Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 480(7375): 113-7, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22020285

RESUMO

Selective autophagy involves the recognition and targeting of specific cargo, such as damaged organelles, misfolded proteins, or invading pathogens for lysosomal destruction. Yeast genetic screens have identified proteins required for different forms of selective autophagy, including cytoplasm-to-vacuole targeting, pexophagy and mitophagy, and mammalian genetic screens have identified proteins required for autophagy regulation. However, there have been no systematic approaches to identify molecular determinants of selective autophagy in mammalian cells. Here, to identify mammalian genes required for selective autophagy, we performed a high-content, image-based, genome-wide small interfering RNA screen to detect genes required for the colocalization of Sindbis virus capsid protein with autophagolysosomes. We identified 141 candidate genes required for viral autophagy, which were enriched for cellular pathways related to messenger RNA processing, interferon signalling, vesicle trafficking, cytoskeletal motor function and metabolism. Ninety-six of these genes were also required for Parkin-mediated mitophagy, indicating that common molecular determinants may be involved in autophagic targeting of viral nucleocapsids and autophagic targeting of damaged mitochondria. Murine embryonic fibroblasts lacking one of these gene products, the C2-domain containing protein, SMURF1, are deficient in the autophagosomal targeting of Sindbis and herpes simplex viruses and in the clearance of damaged mitochondria. Moreover, SMURF1-deficient mice accumulate damaged mitochondria in the heart, brain and liver. Thus, our study identifies candidate determinants of selective autophagy, and defines SMURF1 as a newly recognized mediator of both viral autophagy and mitophagy.


Assuntos
Autofagia/genética , Estudo de Associação Genômica Ampla , RNA Interferente Pequeno/genética , Animais , Proteínas do Capsídeo/metabolismo , Células HeLa , Humanos , Lisossomos/metabolismo , Camundongos , Mitocôndrias/metabolismo , Transporte Proteico/genética , Sindbis virus/metabolismo , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética
2.
Nat Chem Biol ; 7(10): 712-9, 2011 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-21909097

RESUMO

A chemical genetics approach was taken to identify inhibitors of NS1, a major influenza A virus virulence factor that inhibits host gene expression. A high-throughput screen of 200,000 synthetic compounds identified small molecules that reversed NS1-mediated inhibition of host gene expression. A counterscreen for suppression of influenza virus cytotoxicity identified naphthalimides that inhibited replication of influenza virus and vesicular stomatitis virus (VSV). The mechanism of action occurs through activation of REDD1 expression and concomitant inhibition of mammalian target of rapamycin complex 1 (mTORC1) via TSC1-TSC2 complex. The antiviral activity of naphthalimides was abolished in REDD1(-/-) cells. Inhibition of REDD1 expression by viruses resulted in activation of the mTORC1 pathway. REDD1(-/-) cells prematurely upregulated viral proteins via mTORC1 activation and were permissive to virus replication. In contrast, cells conditionally expressing high concentrations of REDD1 downregulated the amount of viral protein. Thus, REDD1 is a new host defense factor, and chemical activation of REDD1 expression represents a potent antiviral intervention strategy.


Assuntos
Antivirais/farmacologia , Naftalimidas/farmacologia , Orthomyxoviridae/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Vesiculovirus/efeitos dos fármacos , Animais , Antivirais/química , Linhagem Celular , Cães , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Naftalimidas/química , Orthomyxoviridae/genética , Orthomyxoviridae/metabolismo , Relação Estrutura-Atividade , Fatores de Transcrição/deficiência , Vesiculovirus/genética , Vesiculovirus/metabolismo , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/efeitos dos fármacos
3.
Viruses ; 15(10)2023 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-37896896

RESUMO

HIV-1 latency is a major barrier to curing infections with antiretroviral therapy and, consequently, to eliminating the disease globally. The establishment, maintenance, and potential clearance of latent infection are complex dynamic processes and can be best described with the help of mathematical models followed by experimental validation. Here, we review the use of viral dynamics models for HIV-1, with a focus on applications to the latent reservoir. Such models have been used to explain the multi-phasic decay of viral load during antiretroviral therapy, the early seeding of the latent reservoir during acute infection and the limited inflow during treatment, the dynamics of viral blips, and the phenomenon of post-treatment control. Finally, we discuss that mathematical models have been used to predict the efficacy of potential HIV-1 cure strategies, such as latency-reversing agents, early treatment initiation, or gene therapies, and to provide guidance for designing trials of these novel interventions.


Assuntos
Infecções por HIV , Soropositividade para HIV , HIV-1 , Humanos , HIV-1/genética , Latência Viral , Modelos Biológicos , Modelos Teóricos , Infecções por HIV/tratamento farmacológico , Linfócitos T CD4-Positivos
4.
FEBS Lett ; 597(10): 1384-1402, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36951513

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic has affected tens of millions of individuals and caused hundreds of thousands of deaths worldwide. Here, we present a comprehensive, multiscale network analysis of the transcriptional response to the virus. In particular, we focused on key regulators, cell receptors, and host processes that were hijacked by the virus for its advantage. ACE2-controlled processes involved CD300e (a TYROBP receptor) as a key regulator and the activation of IL-2 pro-inflammatory cytokine signaling. We further investigated the age dependency of such receptors in different tissues. In summary, this study provides novel insights into the gene regulatory organization during the SARS-CoV-2 infection and the tissue-specific, age-dependent expression of the cell receptors involved in COVID-19.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Citocinas
5.
iScience ; 25(5): 104192, 2022 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-35479404

RESUMO

Sex differences in the pathogenesis of infectious diseases because of differential immune responses between females and males have been well-documented for multiple pathogens. However, the molecular mechanism underlying the observed sex differences in influenza virus infection remains poorly understood. In this study, we used a network-based approach to characterize the blood transcriptome collected over the course of infection with influenza A virus from female and male ferrets to dissect sex-biased gene expression. We identified significant differences in the temporal dynamics and regulation of immune responses between females and males. Our results elucidate sex-differentiated pathways involved in the unfolded protein response (UPR), lipid metabolism, and inflammatory responses, including a female-biased IRE1/XBP1 activation and male-biased crosstalk between metabolic reprogramming and IL-1 and AP-1 pathways. Overall, our study provides molecular insights into sex differences in transcriptional regulation of immune responses and contributes to a better understanding of sex biases in influenza pathogenesis.

6.
Front Immunol ; 13: 840457, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35273617

RESUMO

Costimulation pathways play an essential role in T cell activation, differentiation, and regulation. CD155 expressed on antigen-presenting cells (APCs) interacts with TIGIT, an inhibitory costimulatory molecule, and CD226, an activating costimulatory molecule, on T cells. TIGIT and CD226 are expressed at varying levels depending on the T cell subset and activation state. T follicular helper cells in germinal centers (GC-Tfh) in human tonsils express high TIGIT and low CD226. However, the biological role of the CD155/TIGIT/CD226 axis in human Tfh cell biology has not been elucidated. To address this, we analyzed tonsillar CD4+ T cell subsets cultured with artificial APCs constitutively expressing CD155. Here we show that CD226 signals promote the early phase of Tfh cell differentiation in humans. CD155 signals promoted the proliferation of naïve CD4+ T cells and Tfh precursors (pre-Tfh) isolated from human tonsils and upregulated multiple Tfh molecules and decreased IL-2, a cytokine detrimental for Tfh cell differentiation. Blocking CD226 potently inhibited their proliferation and expression of Tfh markers. By contrast, while CD155 signals promoted the proliferation of tonsillar GC-Tfh cells, their proliferation required only weak CD226 signals. Furthermore, attenuating CD226 signals rather increased the expression of CXCR5, ICOS, and IL-21 by CD155-stimulated GC-Tfh cells. Thus, the importance of CD226 signals changes according to the differentiation stage of human Tfh cells and wanes in mature GC-Tfh cells. High TIGIT expression on GC-Tfh may play a role in attenuating the detrimental CD226 signals post GC-Tfh cell maturation.


Assuntos
Antígenos de Diferenciação de Linfócitos T , Receptores Imunológicos , Células T Auxiliares Foliculares , Antígenos de Diferenciação de Linfócitos T/metabolismo , Diferenciação Celular , Humanos , Ativação Linfocitária , Receptores Imunológicos/metabolismo , Subpopulações de Linfócitos T
7.
Viruses ; 14(11)2022 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-36366544

RESUMO

Seasonal influenza is a primary public health burden in the USA and globally. Annual vaccination programs are designed on the basis of circulating influenza viral strains. However, the effectiveness of the seasonal influenza vaccine is highly variable between seasons and among individuals. A number of factors are known to influence vaccination effectiveness including age, sex, and comorbidities. Here, we sought to determine whether whole blood gene expression profiling prior to vaccination is informative about pre-existing immunological status and the immunological response to vaccine. We performed whole transcriptome analysis using RNA sequencing (RNAseq) of whole blood samples obtained prior to vaccination from 275 participants enrolled in an annual influenza vaccine trial. Serological status prior to vaccination and 28 days following vaccination was assessed using the hemagglutination inhibition assay (HAI) to define baseline immune status and the response to vaccination. We find evidence that genes with immunological functions are increased in expression in individuals with higher pre-existing immunity and in those individuals who mount a greater response to vaccination. Using a random forest model, we find that this set of genes can be used to predict vaccine response with a performance similar to a model that incorporates physiological and prior vaccination status alone. A model using both gene expression and physiological factors has the greatest predictive power demonstrating the potential utility of molecular profiling for enhancing prediction of vaccine response. Moreover, expression of genes that are associated with enhanced vaccination response may point to additional biological pathways that contribute to mounting a robust immunological response to the seasonal influenza vaccine.


Assuntos
Vacinas contra Influenza , Influenza Humana , Humanos , Vacinas contra Influenza/genética , Influenza Humana/prevenção & controle , Índice de Massa Corporal , Anticorpos Antivirais , Vacinação , Testes de Inibição da Hemaglutinação , Estações do Ano , Expressão Gênica
8.
Sci Adv ; 8(40): eabm5859, 2022 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-36197970

RESUMO

Molecular responses to influenza A virus (IAV) infections vary between mammalian species. To identify conserved and species-specific molecular responses, we perform a comparative study of transcriptomic data derived from blood cells, primary epithelial cells, and lung tissues collected from IAV-infected humans, ferrets, and mice. The molecular responses in the human host have unique functions such as antigen processing that are not observed in mice or ferrets. Highly conserved gene coexpression modules across the three species are enriched for IAV infection-induced pathways including cell cycle and interferon (IFN) signaling. TDRD7 is predicted as an IFN-inducible host factor that is up-regulated upon IAV infection in the three species. TDRD7 is required for antiviral IFN response, potentially modulating IFN signaling via the JAK/STAT/IRF9 pathway. Identification of the common and species-specific molecular signatures, networks, and regulators of IAV infection provides insights into host-defense mechanisms and will facilitate the development of novel therapeutic interventions against IAV infection.


Assuntos
Doenças Transmissíveis , Vírus da Influenza A , Influenza Humana , Infecções por Orthomyxoviridae , Animais , Antivirais , Furões/metabolismo , Humanos , Vírus da Influenza A/fisiologia , Influenza Humana/genética , Interferons/metabolismo , Camundongos , Infecções por Orthomyxoviridae/genética , Ribonucleoproteínas
9.
Sci Adv ; 7(41): eabj1249, 2021 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-34623911

RESUMO

Memory T follicular helper (TFH) cells play an essential role to induce secondary antibody response by providing help to memory and naïve B cells. Here, we show that the transcription factor Tox2 is vital for the maintenance of TFH cells in germinal centers (GCs) and the generation of memory TFH cells. High Tox2 expression was almost exclusive to GC TFH cells among human tonsillar and blood CD4+ T cell subsets. Tox2 overexpression maintained the expression of TFH-associated genes in T cell receptor­stimulated human GC TFH cells and inhibited their spontaneous conversion into TH1-like cells. Tox2-deficient mice displayed impaired secondary TFH cell expansion upon reimmunization with an antigen and upon secondary infection with a heterologous influenza virus. Collectively, our study shows that Tox2 is highly integrated into establishment of durable GC TFH cell responses and development of memory TFH cells in mice and humans.

10.
Nat Microbiol ; 6(10): 1245-1258, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34465900

RESUMO

Respiratory failure is associated with increased mortality in COVID-19 patients. There are no validated lower airway biomarkers to predict clinical outcome. We investigated whether bacterial respiratory infections were associated with poor clinical outcome of COVID-19 in a prospective, observational cohort of 589 critically ill adults, all of whom required mechanical ventilation. For a subset of 142 patients who underwent bronchoscopy, we quantified SARS-CoV-2 viral load, analysed the lower respiratory tract microbiome using metagenomics and metatranscriptomics and profiled the host immune response. Acquisition of a hospital-acquired respiratory pathogen was not associated with fatal outcome. Poor clinical outcome was associated with lower airway enrichment with an oral commensal (Mycoplasma salivarium). Increased SARS-CoV-2 abundance, low anti-SARS-CoV-2 antibody response and a distinct host transcriptome profile of the lower airways were most predictive of mortality. Our data provide evidence that secondary respiratory infections do not drive mortality in COVID-19 and clinical management strategies should prioritize reducing viral replication and maximizing host responses to SARS-CoV-2.


Assuntos
Líquido da Lavagem Broncoalveolar/microbiologia , COVID-19/terapia , Respiração Artificial , SARS-CoV-2/patogenicidade , Imunidade Adaptativa , Adulto , Idoso , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Carga Bacteriana , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/virologia , COVID-19/imunologia , COVID-19/microbiologia , COVID-19/mortalidade , Estado Terminal , Feminino , Hospitalização , Humanos , Imunidade Inata , Masculino , Microbiota , Pessoa de Meia-Idade , Razão de Chances , Prognóstico , Estudos Prospectivos , Sistema Respiratório/imunologia , Sistema Respiratório/microbiologia , Sistema Respiratório/virologia , SARS-CoV-2/imunologia , Carga Viral
11.
medRxiv ; 2021 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-33655261

RESUMO

Mortality among patients with COVID-19 and respiratory failure is high and there are no known lower airway biomarkers that predict clinical outcome. We investigated whether bacterial respiratory infections and viral load were associated with poor clinical outcome and host immune tone. We obtained bacterial and fungal culture data from 589 critically ill subjects with COVID-19 requiring mechanical ventilation. On a subset of the subjects that underwent bronchoscopy, we also quantified SARS-CoV-2 viral load, analyzed the microbiome of the lower airways by metagenome and metatranscriptome analyses and profiled the host immune response. We found that isolation of a hospital-acquired respiratory pathogen was not associated with fatal outcome. However, poor clinical outcome was associated with enrichment of the lower airway microbiota with an oral commensal ( Mycoplasma salivarium ), while high SARS-CoV-2 viral burden, poor anti-SARS-CoV-2 antibody response, together with a unique host transcriptome profile of the lower airways were most predictive of mortality. Collectively, these data support the hypothesis that 1) the extent of viral infectivity drives mortality in severe COVID-19, and therefore 2) clinical management strategies targeting viral replication and host responses to SARS-CoV-2 should be prioritized.

12.
BMC Bioinformatics ; 11: 170, 2010 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-20370926

RESUMO

BACKGROUND: The recent emergence of the H5N1 influenza virus from avian reservoirs has raised concern about future influenza strains of high virulence emerging that could easily infect humans. We analyzed differential gene expression of lung epithelial cells to compare the response to H5N1 infection with a more benign infection with Respiratory Syncytial Virus (RSV). These gene expression data are then used as seeds to find important nodes by using a novel combination of the Gene Ontology database and the Human Network of gene interactions. Additional analysis of the data is conducted by training support vector machines (SVM) with the data and examining the orientations of the optimal hyperplanes generated. RESULTS: Analysis of gene clustering in the Gene Ontology shows no significant clustering of genes unique to H5N1 response at 8 hours post infection. At 24 hours post infection, however, a number of significant gene clusters are found for nodes representing "immune response" and "response to virus" terms. There were no significant clusters of genes in the Gene Ontology for the control (Mock) or RSV experiments that were unique relative to the H5N1 response. The genes found to be most important in distinguishing H5N1 infected cells from the controls using SVM showed a large degree of overlap with the list of significantly regulated genes. However, though none of these genes were members of the GO clusters found to be significant. CONCLUSIONS: Characteristics of H5N1 infection compared to RSV infection show several immune response factors that are specific for each of these infections. These include faster timescales within the cell as well as a more focused activation of immunity factors. Many of the genes that are found to be significantly expressed in H5N1 response relative to the control experiments are not found to cluster significantly in the Gene Ontology. These genes are, however, often closely linked to the clustered genes through the Human Network. This may suggest the need for more diverse annotations of these genes and verification of their action in immune response.


Assuntos
Células Epiteliais/virologia , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Virus da Influenza A Subtipo H5N1/fisiologia , Pulmão/virologia , Animais , Células Epiteliais/imunologia , Humanos , Pulmão/citologia , Vírus Sinciciais Respiratórios/fisiologia
13.
Microbiome ; 8(1): 39, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32178738

RESUMO

BACKGROUND: The abundance and diversity of antibiotic resistance genes (ARGs) in the human respiratory microbiome remain poorly characterized. In the context of influenza virus infection, interactions between the virus, the host, and resident bacteria with pathogenic potential are known to complicate and worsen disease, resulting in coinfection and increased morbidity and mortality of infected individuals. When pathogenic bacteria acquire antibiotic resistance, they are more difficult to treat and of global health concern. Characterization of ARG expression in the upper respiratory tract could help better understand the role antibiotic resistance plays in the pathogenesis of influenza-associated bacterial secondary infection. RESULTS: Thirty-seven individuals participating in the Household Influenza Transmission Study (HITS) in Managua, Nicaragua, were selected for this study. We performed metatranscriptomics and 16S rRNA gene sequencing analyses on nasal and throat swab samples, and host transcriptome profiling on blood samples. Individuals clustered into two groups based on their microbial gene expression profiles, with several microbial pathways enriched with genes differentially expressed between groups. We also analyzed antibiotic resistance gene expression and determined that approximately 25% of the sequence reads that corresponded to antibiotic resistance genes mapped to Streptococcus pneumoniae and Staphylococcus aureus. Following construction of an integrated network of ARG expression with host gene co-expression, we identified several host key regulators involved in the host response to influenza virus and bacterial infections, and host gene pathways associated with specific antibiotic resistance genes. CONCLUSIONS: This study indicates the host response to influenza infection could indirectly affect antibiotic resistance gene expression in the respiratory tract by impacting the microbial community structure and overall microbial gene expression. Interactions between the host systemic responses to influenza infection and antibiotic resistance gene expression highlight the importance of viral-bacterial co-infection in acute respiratory infections like influenza. Video abstract.


Assuntos
Bactérias/efeitos dos fármacos , Interações entre Hospedeiro e Microrganismos , Influenza Humana/microbiologia , Microbiota , Infecções Respiratórias/virologia , Adolescente , Adulto , Bactérias/genética , Bactérias/patogenicidade , Criança , Pré-Escolar , Coinfecção/microbiologia , Coinfecção/fisiopatologia , Coinfecção/virologia , Farmacorresistência Bacteriana/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Influenza Humana/fisiopatologia , Masculino , Nicarágua , RNA Ribossômico 16S/genética , Staphylococcus aureus/genética , Streptococcus pneumoniae/genética , Adulto Jovem
14.
mBio ; 11(1)2020 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-31937643

RESUMO

Virus and host factors contribute to cell-to-cell variation in viral infections and determine the outcome of the overall infection. However, the extent of the variability at the single-cell level and how it impacts virus-host interactions at a system level are not well understood. To characterize the dynamics of viral transcription and host responses, we used single-cell RNA sequencing to quantify at multiple time points the host and viral transcriptomes of human A549 cells and primary bronchial epithelial cells infected with influenza A virus. We observed substantial variability in viral transcription between cells, including the accumulation of defective viral genomes (DVGs) that impact viral replication. We show (i) a correlation between DVGs and virus-induced variation of the host transcriptional program and (ii) an association between differential inductions of innate immune response genes and attenuated viral transcription in subpopulations of cells. These observations at the single-cell level improve our understanding of the complex virus-host interplay during influenza virus infection.IMPORTANCE Defective influenza virus particles generated during viral replication carry incomplete viral genomes and can interfere with the replication of competent viruses. These defective genomes are thought to modulate the disease severity and pathogenicity of an influenza virus infection. Different defective viral genomes also introduce another source of variation across a heterogeneous cell population. Evaluating the impact of defective virus genomes on host cell responses cannot be fully resolved at the population level, requiring single-cell transcriptional profiling. Here, we characterized virus and host transcriptomes in individual influenza virus-infected cells, including those of defective viruses that arise during influenza A virus infection. We established an association between defective virus transcription and host responses and validated interfering and immunostimulatory functions of identified dominant defective viral genome species in vitro This study demonstrates the intricate effects of defective viral genomes on host transcriptional responses and highlights the importance of capturing host-virus interactions at the single-cell level.


Assuntos
Vírus Defeituosos/genética , Células Epiteliais/virologia , Perfilação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos/imunologia , Células A549 , Brônquios/citologia , Brônquios/virologia , Células Cultivadas , Vírus Defeituosos/imunologia , Genoma Viral , Humanos , Vírus da Influenza A/fisiologia , RNA Viral/genética , Análise de Sequência de RNA , Análise de Célula Única , Replicação Viral
15.
Cell Rep ; 27(1): 154-171.e9, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30943398

RESUMO

Transcriptional circuit architectures in several organisms have been evolutionarily selected to dictate precise given responses. Unlike these cellular systems, HIV is regulated through a complex circuit composed of two successive phases (host and viral), which create a positive feedback loop facilitating viral replication. However, it has long remained unclear whether both phases operate identically and to what extent the host phase influences the entire circuit. Here, we report that, although the host phase is regulated by a checkpoint whereby KAP1 mediates transcription activation, the virus evolved a minimalist system bypassing KAP1. Given the complex circuit's architecture, cell-to-cell KAP1 fluctuations impart heterogeneity in the host transcriptional responses, thus affecting the feedback loop. Mathematical modeling of a complete circuit reveals how these oscillations ultimately influence homogeneous reactivation potential of a latent virus. Thus, although HIV drives molecular innovation to fuel robust gene activation, it experiences transcriptional fragility, thereby influencing viral fate and cure efforts.


Assuntos
Redes Reguladoras de Genes/fisiologia , Infecções por HIV/virologia , HIV-1/genética , Provírus , Ativação Viral/genética , Latência Viral/genética , Células Cultivadas , Regulação Viral da Expressão Gênica , Genoma Viral , Instabilidade Genômica/fisiologia , Células HEK293 , Infecções por HIV/genética , HIV-1/fisiologia , Humanos , Células Jurkat , Provírus/genética , Provírus/fisiologia , Transcrição Gênica , Replicação Viral/genética
16.
Cell Rep ; 29(12): 3997-4009.e5, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31851929

RESUMO

Influenza A viruses (IAVs) have a remarkable tropism in their ability to circulate in both mammalian and avian species. The IAV NS1 protein is a multifunctional virulence factor that inhibits the type I interferon host response through a myriad of mechanisms. How NS1 has evolved to enable this remarkable property across species and its specific impact in the overall replication, pathogenicity, and host preference remain unknown. Here we analyze the NS1 evolutionary landscape and host tropism using a barcoded library of recombinant IAVs. Results show a surprisingly great variety of NS1 phenotypes according to their ability to replicate in different hosts. The IAV NS1 genes appear to have taken diverse and random evolutionary pathways within their multiple phylogenetic lineages. In summary, the high evolutionary plasticity of this viral protein underscores the ability of IAVs to adapt to multiple hosts and aids in our understanding of its global prevalence.


Assuntos
Especificidade de Hospedeiro/genética , Interações Hospedeiro-Patógeno/genética , Vírus da Influenza A/patogenicidade , Mutação , Infecções por Orthomyxoviridae/virologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Cães , Feminino , Imunidade Inata , Vírus da Influenza A/genética , Células Madin Darby de Rim Canino , Camundongos , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/patologia , Filogenia , Proteínas não Estruturais Virais/genética
17.
J Comput Biol ; 15(2): 185-94, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18312149

RESUMO

In order to understand the behavior of a gene regulatory network, it is essential to know the genes that belong to it. Identifying the correct members (e.g., in order to build a model) is a difficult task even for small subnetworks. Usually only few members of a network are known and one needs to guess the missing members based on experience or informed speculation. It is beneficial if one can additionally rely on experimental data to support this guess. In this work we present a new method based on formal concept analysis to detect unknown members of a gene regulatory network from gene expression time series data. We show that formal concept analysis is able to find a list of candidate genes for inclusion into a partially known basic network. This list can then be reduced by a statistical analysis so that the resulting genes interact strongly with the basic network and therefore should be included when modeling the network. The method has been applied to the DNA repair system of Mycobacterium tuberculosis. In this application, our method produces comparable results to an already existing method of component selection while it is applicable to a broader range of problems.


Assuntos
Biologia Computacional/métodos , Redes Reguladoras de Genes/genética , Modelos Estatísticos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Reparo do DNA , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Análise de Sequência de DNA
18.
Mol Biol Cell ; 16(1): 396-404, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15525680

RESUMO

Trends in increased tuberculosis infection and a fatality rate of approximately 23% have necessitated the search for alternative biomarkers using newly developed postgenomic approaches. Here we provide a systematic analysis of Mycobacterium tuberculosis (Mtb) by directly profiling its gene products. This analysis combines high-throughput proteomics and computational approaches to elucidate the globally expressed complements of the three subcellular compartments (the cell wall, membrane, and cytosol) of Mtb. We report the identifications of 1044 proteins and their corresponding localizations in these compartments. Genome-based computational and metabolic pathways analyses were performed and integrated with proteomics data to reconstruct response networks. From the reconstructed response networks for fatty acid degradation and lipid biosynthesis pathways in Mtb, we identified proteins whose involvements in these pathways were not previously suspected. Furthermore, the subcellular localizations of these expressed proteins provide interesting insights into the compartmentalization of these pathways, which appear to traverse from cell wall to cytoplasm. Results of this large-scale subcellular proteome profile of Mtb have confirmed and validated the computational network hypothesis that functionally related proteins work together in larger organizational structures.


Assuntos
Mycobacterium tuberculosis/fisiologia , Análise Serial de Proteínas/métodos , Automação , Membrana Celular/metabolismo , Parede Celular/metabolismo , Biologia Computacional , Citosol/metabolismo , Bases de Dados de Proteínas , Ácidos Graxos/metabolismo , Genoma , Metabolismo dos Lipídeos , Modelos Biológicos , Modelos Estatísticos , Proteínas/química , Proteoma , Proteômica , Software , Espectrometria de Massas por Ionização por Electrospray , Frações Subcelulares/metabolismo
19.
Bioinformatics ; 22(21): 2674-80, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16926218

RESUMO

MOTIVATION: A quantitative description of interactions between cell components is a major challenge in Computational Biology. As a method of choice, differential equations are used for this purpose, because they provide a detailed insight into the dynamic behavior of the system. In most cases, the number of time points of experimental time series is usually too small to estimate the parameters of a model of a whole gene regulatory network based on differential equations, such that one needs to focus on subnetworks consisting of only a few components. For most approaches, the set of components of the subsystem is given in advance and only the structure has to be estimated. However, the set of components that influence the system significantly are not always known in advance, making a method desirable that determines both, the components that are included into the model and the parameters. RESULTS: We have developed a method that uses gene expression data as well as interaction data between cell components to define a set of genes that we use for our modeling. In a subsequent step, we estimate the parameters of our model of piecewise linear differential equations and evaluate the results simulating the behavior of the system with our model. We have applied our method to the DNA repair system of Mycobacterium tuberculosis. Our analysis predicts that the gene Rv2719c plays an important role in this system.


Assuntos
Proteínas de Bactérias/metabolismo , Reparo do DNA/fisiologia , Perfilação da Expressão Gênica/métodos , Modelos Biológicos , Mycobacterium tuberculosis/fisiologia , Mapeamento de Interação de Proteínas/métodos , Transdução de Sinais/fisiologia , Algoritmos , Simulação por Computador , Regulação da Expressão Gênica/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos/métodos
20.
NPJ Syst Biol Appl ; 3: 35, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29214055

RESUMO

Influenza A virus, with the limited coding capacity of 10-14 proteins, requires the host cellular machinery for many aspects of its life cycle. Knowledge of these host cell requirements not only reveals molecular pathways exploited by the virus or triggered by the immune system, but also provides further targets for antiviral drug development. To uncover novel pathways and key targets of influenza infection, we assembled a large amount of data from 12 cell-based gene-expression studies of influenza infection for an integrative network analysis. We systematically identified differentially expressed genes and gene co-expression networks induced by influenza infection. We revealed the dedicator of cytokinesis 5 (DOCK5) played potentially an important role for influenza virus replication. CRISPR/Cas9 knockout of DOCK5 reduced influenza virus replication, indicating that DOCK5 is a key regulator for the viral life cycle. DOCK5's targets determined by the DOCK5 knockout experiments strongly validated the predicted gene signatures and networks. This study systematically uncovered and validated fundamental patterns of molecular responses, intrinsic structures of gene co-regulation, and novel key targets in influenza virus infection.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA