboratory. The PICV strain P18, derived from sequential passaging of the virus 18 times in strain 13 inbred guinea pigs, causes severe febrile illness in guinea pigs that is reminiscent of lethal LF in humans. As inbred guinea pigs are not readily available and are difficult to maintain, outbred Hartley guinea pigs have been used but they show a high degree of disease heterogeneity upon virulent P18 PICV infection. Here, we describe an improved outbred guinea-pig infection model using recombinant rP18 PICV generated by reverse genetics technique followed by plaque purification, which consistently shows >90% mortality and virulent infection. Comprehensive virological, histopathological, and immunohistochemical analyses of the rP18-virus infected animals show similar features of human LASV infection. Our data demonstrate that this improved animal model can serve as a safe, affordable, and convenient surrogate small animal model for studying human LF pathogenesis and for evaluating efficacy of preventative or therapeutic approaches.


Subject(s)
Disease Models, Animal , Guinea Pigs , Lassa Fever/pathology , Lassa Fever/virology , Pichinde virus/genetics , Pichinde virus/pathogenicity , Animals , Animals, Outbred Strains , Arenaviridae Infections/virology , Cell Line , Chlorocebus aethiops , Cricetinae , Humans , Recombination, Genetic , Reverse Genetics , Vero Cells , Virulence
2.
J Virol ; 87(12): 6635-43, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23552411

ABSTRACT

Several arenaviruses are responsible for causing viral hemorrhagic fevers (VHF) in humans. Lassa virus (LASV), the causative agent of Lassa fever, is a biosafety level 4 (BSL4) pathogen that requires handling in BSL4 facilities. In contrast, the Pichinde arenavirus (PICV) is a BSL2 pathogen that can cause hemorrhagic fever-like symptoms in guinea pigs that resemble those observed in human Lassa fever. Comparative sequence analysis of the avirulent P2 strain of PICV and the virulent P18 strain shows a high degree of sequence homology in the bisegmented genome between the two strains despite the polarized clinical outcomes noted for the infected animals. Using reverse genetics systems that we have recently developed, we have mapped the sequence changes in the large (L) segment of the PICV genome that are responsible for the heightened virulence phenotype of the P18 strain. By monitoring the degree of disease severity and lethality caused by the different mutant viruses, we have identified specific residues located within the viral L polymerase gene encoded on the L segment essential for mediating disease pathogenesis. Through quantitative reverse transcription-PCR (RT-PCR) analysis, we have confirmed that the same set of residues is responsible for the increased viral replicative potential of the P18 strain and its heightened disease severity in vivo. Our laboratory findings serve to reinforce field observations that a high level of viremia often correlates with severe disease outcomes in LASV-infected patients.


Subject(s)
Arenaviridae Infections/pathology , DNA-Directed RNA Polymerases/genetics , Genomics , Pichinde virus/classification , Pichinde virus/pathogenicity , Animals , Arenaviridae Infections/mortality , Arenaviridae Infections/virology , Chlorocebus aethiops , DNA-Directed RNA Polymerases/chemistry , Guinea Pigs , Humans , Liver/pathology , Male , Pichinde virus/genetics , Point Mutation , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Virulence/genetics , Virus Replication
3.
Virology ; 433(1): 97-103, 2012 Nov 10.
Article in English | MEDLINE | ID: mdl-22877842

ABSTRACT

We use a small animal model, based on guinea pigs infected with a non-pathogenic Pichinde virus (PICV), to understand the virulence mechanisms of arenavirus infections in the hosts. PICV P2 strain causes a mild febrile reaction in guinea pigs, while P18 causes severe disease with clinical and pathological features reminiscent of Lassa hemorrhagic fever in humans. The envelope glycoproteins (GPC) of P2 and P18 viruses differ at positions 119, 140, and 164, all localized to the receptor-binding G1 subunit. We found that lentiviral pseudotyped virions (VLPs) bearing P18 GPC show more efficient cell entry than those with P2 GPC, and that the E140 residue plays a critical role in this process. Infection of guinea pigs with the recombinant viruses containing the E140K change demonstrated that E140 of GPC is a necessary virulence determinant of P18 infections, possibly by enhancing the ability of virus to enter target cells.


Subject(s)
Arenaviridae Infections/virology , Liver/virology , Pichinde virus/pathogenicity , Protein Subunits/genetics , Viral Envelope Proteins/genetics , Amino Acid Substitution , Animals , Arenaviridae Infections/pathology , Cell Line , Disease Models, Animal , Guinea Pigs , Humans , Lassa Fever/pathology , Lassa Fever/virology , Liver/pathology , Mutation , Pichinde virus/physiology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Viral Load , Virulence , Virus Internalization
4.
J Virol ; 86(18): 9794-801, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22761375

ABSTRACT

Arenaviruses can cause severe hemorrhagic fever diseases in humans, with limited prophylactic or therapeutic measures. A small RING-domain viral protein Z has been shown to mediate the formation of virus-like particles and to inhibit viral RNA synthesis, although its biological roles in an infectious viral life cycle have not been directly addressed. By taking advantage of the available reverse genetics system for a model arenavirus, Pichinde virus (PICV), we provide the direct evidence for the essential biological roles of the Z protein's conserved residues, including the G2 myristylation site, the conserved C and H residues of RING domain, and the poorly characterized C-terminal L79 and P80 residues. Dicodon substitutions within the late (L) domain (PSAPPYEP) of the PICV Z protein, although producing viable mutant viruses, have significantly reduced virus growth, a finding suggestive of an important role for the intact L domain in viral replication. Further structure-function analyses of both PICV and Lassa fever virus Z proteins suggest that arenavirus Z proteins have similar molecular mechanisms in mediating their multiple functions, with some interesting variations, such as the role of the G2 residue in blocking viral RNA synthesis. In summary, our studies have characterized the biological roles of the Z protein in an infectious arenavirus system and have shed important light on the distinct functions of its domains in virus budding and viral RNA regulation, the knowledge of which may lead to the development of novel antiviral drugs.


Subject(s)
Arenavirus/physiology , Viral Proteins/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Arenaviridae Infections/etiology , Arenaviridae Infections/virology , Arenavirus/genetics , Arenavirus/pathogenicity , Cell Line , Conserved Sequence , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Pichinde virus/genetics , Pichinde virus/pathogenicity , Pichinde virus/physiology , Protein Structure, Tertiary , RNA, Viral/biosynthesis , Sequence Homology, Amino Acid , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Release/genetics , Virus Release/physiology , Virus Replication/genetics , Virus Replication/physiology
5.
Virol J ; 7: 240, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20846417

ABSTRACT

BACKGROUND: A number of RNA viruses cause viral hemorrhagic fever (VHF), in which proinflammatory mediators released from infected cells induce increased permeability of the endothelial lining of blood vessels, leading to loss of plasma volume, hypotension, multi-organ failure, shock and death. The optimal treatment of VHF should therefore include both the use of antiviral drugs to inhibit viral replication and measures to prevent or correct changes in vascular function. Although rodent models have been used to evaluate treatments for increased vascular permeability (VP) in bacterial sepsis, such studies have not been performed for VHF. RESULTS: Here, we use an established model of Pichinde virus infection of hamsters to demonstrate how changes in VP can be detected by intravenous infusion of Evans blue dye (EBD), and compare those measurements to changes in hematocrit, serum albumin concentration and serum levels of proinflammatory mediators. We show that EBD injected into sick animals in the late stage of infection is rapidly sequestered in the viscera, while in healthy animals it remains within the plasma, causing the skin to turn a marked blue color. This test could be used in live animals to detect increased VP and to assess the ability of antiviral drugs and vasoactive compounds to prevent its onset. Finally, we describe a multiplexed assay to measure levels of serum factors during the course of Pichinde arenavirus infection and demonstrate that viremia and subsequent increase in white blood cell counts precede the elaboration of inflammatory mediators, which is followed by increased VP and death. CONCLUSIONS: This level of model characterization is essential to the evaluation of novel interventions designed to control the effects of virus-induced hypercytokinemia on host vascular function in VHF, which could lead to improved survival.


Subject(s)
Capillary Permeability/physiology , Evans Blue/pharmacokinetics , Hemorrhagic Fevers, Viral/pathology , Hemorrhagic Fevers, Viral/physiopathology , Pichinde virus/pathogenicity , Animals , Cricetinae , Cytokines/blood , Disease Models, Animal , Female , Hematocrit , Mesocricetus , Serum Albumin/analysis
6.
Viral Immunol ; 22(6): 457-62, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19951183

ABSTRACT

Lassa virus pathogenesis is believed to involve dysregulation of cytokines. We have previously shown nuclear factor-kappaB (NF-kappaB) inhibition using a BSL-2 model for Lassa fever. Here we further define the potential mechanism for NF-kappaB inhibition as involving increased levels of repressive p50/p50 homodimers, and suggest a novel therapeutic strategy that acts via modulation of host signaling.


Subject(s)
Arenaviridae Infections/drug therapy , Defective Viruses/pathogenicity , Monocytes/virology , NF-kappa B/metabolism , Pichinde virus/pathogenicity , Animals , CSK Tyrosine-Protein Kinase , Defective Viruses/physiology , Drug Delivery Systems , In Vitro Techniques , Mice , NF-kappa B p50 Subunit/metabolism , Phosphorylation , Pichinde virus/physiology , Protein Processing, Post-Translational , Protein Transport , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , Transcription, Genetic , Virulence , src-Family Kinases
7.
Virol J ; 6: 162, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19814828

ABSTRACT

This report is the first to demonstrate infection of human endothelial cells by Pichinde virus (PIC). PIC infection induces an upregulation of the inducible nitric oxide synthase gene; as well as an increase in detectable nitric oxide (NO). PIC induces an increase in permeability in endothelial cell monolayers which can be abrogated at all measured timepoints with the addition of a nitric oxide synthase inhibitor, indicating a role for NO in the alteration of endothelial barrier function. Because NO has shown antiviral activity against some viruses, viral titer was measured after addition of the NO synthase inhibitor and found to have no effect in altering virus load in infected EC. The NO synthase inhibition also has no effect on levels of activated caspases induced by PIC infection. Taken together, these data indicate NO production induced by Pichinde virus infection has a pathogenic effect on endothelial cell monolayer permeability.


Subject(s)
Endothelial Cells/virology , Nitric Oxide/toxicity , Permeability/drug effects , Pichinde virus/pathogenicity , Cell Line , Humans , Nitric Oxide/metabolism , Pichinde virus/immunology
8.
Ann N Y Acad Sci ; 1171 Suppl 1: E65-74, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19751405

ABSTRACT

Arenaviruses are enveloped single-strand RNA viruses that mostly have natural hosts in rodents. Upon infection of humans, several arenaviruses can cause severe hemorrhagic fever diseases, including Lassa fever that is endemic in West Africa. The virulence mechanism of these deadly arenaviruses can be studied in a safe and economical small animal model-guinea pigs infected by a nonpathogenic arenavirus Pichinde virus (PICV), a virulent strain of which can cause similar disease syndromes in guinea pigs as arenaviral hemorrhagic fevers in humans. We have recently developed molecular clones for both the virulent and avirulent strains of PICV. Using the available reverse genetics tools, we are characterizing the molecular determinants of virulent arenavirus infections in vivo.


Subject(s)
Arenaviridae Infections/genetics , Pichinde virus/genetics , Animals , Arenaviridae/genetics , Arenaviridae Infections/physiopathology , Body Temperature , Conserved Sequence , DNA, Viral/genetics , Disease Models, Animal , Genome, Viral , Guinea Pigs , Humans , Pichinde virus/pathogenicity , Plasmids/genetics , RNA, Viral/genetics , Recombination, Genetic , Transcription, Genetic , Viremia/genetics , Viremia/physiopathology
9.
J Virol ; 83(13): 6357-62, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19386714

ABSTRACT

Several arenaviruses can cause hemorrhagic fever diseases (VHFs) in humans, the pathogenic mechanism of which is poorly understood due to their virulent nature and the lack of molecular clones. A safe, convenient, and economical small animal model of arenavirus hemorrhagic fever is based on guinea pigs infected by the arenavirus Pichinde (PICV). PICV does not cause disease in humans, but an adapted strain of PICV (P18) causes a disease in guinea pigs that mimics arenavirus hemorrhagic fever in humans in many aspects, while a low-passaged strain (P2) remains avirulent in infected animals. In order to identify the virulence determinants within the PICV genome, we developed the molecular clones for both the avirulent P2 and virulent P18 viruses. Recombinant viruses were generated by transfecting plasmids that contain the antigenomic L and S RNA segments of PICV under the control of the T7 promoter into BSRT7-5 cells, which constitutively express T7 RNA polymerase. By analyzing viral growth kinetics in vitro and virulence in vivo, we show that the recombinant viruses accurately recapitulate the replication and virulence natures of their respective parental viruses. Both parental and recombinant virulent viruses led to high levels of viremia and titers in different organs of the infected animals, whereas the avirulent viruses were effectively controlled and cleared by the hosts. These novel infectious clones for the PICV provide essential tools to identify the virulence factors that are responsible for the severe VHF-like disease in infected animals.


Subject(s)
Hemorrhagic Fever, American/virology , Pichinde virus/pathogenicity , Virulence/genetics , Animals , Chlorocebus aethiops , DNA, Complementary , Disease Models, Animal , Genome, Viral , Guinea Pigs , Macrophages, Peritoneal/virology , Male , Pichinde virus/genetics , Pichinde virus/growth & development , Vero Cells
10.
J Gen Virol ; 89(Pt 10): 2569-2579, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18796726

ABSTRACT

Arenaviruses such as Lassa virus cause a spectrum of disease in humans ranging from mild febrile illness to lethal haemorrhagic fever. The contributions of innate immunity to protection or pathogenicity are unknown. We compared patterns of expression of cytokines of innate immunity in mild versus severe arenavirus disease using an established guinea pig model based on the macrophage-tropic arenavirus Pichinde virus (PICV). Cytokine transcripts were measured by using real-time RT-PCR in target organs and blood during mild infection (caused by PICV, P2 variant) and lethal haemorrhagic fever (caused by PICV, P18 variant). In the initial peritoneal target cells, virulent P18 infection was associated with significantly increased gamma interferon (IFN-gamma) and monocyte chemoattractant protein-1 (MCP-1, CCL2) mRNA levels relative to P2 infection. Peritoneal cells from P18-infected animals had decreased tumour necrosis factor alpha (TNF-alpha), interleukin (IL)-8 (CXCL-8) and IL-12p40 transcripts relative to mock-infected animals. Late in infection, P18-infected peripheral blood leukocytes (PBL) had decreased TNF-alpha, IFN-gamma, and regulated upon activation, normal T cell expressed and secreted (RANTES, CCL-5) cytokine transcripts relative to P2-infected PBL. We conclude that, in severe arenavirus disease, patterns of cytokine expression in the initially infected cells favour recruitment of additional target monocytes, while inhibiting some of their pro-inflammatory responses. Suppression rather than overexpression of pro-inflammatory cytokines accompanied the terminal shock in this model of arenavirus haemorrhagic fever.


Subject(s)
Arenaviridae Infections/immunology , Arenaviridae Infections/physiopathology , Cytokines/metabolism , Hemorrhagic Fevers, Viral/immunology , Pichinde virus/pathogenicity , Animals , Arenaviridae Infections/virology , Cytokines/genetics , Disease Models, Animal , Guinea Pigs , Hemorrhagic Fevers, Viral/virology , Humans , Immunity, Innate/immunology , Inflammation/immunology , Pichinde virus/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Severity of Illness Index , Virulence
11.
J Virol ; 80(20): 10248-52, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17005702

ABSTRACT

The family Arenaviridae includes several National Institutes of Allergy and Infections Diseases category A select agents which cause hemorrhagic fever. There are few vaccines available, and treatment is limited to ribavirin, which varies in efficacy. Development of new antiviral compounds has been hindered by a lack of understanding of the molecular basis of pathogenesis. We used two variants of Pichinde virus, one attenuated and one virulent in the guinea pig model, to delineate the host determinants which lead to either viral clearance or lethal disease. By analyzing protein level changes using pathway analysis, we have identified key intermediates which may be targets for therapeutic intervention.


Subject(s)
Arenaviridae Infections/metabolism , Gene Expression Regulation , Pichinde virus/pathogenicity , Proteome/analysis , Animals , Disease Models, Animal , Guinea Pigs , Immunoblotting , Macrophages/chemistry , Pichinde virus/genetics
12.
Virology ; 335(1): 87-98, 2005 Apr 25.
Article in English | MEDLINE | ID: mdl-15823608

ABSTRACT

Several viruses in the Arenavirus genus of the family Arenaviridae cause severe, often fatal, hemorrhagic fever. One such virus, Lassa virus (LV), is a frequent cause of disease in Africa, and survivors often are left with substantial neurological impairment. The feasibility of protective immunization against LV infection, and the associated disease, has been demonstrated in animal models, using recombinant vaccinia viruses to deliver Lassa proteins. Circumstantial evidence implicates cellular immunity in this Lassa-induced protection, but this has not been confirmed. Here, we describe DNA vaccines that encode LV proteins. A single inoculation of a plasmid encoding full-length Lassa nucleoprotein (LNP) can induce CD8(+) T cell responses in mice and can protect against challenge with two arenaviruses, lymphocytic choriomeningitis virus (LCMV) and Pichinde virus (PV). A DNA minigene vaccine encoding a 9 amino acid sequence from LNP also induces CD8(+) T cells and protects against arenavirus challenge, thus confirming prior speculation that protective cellular immunity is induced by LV proteins.


Subject(s)
Arenaviridae Infections/prevention & control , CD8-Positive T-Lymphocytes/immunology , Lassa virus/immunology , Lymphocytic Choriomeningitis/prevention & control , Nucleoproteins/genetics , Nucleoproteins/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Amino Acid Sequence , Animals , Arenaviridae Infections/virology , HeLa Cells , Humans , Immunization , Lassa virus/genetics , Lassa virus/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Nucleoproteins/metabolism , Pichinde virus/pathogenicity , Vaccines, DNA/administration & dosage , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism , Viral Vaccines/administration & dosage
13.
Virology ; 290(1): 30-8, 2001 Nov 10.
Article in English | MEDLINE | ID: mdl-11883003

ABSTRACT

The new world arenavirus Pichinde (PIC) is the basis of an accepted small animal model for human Lassa fever. PIC (Munchique strain) variant P2 is attenuated in guinea pigs, whereas variant P18 is extremely virulent. Previous sequence analysis of the S segments of these two viruses indicated a small number of possible virulence markers in the glycoprotein precursor (GPC) and nucleoprotein (NP) genes. In order to determine the role of these S segment genes in guinea pig virulence in this system, we have generated reassortant viruses. When tested in outbred guinea pigs, the reassortant containing the S segment from the virulent parent P18 (S18L2) caused significantly higher morbidity than the reciprocal reassortant. This increased morbidity was associated with higher viral titers in serum and spleen. However, the S18L2 reassortant was not as fully virulent in this system as the P18 parent, indicating a role for L segment genes in virulence.


Subject(s)
Arenaviridae Infections/virology , Pichinde virus/genetics , Reassortant Viruses/genetics , Animals , Arenaviridae Infections/physiopathology , Base Sequence , Chlorocebus aethiops , DNA, Viral , Disease Models, Animal , Genetic Variation , Guinea Pigs , Male , Molecular Sequence Data , Pichinde virus/pathogenicity , Reassortant Viruses/pathogenicity , Recombination, Genetic , Sequence Homology, Nucleic Acid , Vero Cells
14.
Am J Trop Med Hyg ; 61(2): 220-5, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10463670

ABSTRACT

The established animal model for Lassa fever is based on the new world arenavirus Pichinde (PIC). Natural isolates of PIC virus are attenuated in guinea pigs, but serial guinea pig passage renders them extremely virulent in that host. We have compared the nucleotide sequences of the small RNA segments of two attenuated, low-passage variants of the PIC virus Munchique strain (CoAn 4763) and two virulent, high-passage derivatives. Missense mutations in the glycoprotein precursor (GPC) gene at codons GPC-119, GPC-140, and GPC-164 and the nucleoprotein gene (NP) codons NP-35 and NP-374 were most closely associated with virulence. Codon GPC-140 is predicted to represent a region of peak hydrophilicity of the glycoprotein 1 (GP1); it is conceivable that mutations at this site could influence virulence by altering B cell epitopes or virus attachment protein conformation.


Subject(s)
Hemorrhagic Fever, American/virology , Pichinde virus/genetics , RNA/genetics , Animals , Base Sequence , Codon , Genetic Variation , Guinea Pigs , Lethal Dose 50 , Male , Models, Genetic , Mutation, Missense , Phenotype , Pichinde virus/classification , Pichinde virus/isolation & purification , Pichinde virus/pathogenicity , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA
15.
J Exp Med ; 188(9): 1705-15, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9802982

ABSTRACT

A basic principle of immunology is that prior immunity results in complete protection against a homologous agent. In this study, we show that memory T cells specific to unrelated viruses may alter the host's primary immune response to a second virus. Studies with a panel of heterologous viruses, including lymphocytic choriomeningitis (LCMV), Pichinde (PV), vaccinia (VV), and murine cytomegalo (MCMV) viruses showed that prior immunity with one of these viruses in many cases enhanced clearance of a second unrelated virus early in infection. Such protective immunity was common, but it depended on the virus sequence and was not necessarily reciprocal. Cell transfer studies showed that both CD4 and CD8 T cell populations from LCMV-immune mice were required to transfer protective immunity to naive hosts challenged with PV or VV. In the case of LCMV-immune versus naive mice challenged with VV, there was an enhanced early recruitment of memory phenotype interferon (IFN) gamma-secreting CD4(+) and CD8(+) cells into the peritoneal cavity and increased IFN-gamma levels in this initial site of virus replication. Studies with IFN-gamma receptor knockout mice confirmed a role for IFN-gamma in mediating the protective effect by LCMV-immune T cell populations when mice were challenged with VV but not PV. In some virus sequences memory cell populations, although clearing the challenge virus more rapidly, elicited enhanced IFN-gamma-dependent immunopathogenesis in the form of acute fatty necrosis. These results indicate that how a host responds to an infectious agent is a function of its history of previous infections and their influence on the memory T cell pool.


Subject(s)
Immunologic Memory , T-Lymphocytes/immunology , Viruses/immunology , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Female , Immunization , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/immunology , Muromegalovirus/pathogenicity , Necrosis , Phenotype , Pichinde virus/immunology , Pichinde virus/pathogenicity , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/immunology , Vaccinia virus/pathogenicity , Virus Diseases/immunology , Virus Diseases/pathology , Virus Diseases/prevention & control , Viruses/pathogenicity
16.
Am J Trop Med Hyg ; 52(3): 262-9, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7694969

ABSTRACT

Pichinde virus (PIC) is a reticuloendothelial arenavirus of the New World tropics. A guinea pig passage-adapted strain of this virus (adPIC) is uniformly lethal for inbred guinea pigs, while the related, prototype strain (PIC3739) has attenuated virulence. The abilities of adPIC and PIC3739 to induce tumor necrosis factor (TNF) in vivo and in cultured macrophages were compared. Infection with adPIC, but not PIC3739, was associated with detectable serum TNF that peaked in week 2 of infection. Tumor necrosis factor was found in the spleens of adPIC- and PIC3739-infected animals in week 1 of infection; TNF alpha mRNA levels in spleens and livers of adPIC infected animals increased and remained high throughout infection, whereas PIC3739-infected organs showed down regulation of TNF alpha mRNA late in infection. Peritoneal macrophages explanted from adPIC-infected animals showed enhanced lipopolysaccharide-inducible TNF production. Altered regulation of TNF production may play a role in the pathogenesis of guinea pig arenavirus disease.


Subject(s)
Disease Models, Animal , Hemorrhagic Fever, American/etiology , Pichinde virus/pathogenicity , Tumor Necrosis Factor-alpha/analysis , Animals , Blotting, Northern , Cells, Cultured , Guinea Pigs , Hemorrhagic Fever, American/virology , Liver/metabolism , Macrophages/metabolism , Macrophages, Peritoneal/metabolism , Male , RNA, Messenger/analysis , Spleen/cytology , Spleen/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
17.
Am J Pathol ; 145(1): 228-35, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8030751

ABSTRACT

A guinea pig passage-adapted strain of the arena-virus Pichinde (adPIC) is highly virulent in inbred guinea pigs, whereas the related strain PIC3739 is attenuated. Both viruses were macrophage tropic and infected peritoneal, splenic, liver, and alveolar macrophages during experimental Pichinde virus infection. Infection with the virulent strain was associated with unlimited viral replication in the face of exaggerated delayed-type hypersensitivity response, manifested by the macrophage disappearance reaction. Histopathological lesions unique to adPIC-infected guinea pigs included intestinal villus blunting with mucosal infiltration by pyknotic debris-laden macrophages and apoptosis of crypt epithelial cells. Splenic red pulp necrosis was also significantly associated with adPIC infection but not PIC3739 infection. These findings may provide clues to the pathogenesis of a group of poorly understood human viral hemorrhagic fevers.


Subject(s)
Hemorrhagic Fever, American/microbiology , Hemorrhagic Fever, American/pathology , Pichinde virus/pathogenicity , Animals , Disease Models, Animal , Guinea Pigs , Hemorrhagic Fever, American/mortality , Macrophages/microbiology , Macrophages/pathology , Necrosis , Species Specificity , Spleen/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...