Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Gen Virol ; 104(9)2023 09.
Article in English | MEDLINE | ID: mdl-37698490

ABSTRACT

Arenaviridae is a family for ambisense RNA viruses with genomes of about 10.5 kb that infect mammals, snakes, and fish. The arenavirid genome consists of two or three single-stranded RNA segments and encodes a nucleoprotein (NP), a glycoprotein (GP) and a large (L) protein containing RNA-directed RNA polymerase (RdRP) domains; some arenavirids encode a zinc-binding protein (Z). This is a summary of the International Committee on Taxonomy of Viruses (ICTV) report on the family Arenaviridae, which is available at www.ictv.global/report/arenaviridae.


Subject(s)
Arenaviridae , Animals , Arenaviridae/genetics , Nucleoproteins/genetics , RNA , RNA-Dependent RNA Polymerase , Mammals
2.
J Gen Virol ; 100(8): 1200-1201, 2019 08.
Article in English | MEDLINE | ID: mdl-31192784

ABSTRACT

Members of the family Arenaviridae produce enveloped virions containing genomes consisting of two or three single-stranded RNA segments totalling about 10.5 kb. Arenaviruses can infect mammals, including humans and other primates, snakes, and fish. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Arenaviridae, which is available at www.ictv.global/report/arenaviridae.


Subject(s)
Arenaviridae Infections/veterinary , Arenaviridae Infections/virology , Arenaviridae/classification , Arenaviridae/genetics , Animals , Arenaviridae/isolation & purification , Arenaviridae/ultrastructure , Fishes , Genome, Viral , Humans , Phylogeny , RNA, Viral/genetics , Reptiles , Viral Proteins/genetics
3.
J Virol ; 88(6): 3058-66, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24335292

ABSTRACT

The attenuated Lassa vaccine candidate ML29 is a laboratory-produced reassortant between Lassa and Mopeia viruses, two Old World arenaviruses that differ by 40% in nucleic acid sequence. In our previous studies, ML29 elicited sterilizing immunity against Lassa virus challenge in guinea pigs and marmosets and virus-specific cell-mediated immunity in both simian immunodeficiency virus (SIV)-infected and uninfected rhesus macaques. Here, we show that ML29 is stable after 12 passages in vitro without losing its plaque morphology or its attenuated phenotype in suckling mice. Additionally, we used deep sequencing to characterize the viral population comprising the original stock of ML29, the stock of ML29 after 12 passages in Vero cells, and the ML29 isolates obtained from vaccinated animals. Twenty-seven isolates bore approximately 77 mutations that exceeded 20% of the single-nucleotide polymorphism (SNP) changes at any single locus. Of these 77 mutations, 5 appeared to be host specific, for example, appearing in mice but not in primates. None of these mutations were reversions of ML29 to the sequences of the parental Lassa and Mopeia viruses. The host-specific mutations indicate viral adaptations to virus-host interactions, and such interactions make reasonable targets for antiviral approaches. Variants capable of chronic infection did not emerge from any of the primate infections, even in immune-deficient animals, indicating that the ML29 reassortant is reasonably stable in vivo. In conclusion, the preclinical studies of ML29 as a Lassa virus vaccine candidate have been advanced, showing high levels of protection in nonhuman primates and acceptable stability both in vitro and in vivo.


Subject(s)
Genetic Variation , Lassa Fever/prevention & control , Lassa virus/genetics , Lassa virus/immunology , Viral Vaccines/genetics , Animals , Callithrix , Chlorocebus aethiops , Humans , Immunity, Cellular , Lassa Fever/immunology , Lassa Fever/virology , Macaca mulatta , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vero Cells , Viral Vaccines/immunology
4.
Arch Virol ; 160(7): 1851-74, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25935216

ABSTRACT

Until recently, members of the monogeneric family Arenaviridae (arenaviruses) have been known to infect only muroid rodents and, in one case, possibly phyllostomid bats. The paradigm of arenaviruses exclusively infecting small mammals shifted dramatically when several groups independently published the detection and isolation of a divergent group of arenaviruses in captive alethinophidian snakes. Preliminary phylogenetic analyses suggest that these reptilian arenaviruses constitute a sister clade to mammalian arenaviruses. Here, the members of the International Committee on Taxonomy of Viruses (ICTV) Arenaviridae Study Group, together with other experts, outline the taxonomic reorganization of the family Arenaviridae to accommodate reptilian arenaviruses and other recently discovered mammalian arenaviruses and to improve compliance with the Rules of the International Code of Virus Classification and Nomenclature (ICVCN). PAirwise Sequence Comparison (PASC) of arenavirus genomes and NP amino acid pairwise distances support the modification of the present classification. As a result, the current genus Arenavirus is replaced by two genera, Mammarenavirus and Reptarenavirus, which are established to accommodate mammalian and reptilian arenaviruses, respectively, in the same family. The current species landscape among mammalian arenaviruses is upheld, with two new species added for Lunk and Merino Walk viruses and minor corrections to the spelling of some names. The published snake arenaviruses are distributed among three new separate reptarenavirus species. Finally, a non-Latinized binomial species name scheme is adopted for all arenavirus species. In addition, the current virus abbreviations have been evaluated, and some changes are introduced to unequivocally identify each virus in electronic databases, manuscripts, and oral proceedings.


Subject(s)
Arenaviridae Infections/veterinary , Arenaviridae Infections/virology , Arenavirus/classification , Animals , Arenaviridae Infections/history , Arenavirus/genetics , Arenavirus/isolation & purification , History, 20th Century , History, 21st Century , Humans , Phylogeny , Virology/history , Virology/trends
5.
J Virol ; 86(13): 7216-26, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532679

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV), the prototype arenavirus, and Lassa virus (LASV), the causative agent of Lassa fever (LF), have extensive strain diversity and significant variations in pathogenicity for humans and experimental animals. The WE strain of LCMV (LCMV-WE), but not the Armstrong (Arm) strain, induces a fatal LF-like disease in rhesus macaques. We also demonstrated that LASV infection of human macrophages and endothelial cells resulted in reduced levels of proinflammatory cytokines. Here we have shown that cells infected with LASV or with LCMV-WE suppressed Toll-like receptor 2 (TLR2)-dependent proinflammatory cytokine responses. The persisting isolate LCMV clone 13 (CL13) also failed to stimulate interleukin-6 (IL-6) in macrophages. In contrast, nonpathogenic Mopeia virus, which is a genetic relative of LASV and LCMV-Arm induced robust responses that were TLR2/Mal dependent, required virus replication, and were enhanced by CD14. Superinfection experiments demonstrated that the WE strain of LCMV inhibited the Arm-mediated IL-8 response during the early stage of infection. In cells transfected with the NF-κB-luciferase reporter, infection with LCMV-Arm resulted in the induction of NF-κB, but cells infected with LCMV-WE and CL13 did not. These results suggest that pathogenic arenaviruses suppress NF-κB-mediated proinflammatory cytokine responses in infected cells.


Subject(s)
Cytokines/antagonists & inhibitors , Immune Evasion , Lassa virus/pathogenicity , Lymphocytic choriomeningitis virus/pathogenicity , Myelin Proteins/antagonists & inhibitors , Proteolipids/antagonists & inhibitors , Toll-Like Receptor 2/antagonists & inhibitors , Animals , Cell Line , Humans , Lassa virus/immunology , Lymphocytic choriomeningitis virus/immunology , Membrane Transport Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins/immunology , Myelin and Lymphocyte-Associated Proteolipid Proteins , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Proteolipids/immunology , Toll-Like Receptor 2/immunology
6.
Virol J ; 10: 52, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23402317

ABSTRACT

BACKGROUND: Lassa hemorrhagic fever (LHF) is a rodent-borne viral disease that can be fatal for human beings. In this study, an attenuated Lassa vaccine candidate, ML29, was tested in SIV-infected rhesus macaques for its ability to elicit immune responses without instigating signs pathognomonic for arenavirus disease. ML29 is a reassortant between Lassa and Mopeia viruses that causes a transient infection in non-human primates and confers sterilizing protection from lethal Lassa viral challenge. However, since the LHF endemic area of West Africa also has high HIV seroprevalence, it is important to determine whether vaccination could be safe in the context of HIV infection. RESULTS: SIV-infected and uninfected rhesus macaques were vaccinated with the ML29 virus and monitored for specific humoral and cellular immune responses, as well as for classical and non-classical signs of arenavirus disease. Classical disease signs included viremia, rash, respiratory distress, malaise, high liver enzyme levels, and virus invasion of the central nervous system. Non-classical signs, derived from profiling the blood transcriptome of virulent and non-virulent arenavirus infections, included increased expression of interferon-stimulated genes (ISG) and decreased expression of COX2, IL-1ß, coagulation intermediates and nuclear receptors needed for stress signaling. All vaccinated monkeys showed ML29-specific antibody responses and ML29-specific cell-mediated immunity. CONCLUSION: SIV-infected and uninfected rhesus macaques responded similarly to ML29 vaccination, and none developed chronic arenavirus infection. Importantly, none of the macaques developed signs, classical or non-classical, of arenavirus disease.


Subject(s)
Coinfection/immunology , HIV Infections/immunology , Lassa Fever/prevention & control , Lassa virus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Coinfection/prevention & control , Coinfection/virology , HIV Infections/complications , HIV Infections/virology , Humans , Lassa Fever/complications , Lassa Fever/immunology , Lassa Fever/virology , Macaca mulatta , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Vaccines/administration & dosage
7.
Blood ; 114(6): 1196-204, 2009 Aug 06.
Article in English | MEDLINE | ID: mdl-19498020

ABSTRACT

Immune deficiency viruses such as SIV in macaques or HIV-1 in human beings have evolved mechanisms to defeat host immunity that also impact the efficacy of vaccines. A key factor for vaccine protection is whether immune responses elicited by prior immunization remain at levels sufficient to limit disease progression once a host is exposed to the pathogen. One potential mechanism for escaping pre-existing immunity is to trigger death among antigen-activated cells. We tested whether FasL/CD178 is involved in destroying preexisting immunity. Rhesus macaques were immunized with recombinant vesicular stomatitis virus vaccine expressing SIV Gag to elicit cellular immune responses, then treated with antibody that neutralizes FasL and challenged with intravenous SIVmac251. Compared with animals injected with control antibody, anti-FasL-treated macaques had superior preservation of central memory CD4(+) and CD8(+) cells and decreased regulatory T cells in the blood. The CD4(+) and CD8(+) lymphocytes from treated animals responded better to SIV Gag compared with controls, evidenced by higher cell-mediated immune responses to viral antigens for at least 17 weeks after SIV challenge. Anti-FasL treatment during the initial stages of acute SIV infection preserved the T-cell compartment and sustained cell-mediated immunity to SIV.


Subject(s)
Antibodies/pharmacology , Fas Ligand Protein/immunology , Gene Products, gag/immunology , Immunologic Memory , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Fas Ligand Protein/antagonists & inhibitors , HIV-1/immunology , Humans , Immunity, Cellular , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/therapy , T-Lymphocytes, Regulatory/immunology
8.
Pathogens ; 9(12)2020 Nov 26.
Article in English | MEDLINE | ID: mdl-33255985

ABSTRACT

Interferons (IFN) are crucial for the innate immune response. Slightly more than two decades ago, a new type of IFN was discovered: the lambda IFN (type III IFN). Like other IFN, the type III IFN display antiviral activity against a wide variety of infections, they induce expression of antiviral, interferon-stimulated genes (MX1, OAS, IFITM1), and they have immuno-modulatory activities that shape adaptive immune responses. Unlike other IFN, the type III IFN signal through distinct receptors is limited to a few cell types, primarily mucosal epithelial cells. As a consequence of their greater and more durable production in nasal and respiratory tissues, they can determine the outcome of respiratory infections. This review is focused on the role of IFN-λ in the pathogenesis of respiratory viral infections, with influenza as a prime example. The influenza virus is a major public health problem, causing up to half a million lethal infections annually. Moreover, the virus has been the cause of four pandemics over the last century. Although IFN-λ are increasingly being tested in antiviral therapy, they can have a negative influence on epithelial tissue recovery and increase the risk of secondary bacterial infections. Therefore, IFN-λ expression deserves increased scrutiny as a key factor in the host immune response to infection.

9.
Biomed Res Int ; 2020: 7465242, 2020.
Article in English | MEDLINE | ID: mdl-32258141

ABSTRACT

Recent comparisons between plant and animal viruses reveal many common principles that underlie how all viruses express their genetic material, amplify their genomes, and link virion assembly with replication. Cauliflower mosaic virus (CaMV) is not infectious for human beings. Here, we show that CaMV transactivator/viroplasmin protein (TAV) shares sequence similarity with and behaves like the human ribonuclease H1 (RNase H1) in reducing DNA/RNA hybrids detected with S9.6 antibody in HEK293T cells. We showed that TAV is clearly expressed in the cytosol and in the nuclei of transiently transfected human cells, similar to its distribution in plants. TAV also showed remarkable cytotoxic effects in U251 human glioma cells in vitro. These characteristics pave the way for future analysis on the use of the plant virus protein TAV, as an alternative to human RNAse H1 during gene therapy in human cells.


Subject(s)
Caulimovirus/enzymology , Glioma/drug therapy , Ribonuclease H , Viral Proteins , Cell Line, Tumor , Cytotoxins/chemistry , Cytotoxins/pharmacology , Glioma/metabolism , Glioma/pathology , HEK293 Cells , Humans , Ribonuclease H/chemistry , Ribonuclease H/pharmacology , Viral Proteins/chemistry , Viral Proteins/pharmacology
10.
Retrovirology ; 6: 91, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19832988

ABSTRACT

Human immunodeficiency virus disease involves progressive destruction of host immunity leading to opportunistic infections and increased rates for malignancies. Both depletion in immune cell numbers as well as defects in their effector functions are responsible for this immunodeficiency The broad impact of HIV reflects a similarly broad pattern of cell depletion including subsets that do not express viral receptors or support viral replication. Indirect cell killing, the destruction of uninfected cells, is due partly to activation of the Fas/FasL system for cell death. This death-signaling pathway is induced during HIV disease and contributes significantly to viral pathogenesis and disease.


Subject(s)
Cell Death , Fas Ligand Protein/physiology , HIV Infections/virology , HIV/pathogenicity , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , fas Receptor/physiology , Animals , Haplorhini , Humans , Models, Biological
11.
Virol J ; 6: 20, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19216742

ABSTRACT

BACKGROUND: Rhesus macaques infected with lymphocytic choriomeningitis virus (LCMV) provide a model for human Lassa fever. Disease begins with flu-like symptoms and progresses rapidly with fatal consequences. Previously, we profiled the blood transcriptome of LCMV-infected monkeys (M. Djavani et al J. Virol. 2007) showing distinct pre-viremic and viremic stages that discriminated virulent from benign infections. In the present study, changes in liver gene expression from macaques infected with virulent LCMV-WE were compared to gene expression in uninfected monkeys as well as to monkeys that were infected but not diseased. RESULTS: Based on a functional pathway analysis of differentially expressed genes, virulent LCMV-WE had a broader effect on liver cell function than did infection with non-virulent LCMV-Armstrong. During the first few days after infection, LCMV altered expression of genes associated with energy production, including fatty acid and glucose metabolism. The transcriptome profile resembled that of an organism in starvation: mRNA for acetyl-CoA carboxylase, a key enzyme of fatty acid synthesis was reduced while genes for enzymes in gluconeogenesis were up-regulated. Expression was also altered for genes associated with complement and coagulation cascades, and with signaling pathways involving STAT1 and TGF-beta. CONCLUSION: Most of the 4500 differentially expressed transcripts represented a general response to both virulent and mild infections. However, approximately 250 of these transcripts had significantly different expression in virulent infections as compared to mild infections, with approximately 30 of these being differentially regulated during the pre-viremic stage of infection. The genes that are expressed early and differently in mild and virulent disease are potential biomarkers for prognosis and triage of acute viral disease.


Subject(s)
Disease Models, Animal , Gene Expression Regulation , Liver/metabolism , Lymphocytic choriomeningitis virus/pathogenicity , Macaca mulatta , Proteins/metabolism , Animals , Arenaviridae Infections/pathology , Arenaviridae Infections/virology , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Proteins/genetics , Virulence
12.
Mem Inst Oswaldo Cruz ; 104(4): 583-91, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19722081

ABSTRACT

Rhesus macaques infected with the WE strain of lymphocytic choriomeningitis virus (LCMV-WE) serve as a model for human infection with Lassa fever virus. To identify the earliest events of acute infection, rhesus macaques were monitored immediately after lethal infection for changes in peripheral blood mononuclear cells (PBMCs). Changes in CD3, CD4, CD8 and CD20 subsets did not vary outside the normal fluctuations of these blood cell populations; however, natural killer (NK) and gammadelta T cells increased slightly on day 1 and then decreased significantly after two days. The NK subsets responsible for the decrease were primarily CD3-CD8+ or CD3-CD16+ and not the NKT (primarily CD3+CD56+) subset. Macaques infected with a non-virulent arenavirus, LCMV-Armstrong, showed a similar drop in circulating NK and gammadelta T cells, indicating that this is not a pathogenic event. V(3)9 T cells, representing the majority of circulating gammadelta T cells in rhesus macaques, displayed significant apoptosis when incubated with LCMV in cell culture; however, the low amount of cell death for virus-co-cultured NK cells was insufficient to account for the observed disappearance of this subset. Our observations in primates are similar to those seen in LCMV-infected mice, where decreased circulating NK cells were attributed to margination and cell death. Thus, the disappearance of these cells during acute hemorrhagic fever in rhesus macaques may be a cytokine-induced lymphopenia common to many virus infections.


Subject(s)
Apoptosis/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes/immunology , Viremia/immunology , Animals , Female , Flow Cytometry , Killer Cells, Natural/immunology , Lymphocytic Choriomeningitis/blood , Macaca mulatta , Reverse Transcriptase Polymerase Chain Reaction
13.
Vaccines (Basel) ; 7(1)2019 Jan 22.
Article in English | MEDLINE | ID: mdl-30678246

ABSTRACT

Viral hemorrhagic fevers (VHF) are a group of clinically similar diseases that can be caused by enveloped RNA viruses primarily from the families Arenaviridae, Filoviridae, Hantaviridae, and Flaviviridae. Clinically, this group of diseases has in common fever, fatigue, dizziness, muscle aches, and other associated symptoms that can progress to vascular leakage, bleeding and multi-organ failure. Most of these viruses are zoonotic causing asymptomatic infections in the primary host, but in human beings, the infection can be lethal. Clinical and experimental evidence suggest that the T-cell response is needed for protection against VHF, but can also cause damage to the host, and play an important role in disease pathogenesis. Here, we present a review of the T-cell immune responses to VHF and insights into the possible ways to improve counter-measures for these viral agents.

14.
Pathogens ; 8(3)2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31466243

ABSTRACT

Lassa fever surpasses Ebola, Marburg, and all other hemorrhagic fevers except Dengue in its public health impact. Caused by Lassa virus (LASV), the disease is a scourge on populations in endemic areas of West Africa, where reported incidence is higher. Here, we report construction, characterization, and preclinical efficacy of a novel recombinant vaccine candidate GEO-LM01. Constructed in the Modified Vaccinia Ankara (MVA) vector, GEO-LM01 expresses the glycoprotein precursor (GPC) and zinc-binding matrix protein (Z) from the prototype Josiah strain lineage IV. When expressed together, GP and Z form Virus-Like Particles (VLPs) in cell culture. Immunogenicity and efficacy of GEO-LM01 was tested in a mouse challenge model. A single intramuscular dose of GEO-LM01 protected 100% of CBA/J mice challenged with a lethal dose of ML29, a Mopeia/Lassa reassortant virus, delivered directly into the brain. In contrast, all control animals died within one week. The vaccine induced low levels of antibodies but Lassa-specific CD4+ and CD8+ T cell responses. This is the first report showing that a single dose of a replication-deficient MVA vector can confer full protection against a lethal challenge with ML29 virus.

15.
Pathogens ; 7(4)2018 Oct 28.
Article in English | MEDLINE | ID: mdl-30373278

ABSTRACT

In 2017, the global Coalition for Epidemic Preparedness (CEPI) declared Lassa virus disease to be one of the world's foremost biothreats. In January 2018, World Health Organization experts met to address the Lassa biothreat. It was commonly recognized that the diversity of Lassa virus (LASV) isolated from West African patient samples was far greater than that of the Ebola isolates from the West African epidemic of 2013⁻2016. Thus, vaccines produced against Lassa virus disease face the added challenge that they must be broadly-protective against a wide variety of LASV. In this review, we discuss what is known about the immune response to Lassa infection. We also discuss the approaches used to make broadly-protective influenza vaccines and how they could be applied to developing broad vaccine coverage against LASV disease. Recent advances in AIDS research are also potentially applicable to the design of broadly-protective medical countermeasures against LASV disease.

16.
Methods Mol Biol ; 1604: 79-88, 2018.
Article in English | MEDLINE | ID: mdl-28986826

ABSTRACT

There are two types of viral diagnostics: (1) those that detect components of the pathogen (like viral RNA or proteins) and (2) those that detect host molecules that rise or fall as a consequence of pathogen infection (like anti-viral antibodies or virus-induced inflammatory cytokines). Quantitative PCR to detect Lassa RNA, and clinical chemistry to detect high liver enzymes (AST/ALT) are commonly used to diagnose Lassa fever. Here, we discuss the various types of diagnostics for Lassa fever and the urgent need for early diagnosis. We also describe a protocol for using the attenuated Lassa vaccine candidate, ML29 , as an antigen for detecting Lassa-specific antibodies. Since antibodies are developed late in the progression of Lassa fever disease, this is not an early diagnostic, but is more useful in surveillance of the population to determine the sero-prevalence of antibodies to Lassa virus (LASV ), and to define treatment options for people in close contact with a Lassa-infected person.


Subject(s)
Lassa Fever/diagnosis , Lassa Fever/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/genetics , Antibody Formation/immunology , Early Diagnosis , Humans , Lassa Fever/genetics , Lassa virus/genetics , Lassa virus/immunology , Lassa virus/pathogenicity , Polymerase Chain Reaction , Viral Vaccines/immunology
17.
Methods Mol Biol ; 1604: 279-290, 2018.
Article in English | MEDLINE | ID: mdl-28986843

ABSTRACT

Lymphocytic choriomeningitis virus strain WE (LCMV-WE), a Risk Group 3 virus, causes a disease in rhesus monkeys that closely resembles human infection with Lassa fever virus, a Risk Group 4 agent. Three stages of disease progression have been defined and profiled in this model: pre-viremic, viremic, and terminal. The earliest or pre-viremic stage reveals changes in the blood profile predictive of the later stages of disease. In order to identify whether specific changes are pathognomonic, it was necessary to perform a parallel infection with an attenuated virus (LCMV-Armstrong). Here we review the use of nonhuman primates to model viral hemorrhagic fever and offer a step-by-step guide to using a rhesus macaque model for Lassa fever.


Subject(s)
Hemorrhagic Fevers, Viral/pathology , Hemorrhagic Fevers, Viral/virology , Animals , Disease Models, Animal , Humans , Lassa Fever/pathology , Lassa Fever/veterinary , Macaca mulatta
18.
Clin Dev Immunol ; 2007: 93462, 2007.
Article in English | MEDLINE | ID: mdl-18317535

ABSTRACT

Acute SIVmac infection in macaques is accompanied by high levels of plasma viremia that decline with the appearance of viral immunity and is a model for acute HIV disease in man. Despite specific immune responses, the virus establishes a chronic, persistent infection. The destruction of CD4+ and CD4- lymphocyte subsets in macaques contributes to viral persistence and suggests the importance of mechanisms for depleting both infected and uninfected (bystander) cells. Bystander cell killing can occur when FasL binds the Fas receptor on activated lymphocytes, which include T and B cell subpopulations that are responding to the infection. Destruction of specific immune cells could be an important mechanism for blunting viral immunity and establishing persistent infection with chronic disease. We inhibited the Fas pathway in vivo with a monoclonal antibody against FasL (RNOK203). Here we show that treatment with anti-FasL reduced cell death in circulating T and B cells, increased CTL and antibody responses to viral proteins, and lowered the setpoint viremia. By blocking FasL during only the first few weeks after infection, we attenuated SIVmac disease and increased the life span for infected and treated macaques.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cytotoxicity, Immunologic/drug effects , Fas Ligand Protein/antagonists & inhibitors , Simian Acquired Immunodeficiency Syndrome/prevention & control , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Apoptosis/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/virology , Cytotoxicity Tests, Immunologic , Flow Cytometry , Humans , Macaca , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus , T-Lymphocytes/drug effects , T-Lymphocytes/virology , Viral Proteins/immunology , Viremia/drug therapy
19.
J Zhejiang Univ Sci B ; 6(7): 668-72, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15973770

ABSTRACT

In our previous work we reported that HIV Tat and 6 cysteine rich peptides of Tat induce tumor necrosis factor-related apoptosis-induced ligand (TRAIL) in human monocytes (Yang et al., 2003). Here our results showed that HIV Tat and Tat cysteine rich peptide increase CCR5 expression in human monocytes, and this activity is inhibited by rabbit anti-Tat. Boiled Tat does not increase CCR5 expression in monocytes. These results provide insight into a new mechanism by which HIV Tat plays a key role in the pathogenesis of HIV-1 infection.


Subject(s)
Cysteine/chemistry , Gene Products, tat/chemistry , Gene Products, tat/pharmacology , Monocytes/metabolism , Amino Acid Sequence , Cells, Cultured , Dose-Response Relationship, Drug , Extracellular Fluid/chemistry , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Molecular Sequence Data , Monocytes/drug effects , Peptides/chemistry
20.
Future Virol ; 10(3): 233-256, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25844088

ABSTRACT

Lassa virus infection elicits distinctive changes in host gene expression and metabolism. We focus on changes in host gene expression that may be biomarkers that discriminate individual pathogens or may help to provide a prognosis for disease. In addition to assessing mRNA changes, functional studies are also needed to discriminate causes of disease from mechanisms of host resistance. Host responses that drive pathogenesis are likely to be targets for prevention or therapy. Host responses to Lassa or its related arenaviruses have been monitored in cell culture, in animal models of hemorrhagic fever, in Lassa-infected nonhuman primates and, to a limited extent, in infected human beings. Here, we describe results from those studies and discuss potential targets for reducing virus replication and mitigating disease.

SELECTION OF CITATIONS
SEARCH DETAIL