Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Sci Rep ; 12(1): 19911, 2022 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-36402782

RESUMEN

LHF-535 is a small molecule antiviral currently in development for the treatment of Lassa fever, a zoonotic disease endemic in West Africa that generates significant morbidity and mortality. Current treatment options are inadequate, and there are no approved therapeutics or vaccines for Lassa fever. LHF-535 was evaluated in a lethal guinea pig model of Lassa pathogenesis, using once-daily administration of a fixed dose (50 mg/kg/day) initiating either 1 or 3 days after inoculation with a lethal dose of Lassa virus. LHF-535 reduced viremia and clinical signs and protected all animals from lethality. A subset of surviving animals was rechallenged four months later with a second lethal challenge of Lassa virus and were found to be protected from disease. LHF-535 pharmacokinetics at the protective dose in guinea pigs showed plasma concentrations well within the range observed in clinical trials in healthy volunteers, supporting the continued development of LHF-535 as a Lassa therapeutic.


Asunto(s)
Fiebre de Lassa , Cobayas , Animales , Fiebre de Lassa/tratamiento farmacológico , Fiebre de Lassa/prevención & control , Antivirales/farmacología , Antivirales/uso terapéutico , Virus Lassa , Viremia/tratamiento farmacológico , Vacunación
2.
PLoS Pathog ; 14(12): e1007439, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30576397

RESUMEN

Arenaviruses are a significant cause of hemorrhagic fever, an often-fatal disease for which there is no approved antiviral therapy. Lassa fever in particular generates high morbidity and mortality in West Africa, where the disease is endemic, and a recent outbreak in Nigeria was larger and more geographically diverse than usual. We are developing LHF-535, a small-molecule viral entry inhibitor that targets the arenavirus envelope glycoprotein, as a therapeutic candidate for Lassa fever and other hemorrhagic fevers of arenavirus origin. Using a lentiviral pseudotype infectivity assay, we determined that LHF-535 had sub-nanomolar potency against the viral envelope glycoproteins from all Lassa virus lineages, with the exception of the glycoprotein from the LP strain from lineage I, which was 100-fold less sensitive than that of other strains. This reduced sensitivity was mediated by a unique amino acid substitution, V434I, in the transmembrane domain of the envelope glycoprotein GP2 subunit. This position corresponds to the attenuation determinant of Candid#1, a live-attenuated Junín virus vaccine strain used to prevent Argentine hemorrhagic fever. Using a virus-yield reduction assay, we determined that LHF-535 potently inhibited Junín virus, but not Candid#1, and the Candid#1 attenuation determinant, F427I, regulated this difference in sensitivity. We also demonstrated that a daily oral dose of LHF-535 at 10 mg/kg protected mice from a lethal dose of Tacaribe virus. Serial passage of Tacaribe virus in LHF-535-treated Vero cells yielded viruses that were resistant to LHF-535, and the majority of drug-resistant viruses exhibited attenuated pathogenesis. These findings provide a framework for the clinical development of LHF-535 as a broad-spectrum inhibitor of arenavirus entry and provide an important context for monitoring the emergence of drug-resistant viruses.


Asunto(s)
Antivirales/farmacología , Fiebre de Lassa , Virus Lassa/genética , Virulencia/efectos de los fármacos , Virulencia/genética , Animales , Chlorocebus aethiops , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Células HEK293 , Humanos , Virus Lassa/efectos de los fármacos , Ratones , Mutación , Células Vero , Proteínas del Envoltorio Viral/genética
3.
Cell Host Microbe ; 19(2): 254-66, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26867183

RESUMEN

Pandemic influenza viruses modulate proinflammatory responses that can lead to immunopathogenesis. We present an extensive and systematic profiling of lipids, metabolites, and proteins in respiratory compartments of ferrets infected with either 1918 or 2009 human pandemic H1N1 influenza viruses. Integrative analysis of high-throughput omics data with virologic and histopathologic data uncovered relationships between host responses and phenotypic outcomes of viral infection. Proinflammatory lipid precursors in the trachea following 1918 infection correlated with severe tracheal lesions. Using an algorithm to infer cell quantity changes from gene expression data, we found enrichment of distinct T cell subpopulations in the trachea. There was also a predicted increase in inflammatory monocytes in the lung of 1918 virus-infected animals that was sustained throughout infection. This study presents a unique resource to the influenza research community and demonstrates the utility of an integrative systems approach for characterization of lipid metabolism alterations underlying respiratory responses to viruses.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Metabolismo de los Lípidos , Animales , Modelos Animales de Enfermedad , Hurones , Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Gripe Humana/epidemiología , Gripe Humana/genética , Gripe Humana/patología , Lípidos/química , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Metabolómica
4.
J Virol ; 89(5): 2543-52, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25520505

RESUMEN

UNLABELLED: To identify host factors associated with arenavirus virulence, we used a cynomolgus macaque model to evaluate the pathogenesis of Lujo virus (LUJV), a recently emerged arenavirus that caused an outbreak of severe viral hemorrhagic fever in southern Africa. In contrast to human cases, LUJV caused mild, nonlethal illness in macaques. We then compared this to contrasting clinical outcomes during arenavirus infection, specifically to samples obtained from macaques infected with three highly pathogenic lines of Lassa virus (LASV), the causative agent of Lassa fever (LF). We assessed gene expression in peripheral blood mononuclear cells (PBMC) and determined genes that significantly changed expression relative to that in uninfected animals over the course of infection. We detected a 72-h delay in the induction of host responses to infection during LUJV infection compared to that of the animals infected with LASV. This included genes associated with inflammatory and antiviral responses and was particularly apparent among groups of genes promoting cell death. We also observed early differential expression of a subset of genes specific to LUJV infection that accounts for the delayed inflammatory response. Cell type enrichment analysis suggested that host response induction delay and an LUJV-specific profile are due to a different proportion of natural killer cells responding in LUJV infection than that in the LASV-infected animals. Together, these data indicate that delayed proinflammatory and proapoptotic host responses to arenavirus infection could ameliorate disease severity. This conclusion provides insight into the cellular and molecular mechanisms of arenaviral hemorrhagic fever and suggests potential strategies for therapeutic development. IMPORTANCE: Old World arenaviruses are significant human pathogens that often are associated with high mortality. However, mechanisms underlying disease severity and virulence in arenavirus hemorrhagic fever are largely unknown, particularly regarding host responses that contribute to pathogenicity. This study describes a comparison between Lujo and Lassa virus infection in cynomolgus macaques. Lujo virus-infected macaques developed only mild illness, while Lassa virus-infected macaques developed severe illness consistent with Lassa fever. We determined that mild disease is associated with a delay in host expression of genes linked to virulence, such as those causing inflammation and cell death, and with distinct cell types that may mediate this delay. This is the first study to associate the timing and directionality of gene expression with arenaviral pathogenicity and disease outcome and evokes new potential approaches for developing effective therapeutics for treating these deadly emerging pathogens.


Asunto(s)
Infecciones por Arenaviridae/patología , Infecciones por Arenaviridae/virología , Fiebres Hemorrágicas Virales/patología , Fiebres Hemorrágicas Virales/virología , Lujo virus/patogenicidad , Animales , Infecciones por Arenaviridae/inmunología , Muerte Celular , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Fiebres Hemorrágicas Virales/inmunología , Inflamación/patología , Células Asesinas Naturales/inmunología , Fiebre de Lassa/patología , Fiebre de Lassa/virología , Virus Lassa/patogenicidad , Leucocitos Mononucleares/inmunología , Macaca fascicularis , Factores de Tiempo
5.
Science ; 346(6212): 987-91, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25359852

RESUMEN

Existing mouse models of lethal Ebola virus infection do not reproduce hallmark symptoms of Ebola hemorrhagic fever, neither delayed blood coagulation and disseminated intravascular coagulation nor death from shock, thus restricting pathogenesis studies to nonhuman primates. Here we show that mice from the Collaborative Cross panel of recombinant inbred mice exhibit distinct disease phenotypes after mouse-adapted Ebola virus infection. Phenotypes range from complete resistance to lethal disease to severe hemorrhagic fever characterized by prolonged coagulation times and 100% mortality. Inflammatory signaling was associated with vascular permeability and endothelial activation, and resistance to lethal infection arose by induction of lymphocyte differentiation and cellular adhesion, probably mediated by the susceptibility allele Tek. These data indicate that genetic background determines susceptibility to Ebola hemorrhagic fever.


Asunto(s)
Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Fiebre Hemorrágica Ebola/genética , Fiebre Hemorrágica Ebola/inmunología , Interacciones Huésped-Patógeno/genética , Ratones , Receptor TIE-2/genética , Alelos , Animales , Coagulación Sanguínea/genética , Permeabilidad Capilar/genética , Endotelio Vascular/fisiopatología , Fiebre Hemorrágica Ebola/sangre , Hígado/irrigación sanguínea , Hígado/metabolismo , Hígado/patología , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Neovascularización Fisiológica/genética
6.
J Virol ; 88(16): 8768-82, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24850744

RESUMEN

UNLABELLED: Polyadenylated mature mRNAs are the focus of standard transcriptome analyses. However, the profiling of nascent transcripts, which often include nonpolyadenylated RNAs, can unveil novel insights into transcriptional regulation. Here, we separately sequenced total RNAs (Total RNAseq) and mRNAs (mRNAseq) from the same HIV-1-infected human CD4(+) T cells. We found that many nonpolyadenylated RNAs were differentially expressed upon HIV-1 infection, and we identified 8 times more differentially expressed genes at 12 h postinfection by Total RNAseq than by mRNAseq. These expression changes were also evident by concurrent changes in introns and were recapitulated by later mRNA changes, revealing an unexpectedly significant delay between transcriptional initiation and mature mRNA production early after HIV-1 infection. We computationally derived and validated the underlying regulatory programs, and we predicted drugs capable of reversing these HIV-1-induced expression changes followed by experimental confirmation. Our results show that combined total and mRNA transcriptome analysis is essential for fully capturing the early host response to virus infection and provide a framework for identifying candidate drugs for host-directed therapy against HIV/AIDS. IMPORTANCE: In this study, we used mass sequencing to identify genes differentially expressed in CD4(+) T cells during HIV-1 infection. To our surprise, we found many differentially expressed genes early after infection by analyzing both newly transcribed unprocessed pre-mRNAs and fully processed mRNAs, but not by analyzing mRNAs alone, indicating a significant delay between transcription initiation and mRNA production early after HIV-1 infection. These results also show that important findings could be missed by the standard practice of analyzing mRNAs alone. We then derived the regulatory mechanisms driving the observed expression changes using integrative computational analyses. Further, we predicted drugs that could reverse the observed expression changes induced by HIV-1 infection and showed that one of the predicted drugs indeed potently inhibited HIV-1 infection. This shows that it is possible to identify candidate drugs for host-directed therapy against HIV/AIDS using our genomics-based approach.


Asunto(s)
Infecciones por VIH/genética , VIH-1/genética , Transcripción Genética/genética , Linfocitos T CD4-Positivos/virología , Línea Celular , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Infecciones por VIH/virología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , ARN/genética , ARN Mensajero/genética , Replicación Viral/genética
7.
ILAR J ; 54(2): 166-80, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24174440

RESUMEN

Nonhuman primate (NHP) biomedical models are critical to our understanding of human health and disease, yet we are still in the early stages of developing sufficient tools to support primate genomic research that allow us to better understand the basis of phenotypic traits in NHP models of disease. A mere 7 years ago, the limited NHP transcriptome profiling that was being performed was done using complementary DNA arrays based on human genome sequences, and the lack of NHP genomic information and immunologic reagents precluded the use of NHPs in functional genomic studies. Since then, significant strides have been made in developing genomics capabilities for NHP research, from the rhesus macaque genome sequencing project to the construction of the first macaque-specific high-density oligonucleotide microarray, paving the way for further resource development and additional primate sequencing projects. Complete published draft genome sequences are now available for the chimpanzee ( Chimpanzee Sequencing Analysis Consortium 2005), bonobo ( Prufer et al. 2012), gorilla ( Scally et al. 2012), and baboon ( Ensembl.org 2013), along with the recently completed draft genomes for the cynomolgus macaque and Chinese rhesus macaque. Against this backdrop of both expanding sequence data and the early application of sequence-derived DNA microarrays tools, we will contextualize the development of these community resources and their application to infectious disease research through a literature review of NHP models of acquired immune deficiency syndrome and models of respiratory virus infection. In particular, we will review the use of -omics approaches in studies of simian immunodeficiency virus and respiratory virus pathogenesis and vaccine development, emphasizing the acute and innate responses and the relationship of these to the course of disease and to the evolution of adaptive immunity.


Asunto(s)
Cercopithecidae/genética , Genómica/métodos , Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/veterinaria , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Virus de la Inmunodeficiencia de los Simios/genética , Animales , Humanos , Virus de la Influenza A/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos/veterinaria , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Análisis de Secuencia de ADN/veterinaria , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología
8.
Curr Opin Immunol ; 25(5): 588-92, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24011665

RESUMEN

The advent of publicly available databases containing system-wide phenotypic data of the host response to both drugs and pathogens, in conjunction with bioinformatics and computational methods now allows for in silico predictions of FDA-approved drugs as treatments against infection diseases. This systems biology approach captures the complexity of both the pathogen and drug host response in the form of expression patterns or molecular interaction networks without having to understand the underlying mechanisms of action. These drug repurposing techniques have been successful in identifying new drug candidates for several types of cancers and were recently used to identify potential therapeutics against influenza, the newly discovered Middle Eastern Respiratory Syndrome coronavirus and several parasitic diseases. These new approaches have the potential to significantly reduce both the time and cost for infectious diseases drug discovery.


Asunto(s)
Enfermedades Transmisibles/tratamiento farmacológico , Reposicionamiento de Medicamentos , Animales , Redes Reguladoras de Genes , Genómica , Humanos , Biología de Sistemas
9.
Nat Rev Microbiol ; 11(7): 455-66, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23728212

RESUMEN

High-throughput molecular profiling and computational biology are changing the face of virology, providing a new appreciation of the importance of the host in viral pathogenesis and offering unprecedented opportunities for better diagnostics, therapeutics and vaccines. Here, we provide a snapshot of the evolution of systems virology, from global gene expression profiling and signatures of disease outcome, to geometry-based computational methods that promise to yield novel therapeutic targets, personalized medicine and a deeper understanding of how viruses cause disease. To realize these goals, pipettes and Petri dishes need to join forces with the powers of mathematics and computational biology.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Biología de Sistemas/métodos , Virología/métodos , Virosis/tratamiento farmacológico , Virosis/virología , Animales , Biología Computacional , Ensayos Analíticos de Alto Rendimiento/métodos , Interacciones Huésped-Patógeno , Humanos , Virulencia , Virus/patogenicidad
10.
Semin Immunol ; 25(3): 228-39, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23218769

RESUMEN

Influenza virus research has recently undergone a shift from a virus-centric perspective to one that embraces the full spectrum of virus-host interactions and cellular signaling events that determine disease outcome. This change has been brought about by the increasing use and expanding scope of high-throughput molecular profiling and computational biology, which together fuel discovery in systems biology. In this review, we show how these approaches have revealed an uncontrolled inflammatory response as a contributor to the extreme virulence of the 1918 pandemic and avian H5N1 viruses, and how this response differs from that induced by the 2009 H1N1 viruses responsible for the most recent influenza pandemic. We also discuss how new animal models, such as the Collaborative Cross mouse systems genetics platform, are key to the necessary systematic investigation of the impact of host genetics on infection outcome, how genome-wide RNAi screens have identified hundreds of cellular factors involved in viral replication, and how systems biology approaches are making possible the rational design of new drugs and vaccines against an ever-evolving respiratory virus.


Asunto(s)
Interacciones Huésped-Patógeno , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/inmunología , Biología de Sistemas/métodos , Animales , Biología Computacional , Modelos Animales de Enfermedad , Interacción Gen-Ambiente , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunidad/genética , Vacunas contra la Influenza/inmunología , Gripe Humana/epidemiología , Gripe Humana/genética , Ratones , Orthomyxoviridae/patogenicidad , Infecciones por Orthomyxoviridae/genética , Pandemias , Virulencia
11.
Virology ; 429(1): 37-46, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22542004

RESUMEN

Human immunodeficiency virus (HIV-1) depends upon host-encoded proteins to facilitate its replication while at the same time inhibiting critical components of innate and/or intrinsic immune response pathways. To characterize the host cell response on protein levels in CD4+ lymphoblastoid SUP-T1 cells after infection with HIV-1 strain LAI, we used mass spectrometry (MS)-based global quantitation with iTRAQ (isobaric tag for relative and absolute quantification). We found 266, 60 and 22 proteins differentially expressed (DE) (P-value ≤ 0.05) at 4, 8, and 20 hours post-infection (hpi), respectively, compared to time-matched mock-infected samples. The majority of changes in protein abundance occurred at an early stage of infection well before the de novo production of viral proteins. Functional analyses of these DE proteins showed enrichment in several biological pathways including protein synthesis, cell proliferation, and T-cell activation. Importantly, these early changes before the time of robust viral production have not been described before.


Asunto(s)
Linfocitos T CD4-Positivos/química , Infecciones por VIH/genética , VIH-1/fisiología , Proteómica , Linfocitos T/química , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Línea Celular , Proliferación Celular , Redes Reguladoras de Genes , Infecciones por VIH/inmunología , Infecciones por VIH/fisiopatología , Infecciones por VIH/virología , VIH-1/genética , Interacciones Huésped-Patógeno , Humanos , Biosíntesis de Proteínas , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología
12.
Virology ; 430(1): 43-52, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22608059

RESUMEN

Hepatitis C virus/human immunodeficiency virus (HCV/HIV) coinfected patients demonstrate accelerated progression to severe liver injury in comparison to HCV monoinfected patients, although the underlying mechanisms are unclear owing to infection of separate tissue compartments with two distinct viral pathogens. Microarray analysis of paired liver biopsy and peripheral blood mononuclear cell (PBMC) specimens from HCV/HIV coinfected and HCV monoinfected patients identified a gene expression signature associated with increased inflammation and immune activation that was present only in liver and PBMC samples from coinfected patients. We also identified in these samples liver- and PBMC-specific signatures enriched with fibrogenic/hepatic stellate activation and proinflammatory genes, respectively. Finally, Bayesian networks were constructed by assimilating these data with existing data from liver and PBMC samples from other cohorts, augmenting enrichment of biologically important pathways and further indicating that chronic immune activation in HCV/HIV coinfection may exacerbate liver disease progression in coinfected patients.


Asunto(s)
Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Hepatitis C/complicaciones , Hepatitis C/inmunología , Leucocitos Mononucleares/inmunología , Hígado/inmunología , Activación de Linfocitos , Adulto , Biopsia , Citocinas/biosíntesis , Femenino , Perfilación de la Expresión Génica , Células Estrelladas Hepáticas/inmunología , Humanos , Hígado/patología , Masculino , Análisis por Micromatrices , Persona de Mediana Edad
13.
Microbiol Mol Biol Rev ; 76(1): 16-32, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22390970

RESUMEN

The cytokine storm has captured the attention of the public and the scientific community alike, and while the general notion of an excessive or uncontrolled release of proinflammatory cytokines is well known, the concept of a cytokine storm and the biological consequences of cytokine overproduction are not clearly defined. Cytokine storms are associated with a wide variety of infectious and noninfectious diseases. The term was popularized largely in the context of avian H5N1 influenza virus infection, bringing the term into popular media. In this review, we focus on the cytokine storm in the context of virus infection, and we highlight how high-throughput genomic methods are revealing the importance of the kinetics of cytokine gene expression and the remarkable degree of redundancy and overlap in cytokine signaling. We also address evidence for and against the role of the cytokine storm in the pathology of clinical and infectious disease and discuss why it has been so difficult to use knowledge of the cytokine storm and immunomodulatory therapies to improve the clinical outcomes for patients with severe acute infections.


Asunto(s)
Citocinas/metabolismo , Animales , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/virología , Citocinas/inmunología , Humanos , Subtipo H5N1 del Virus de la Influenza A/inmunología , Transducción de Señal/inmunología , Transducción de Señal/fisiología
14.
Hepatology ; 56(1): 17-27, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22278598

RESUMEN

UNLABELLED: Liver failure resulting from chronic hepatitis C virus (HCV) infection is a major cause for liver transplantation worldwide. Recurrent infection of the graft is universal in HCV patients after transplant and results in a rapid progression to severe fibrosis and end-stage liver disease in one third of all patients. No single clinical variable, or combination thereof, has, so far, proven accurate in identifying patients at risk of hepatic decompensation in the transplant setting. A combination of longitudinal, dimensionality reduction and categorical analysis of the transcriptome from 111 liver biopsy specimens taken from 57 HCV-infected patients over time identified a molecular signature of gene expression of patients at risk of developing severe fibrosis. Significantly, alterations in gene expression occur before histologic evidence of liver disease progression, suggesting that events that occur during the acute phase of infection influence patient outcome. Additionally, a common precursor state for different severe clinical outcomes was identified. CONCLUSION: Based on this patient cohort, incidence of severe liver disease is a process initiated early during HCV infection of the donor organ. The probable cellular network at the basis of the initial transition to severe liver disease was identified and characterized.


Asunto(s)
Rechazo de Injerto/genética , Hepatitis C Crónica/complicaciones , Fallo Hepático/cirugía , Trasplante de Hígado/efectos adversos , Activación Transcripcional/genética , Anciano , Biopsia con Aguja , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Regulación de la Expresión Génica , Hepacivirus/genética , Hepacivirus/fisiología , Hepatitis C Crónica/patología , Hepatitis C Crónica/cirugía , Humanos , Inmunohistoquímica , Fallo Hepático/etiología , Fallo Hepático/genética , Trasplante de Hígado/métodos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/genética , Complicaciones Posoperatorias/fisiopatología , Pronóstico , Recurrencia , Medición de Riesgo , Índice de Severidad de la Enfermedad , Donantes de Tejidos
15.
J Virol ; 86(5): 2437-46, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22190720

RESUMEN

Understanding the role of host factors during lethal influenza virus infection is critical to deciphering the events that determine the fate of the host. One such factor is encoded by the Mx1 gene, which confers resistance to influenza virus infection. Here, we compared pathology and global gene expression profiles in lung tissue from BALB/c (Mx1(-)) and BALB · A2G-Mx1 mice (Mx1(+/+)) infected with the fully reconstructed 1918 pandemic influenza virus. Mx1(+/+) mice showed less tissue damage than Mx(-) animals, and pathology and mortality were further reduced by treating the mice with interferon prior to infection. Using global transcriptional profiling, we identified distinct molecular signatures associated with partial protection, complete protection, and the contribution of interferon to the host response. In the absence of interferon treatment, partial protection was characterized by the generation of an acute response with the upregulation of genes associated with apoptosis, reactive oxygen species, and cell migration. Complete protection was characterized by the downregulation of cytokine and chemokine genes previously associated with influenza virus pathogenesis. The contribution of interferon treatment to total protection in virus-infected Mx1(+/+) mice was characterized by the altered regulation of cell cycle genes. These genes were upregulated in Mx1(+/+) mice treated with interferon but downregulated in the absence of interferon treatment. Our results suggest that Mx1(+/+) mice generate a protective antiviral response by controlling the expression of key modulator molecules associated with influenza virus lethality.


Asunto(s)
Proteínas de Unión al GTP/inmunología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Gripe Humana/mortalidad , Animales , Resistencia a la Enfermedad , Femenino , Proteínas de Unión al GTP/genética , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/patología , Gripe Humana/virología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas de Resistencia a Mixovirus , Sobrevida
16.
mBio ; 2(6)2011.
Artículo en Inglés | MEDLINE | ID: mdl-22086488

RESUMEN

UNLABELLED: We previously reported widespread differential expression of long non-protein-coding RNAs (ncRNAs) in response to virus infection. Here, we expanded the study through small RNA transcriptome sequencing analysis of the host response to both severe acute respiratory syndrome coronavirus (SARS-CoV) and influenza virus infections across four founder mouse strains of the Collaborative Cross, a recombinant inbred mouse resource for mapping complex traits. We observed differential expression of over 200 small RNAs of diverse classes during infection. A majority of identified microRNAs (miRNAs) showed divergent changes in expression across mouse strains with respect to SARS-CoV and influenza virus infections and responded differently to a highly pathogenic reconstructed 1918 virus compared to a minimally pathogenic seasonal influenza virus isolate. Novel insights into miRNA expression changes, including the association with pathogenic outcomes and large differences between in vivo and in vitro experimental systems, were further elucidated by a survey of selected miRNAs across diverse virus infections. The small RNAs identified also included many non-miRNA small RNAs, such as small nucleolar RNAs (snoRNAs), in addition to nonannotated small RNAs. An integrative sequencing analysis of both small RNAs and long transcripts from the same samples showed that the results revealing differential expression of miRNAs during infection were largely due to transcriptional regulation and that the predicted miRNA-mRNA network could modulate global host responses to virus infection in a combinatorial fashion. These findings represent the first integrated sequencing analysis of the response of host small RNAs to virus infection and show that small RNAs are an integrated component of complex networks involved in regulating the host response to infection. IMPORTANCE: Most studies examining the host transcriptional response to infection focus only on protein-coding genes. However, mammalian genomes transcribe many short and long non-protein-coding RNAs (ncRNAs). With the advent of deep-sequencing technologies, systematic transcriptome analysis of the host response, including analysis of ncRNAs of different sizes, is now possible. Using this approach, we recently discovered widespread differential expression of host long (>200 nucleotide [nt]) ncRNAs in response to virus infection. Here, the samples described in the previous report were again used, but we sequenced another fraction of the transcriptome to study very short (about 20 to 30 nt) ncRNAs. We demonstrated that virus infection also altered expression of many short ncRNAs of diverse classes. Putting the results of the two studies together, we show that small RNAs may also play an important role in regulating the host response to virus infection.


Asunto(s)
Infecciones por Coronavirus/inmunología , Regulación de la Expresión Génica , Pulmón/inmunología , Pulmón/virología , MicroARNs/biosíntesis , Infecciones por Orthomyxoviridae/inmunología , Transcriptoma , Animales , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , ARN Nucleolar Pequeño
17.
J Virol ; 85(22): 11646-54, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21917952

RESUMEN

We previously employed systems biology approaches to identify the mitochondrial fatty acid oxidation enzyme dodecenoyl coenzyme A delta isomerase (DCI) as a bottleneck protein controlling host metabolic reprogramming during hepatitis C virus (HCV) infection. Here we present the results of studies confirming the importance of DCI to HCV pathogenesis. Computational models incorporating proteomic data from HCV patient liver biopsy specimens recapitulated our original predictions regarding DCI and link HCV-associated alterations in cellular metabolism and liver disease progression. HCV growth and RNA replication in hepatoma cell lines stably expressing DCI-targeting short hairpin RNA (shRNA) were abrogated, indicating that DCI is required for productive infection. Pharmacologic inhibition of fatty acid oxidation also blocked HCV replication. Production of infectious HCV was restored by overexpression of an shRNA-resistant DCI allele. These findings demonstrate the utility of systems biology approaches to gain novel insight into the biology of HCV infection and identify novel, translationally relevant therapeutic targets.


Asunto(s)
Isomerasas de Doble Vínculo Carbono-Carbono/metabolismo , Hepacivirus/patogenicidad , Interacciones Huésped-Patógeno , Mitocondrias/enzimología , Replicación Viral , Biopsia , Línea Celular , Dodecenoil-CoA Isomerasa , Ácidos Grasos/metabolismo , Silenciador del Gen , Hepatocitos/enzimología , Hepatocitos/virología , Humanos , Hígado/química , Hígado/patología , Oxidación-Reducción , Proteoma
18.
J Virol ; 85(17): 9060-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21734050

RESUMEN

Ebola virus is the etiologic agent of a lethal hemorrhagic fever in humans and nonhuman primates with mortality rates of up to 90%. Previous studies with Zaire Ebola virus (ZEBOV), mouse-adapted virus (MA-ZEBOV), and mutant viruses (ZEBOV-NP(ma), ZEBOV-VP24(ma), and ZEBOV-NP/VP24(ma)) allowed us to identify the mutations in viral protein 24 (VP24) and nucleoprotein (NP) responsible for acquisition of high virulence in mice. To elucidate specific molecular signatures associated with lethality, we compared global gene expression profiles in spleen samples from mice infected with these viruses and performed an extensive functional analysis. Our analysis showed that the lethal viruses (MA-ZEBOV and ZEBOV-NP/VP24(ma)) elicited a strong expression of genes 72 h after infection. In addition, we found that although the host transcriptional response to ZEBOV-VP24(ma) was nearly the same as that to ZEBOV-NP/VP24(ma), the contribution of a mutation in the NP gene was required for a lethal phenotype. Further analysis indicated that one of the most relevant biological functions differentially regulated by the lethal viruses was the inflammatory response, as was the induction of specific metalloproteinases, which were present in our newly identify functional network that was associated with Ebola virus lethality. Our results suggest that this dysregulated proinflammatory response increased the severity of disease. Consequently, the newly discovered molecular signature could be used as the starting point for the development of new drugs and therapeutics. To our knowledge, this is the first study that clearly defines unique molecular signatures associated with Ebola virus lethality.


Asunto(s)
Ebolavirus/patogenicidad , Perfilación de la Expresión Génica , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/patología , Inflamación/inmunología , Inflamación/patología , Metaloproteasas/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Fiebre Hemorrágica Ebola/mortalidad , Ratones , Ratones Endogámicos BALB C , Enfermedades de los Roedores/inmunología , Enfermedades de los Roedores/mortalidad , Enfermedades de los Roedores/patología , Bazo/inmunología , Bazo/patología
19.
J Gen Virol ; 92(Pt 9): 2093-2104, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21593271

RESUMEN

The NS1 protein of influenza virus counters host antiviral defences primarily by antagonizing the type I interferon (IFN) response. Both the N-terminal dsRNA-binding domain and the C-terminal effector domain are required for optimal suppression of host responses during infection. To better understand the regulatory role of the NS1 effector domain, we used an NS1-truncated mutant virus derived from human H1N1 influenza isolate A/Texas/36/91 (Tx/91) and assessed global transcriptional profiles from two independent human lung cell-culture models. Relative to the wild-type Tx/91-induced gene expression, the NS1 mutant virus induced enhanced expression of innate immune genes, specifically NF-κB signalling-pathway genes and IFN-α and -ß target genes. We queried an experimentally derived IFN gene set to gauge the proportion of IFN-responsive genes that are suppressed specifically by NS1. We show that the C-terminally truncated NS1 mutant virus is less efficient at suppressing IFN-regulated gene expression associated with activation of antigen-presentation and immune-proteasome pathways. This is the first report integrating genomic analysis from two independent human culture systems, including primary lung cells, using genetically similar H1N1 influenza viruses that differ only in the length of the NS1 protein.


Asunto(s)
Presentación de Antígeno , Subtipo H1N1 del Virus de la Influenza A/inmunología , Interferón-alfa/antagonistas & inhibidores , Interferón beta/antagonistas & inhibidores , Inhibidores de Proteasoma , Proteínas no Estructurales Virales/metabolismo , Factores de Virulencia/metabolismo , Células Cultivadas , Células Epiteliales/inmunología , Células Epiteliales/virología , Perfilación de la Expresión Génica , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Proteínas Mutantes/metabolismo , FN-kappa B/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Factores de Virulencia/inmunología
20.
mBio ; 2(1): e00325-10, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21285433

RESUMEN

The twentieth century was marked by extraordinary advances in our understanding of microbes and infectious disease, but pandemics remain, food and waterborne illnesses are frequent, multidrug-resistant microbes are on the rise, and the needed drugs and vaccines have not been developed. The scientific approaches of the past-including the intense focus on individual genes and proteins typical of molecular biology-have not been sufficient to address these challenges. The first decade of the twenty-first century has seen remarkable innovations in technology and computational methods. These new tools provide nearly comprehensive views of complex biological systems and can provide a correspondingly deeper understanding of pathogen-host interactions. To take full advantage of these innovations, the National Institute of Allergy and Infectious Diseases recently initiated the Systems Biology Program for Infectious Disease Research. As participants of the Systems Biology Program, we think that the time is at hand to redefine the pathogen-host research paradigm.


Asunto(s)
Enfermedades Transmisibles , Interacciones Huésped-Patógeno , Biología de Sistemas/métodos , Enfermedades Transmisibles/genética , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA