Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Open Forum Infect Dis ; 10(3): ofad095, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36949873

RESUMO

Background: The ongoing circulation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a diagnostic challenge because symptoms of coronavirus disease 2019 (COVID-19) are difficult to distinguish from other respiratory diseases. Our goal was to use statistical analyses and machine learning to identify biomarkers that distinguish patients with COVID-19 from patients with influenza. Methods: Cytokine levels were analyzed in plasma and serum samples from patients with influenza and COVID-19, which were collected as part of the Centers for Disease Control and Prevention's Hospitalized Adult Influenza Vaccine Effectiveness Network (inpatient network) and the US Flu Vaccine Effectiveness (outpatient network). Results: We determined that interleukin (IL)-10 family cytokines are significantly different between COVID-19 and influenza patients. The results suggest that the IL-10 family cytokines are a potential diagnostic biomarker to distinguish COVID-19 and influenza infection, especially for inpatients. We also demonstrate that cytokine combinations, consisting of up to 3 cytokines, can distinguish SARS-CoV-2 and influenza infection with high accuracy in both inpatient (area under the receiver operating characteristics curve [AUC] = 0.84) and outpatient (AUC = 0.81) groups, revealing another potential screening tool for SARS-CoV-2 infection. Conclusions: This study not only reveals prospective screening tools for COVID-19 infections that are independent of polymerase chain reaction testing or clinical condition, but it also emphasizes potential pathways involved in disease pathogenesis that act as potential targets for future mechanistic studies.

2.
Viruses ; 14(5)2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35632648

RESUMO

The timing and magnitude of the immune response (i.e., the immunodynamics) associated with the early innate immune response to viral infection display distinct trends across influenza A virus subtypes in vivo. Evidence shows that the timing of the type-I interferon response and the overall magnitude of immune cell infiltration are both correlated with more severe outcomes. However, the mechanisms driving the distinct immunodynamics between infections of different virus strains (strain-specific immunodynamics) remain unclear. Here, computational modeling and strain-specific immunologic data are used to identify the immune interactions that differ in mice infected with low-pathogenic H1N1 or high-pathogenic H5N1 influenza viruses. Computational exploration of free parameters between strains suggests that the production rate of interferon is the major driver of strain-specific immune responses observed in vivo, and points towards the relationship between the viral load and lung epithelial interferon production as the main source of variance between infection outcomes. A greater understanding of the contributors to strain-specific immunodynamics can be utilized in future efforts aimed at treatment development to improve clinical outcomes of high-pathogenic viral strains.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Virus da Influenza A Subtipo H5N1 , Influenza Humana , Interferon Tipo I , Animais , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Virus da Influenza A Subtipo H5N1/fisiologia , Camundongos , Replicação Viral
4.
PLoS Comput Biol ; 17(10): e1008874, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34695114

RESUMO

Respiratory viruses present major public health challenges, as evidenced by the 1918 Spanish Flu, the 1957 H2N2, 1968 H3N2, and 2009 H1N1 influenza pandemics, and the ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. Severe RNA virus respiratory infections often correlate with high viral load and excessive inflammation. Understanding the dynamics of the innate immune response and its manifestations at the cell and tissue levels is vital to understanding the mechanisms of immunopathology and to developing strain-independent treatments. Here, we present a novel spatialized multicellular computational model of RNA virus infection and the type-I interferon-mediated antiviral response that it induces within lung epithelial cells. The model is built using the CompuCell3D multicellular simulation environment and is parameterized using data from influenza virus-infected cell cultures. Consistent with experimental observations, it exhibits either linear radial growth of viral plaques or arrested plaque growth depending on the local concentration of type I interferons. The model suggests that modifying the activity of signaling molecules in the JAK/STAT pathway or altering the ratio of the diffusion lengths of interferon and virus in the cell culture could lead to plaque growth arrest. The dependence of plaque growth arrest on diffusion lengths highlights the importance of developing validated spatial models of cytokine signaling and the need for in vitro measurement of these diffusion coefficients. Sensitivity analyses under conditions leading to continuous or arrested plaque growth found that plaque growth is more sensitive to variations of most parameters and more likely to have identifiable model parameters when conditions lead to plaque arrest. This result suggests that cytokine assay measurements may be most informative under conditions leading to arrested plaque growth. The model is easy to extend to include SARS-CoV-2-specific mechanisms or to use as a component in models linking epithelial cell signaling to systemic immune models.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Interferons , Infecções por Vírus de RNA , Vírus de RNA , Replicação Viral , Células Cultivadas , Biologia Computacional , Células Epiteliais/imunologia , Humanos , Imunidade Inata/imunologia , Interferons/imunologia , Interferons/metabolismo , Pulmão/citologia , Pulmão/imunologia , Modelos Biológicos , Infecções por Vírus de RNA/imunologia , Infecções por Vírus de RNA/virologia , Vírus de RNA/imunologia , Vírus de RNA/fisiologia , Replicação Viral/imunologia , Replicação Viral/fisiologia
5.
Mol Syst Biol ; 17(10): e10387, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34664389

RESUMO

We need to effectively combine the knowledge from surging literature with complex datasets to propose mechanistic models of SARS-CoV-2 infection, improving data interpretation and predicting key targets of intervention. Here, we describe a large-scale community effort to build an open access, interoperable and computable repository of COVID-19 molecular mechanisms. The COVID-19 Disease Map (C19DMap) is a graphical, interactive representation of disease-relevant molecular mechanisms linking many knowledge sources. Notably, it is a computational resource for graph-based analyses and disease modelling. To this end, we established a framework of tools, platforms and guidelines necessary for a multifaceted community of biocurators, domain experts, bioinformaticians and computational biologists. The diagrams of the C19DMap, curated from the literature, are integrated with relevant interaction and text mining databases. We demonstrate the application of network analysis and modelling approaches by concrete examples to highlight new testable hypotheses. This framework helps to find signatures of SARS-CoV-2 predisposition, treatment response or prioritisation of drug candidates. Such an approach may help deal with new waves of COVID-19 or similar pandemics in the long-term perspective.


Assuntos
COVID-19/imunologia , Biologia Computacional/métodos , Bases de Dados Factuais , SARS-CoV-2/imunologia , Software , Antivirais/uso terapêutico , COVID-19/genética , COVID-19/virologia , Gráficos por Computador , Citocinas/genética , Citocinas/imunologia , Mineração de Dados/estatística & dados numéricos , Regulação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/virologia , Redes e Vias Metabólicas/genética , Redes e Vias Metabólicas/imunologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/virologia , Mapeamento de Interação de Proteínas , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/genética , SARS-CoV-2/patogenicidade , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Tratamento Farmacológico da COVID-19
6.
BMC Bioinformatics ; 22(1): 108, 2021 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-33663384

RESUMO

BACKGROUND: Knowledge on the molecular targets of diseases and drugs is crucial for elucidating disease pathogenesis and mechanism of action of drugs, and for driving drug discovery and treatment formulation. In this regard, high-throughput gene transcriptional profiling has become a leading technology, generating whole-genome data on the transcriptional alterations caused by diseases or drug compounds. However, identifying direct gene targets, especially in the background of indirect (downstream) effects, based on differential gene expressions is difficult due to the complexity of gene regulatory network governing the gene transcriptional processes. RESULTS: In this work, we developed a network analysis method, called DeltaNeTS+, for inferring direct gene targets of drugs and diseases from gene transcriptional profiles. DeltaNeTS+ uses a gene regulatory network model to identify direct perturbations to the transcription of genes using gene expression data. Importantly, DeltaNeTS+ is able to combine both steady-state and time-course expression profiles, as well as leverage information on the gene network structure. We demonstrated the power of DeltaNeTS+ in predicting gene targets using gene expression data in complex organisms, including Caenorhabditis elegans and human cell lines (T-cell and Calu-3). More specifically, in an application to time-course gene expression profiles of influenza A H1N1 (swine flu) and H5N1 (avian flu) infection, DeltaNeTS+ shed light on the key differences of dynamic cellular perturbations caused by the two influenza strains. CONCLUSION: DeltaNeTS+ is a powerful network analysis tool for inferring gene targets from gene expression profiles. As demonstrated in the case studies, by incorporating available information on gene network structure, DeltaNeTS+ produces accurate predictions of direct gene targets from a small sample size (~ 10 s). Integrating static and dynamic expression data with transcriptional network structure extracted from genomic information, as enabled by DeltaNeTS+, is crucial toward personalized medicine, where treatments can be tailored to individual patients. DeltaNeTS+ can be freely downloaded from http://www.github.com/cabsel/deltanetsplus .


Assuntos
Biologia Computacional/métodos , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Preparações Farmacêuticas , Algoritmos , Animais , Humanos , Vírus da Influenza A Subtipo H1N1 , Virus da Influenza A Subtipo H5N1
7.
Bioinformatics ; 37(10): 1428-1434, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33196784

RESUMO

MOTIVATION: The cGAS pathway is a component of the innate immune system responsible for the detection of pathogenic DNA and upregulation of interferon beta (IFNß). Experimental evidence shows that IFNß signaling occurs in highly heterogeneous cells and is stochastic in nature; however, the benefits of these attributes remain unclear. To investigate how stochasticity and heterogeneity affect IFNß production, an agent-based model is developed to simulate both DNA transfection and viral infection. RESULTS: We show that heterogeneity can enhance IFNß responses during infection. Furthermore, by varying the degree of IFNß stochasticity, we find that only a percentage of cells (20-30%) need to respond during infection. Going beyond this range provides no additional protection against cell death or reduction of viral load. Overall, these simulations suggest that heterogeneity and stochasticity are important for moderating immune potency while minimizing cell death during infection. AVAILABILITY AND IMPLEMENTATION: Model repository is available at: https://github.com/ImmuSystems-Lab/AgentBasedModel-cGASPathway. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Interferon beta , Nucleotidiltransferases , Células Epiteliais , Humanos , Interferon beta/genética , Nucleotidiltransferases/metabolismo , Transdução de Sinais , Análise de Sistemas
8.
Biophys J ; 119(11): 2290-2298, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33129831

RESUMO

Over 50% of drugs fail in stage 3 clinical trials, many because of a poor understanding of the drug's mechanisms of action (MoA). A better comprehension of drug MoA will significantly improve research and development (R&D). Current proposed algorithms, such as ProTINA and DeMAND, can be overly complex. Additionally, they are unable to predict whether the drug-induced gene expression or the topology of the networks used to model gene regulation primarily impacts accurate drug target inference. In this work, we evaluate how network and gene expression data affect ProTINA's accuracy. We find that network topology predominantly determines the accuracy of ProTINA's predictions. We further show that the size of an interaction network and/or selecting cell-specific networks has a limited effect on accuracy. We then demonstrate that a specific network topology measure, betweenness, can be used to improve drug target prediction. Based on these results, we create a new algorithm, TREAP, that combines betweenness values and adjusted p-values for target inference. TREAP offers an alternative approach to drug target inference and is advantageous because it is not computationally demanding, provides easy-to-interpret results, and is often more accurate at predicting drug targets than current state-of-the-art approaches.


Assuntos
Algoritmos , Preparações Farmacêuticas , Biologia Computacional , Regulação da Expressão Gênica , Redes Reguladoras de Genes
9.
Viruses ; 12(10)2020 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-32993136

RESUMO

In a short time, the COVID-19 pandemic has left the world with over 25 million cases and staggering death tolls that are still rising. Treatments for SARS-CoV-2 infection are desperately needed as there are currently no approved drug therapies. With limited knowledge of viral mechanisms, a network controllability method of prioritizing existing drugs for repurposing efforts is optimal for quickly moving through the drug approval pipeline using limited, available, virus-specific data. Based on network topology and controllability, 16 proteins involved in translation, cellular transport, cellular stress, and host immune response are predicted as regulators of the SARS-CoV-2 infected cell. Of the 16, eight are prioritized as possible drug targets where two, PVR and SCARB1, are previously unexplored. Known compounds targeting these genes are suggested for viral inhibition study. Prioritized proteins in agreement with previous analysis and viral inhibition studies verify the ability of network controllability to predict biologically relevant candidates.


Assuntos
Betacoronavirus/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Reposicionamento de Medicamentos/métodos , Pneumonia Viral/tratamento farmacológico , Betacoronavirus/isolamento & purificação , Betacoronavirus/fisiologia , COVID-19 , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Aprovação de Drogas , Sistemas de Liberação de Medicamentos , Interações Hospedeiro-Patógeno , Humanos , Pandemias , Pneumonia Viral/metabolismo , Pneumonia Viral/virologia , Mapas de Interação de Proteínas/efeitos dos fármacos , Receptores Virais/genética , Receptores Virais/metabolismo , SARS-CoV-2 , Receptores Depuradores Classe B/metabolismo , Integração Viral , Tratamento Farmacológico da COVID-19
10.
J Infect Dis ; 222(7): 1155-1164, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32433769

RESUMO

The avian influenza A(H7N9) virus has caused high mortality rates in humans, especially in the elderly; however, little is known about the mechanistic basis for this. In the current study, we used nonhuman primates to evaluate the effect of aging on the pathogenicity of A(H7N9) virus. We observed that A(H7N9) virus infection of aged animals (defined as age 20-26 years) caused more severe symptoms than infection of young animals (defined as age 2-3 years). In aged animals, lung inflammation was weak and virus infection was sustained. Although cytokine and chemokine expression in the lungs of most aged animals was lower than that in the lungs of young animals, 1 aged animal showed severe symptoms and dysregulated proinflammatory cytokine and chemokine production. These results suggest that attenuated or dysregulated immune responses in aged animals are responsible for the severe symptoms observed among elderly patients infected with A(H7N9) virus.


Assuntos
Envelhecimento , Subtipo H7N9 do Vírus da Influenza A , Pulmão/patologia , Infecções por Orthomyxoviridae/virologia , Animais , Citocinas/imunologia , Modelos Animais de Doenças , Feminino , Pulmão/imunologia , Pulmão/virologia , Macaca fascicularis , Infecções por Orthomyxoviridae/imunologia , Replicação Viral
11.
Int Immunol ; 32(8): 499-507, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32060507

RESUMO

Aluminum precipitates have long been used as adjuvants for human vaccines, but there is a clear need for safer and more effective adjuvants. Here we report in a mouse model that the psoriasis drug Oxarol ointment is a highly effective vaccine adjuvant. By applying Oxarol ointment onto skin, humoral responses and germinal center (GC) reactions were augmented, and the treated mice were protected from death caused by influenza virus infection. Keratinocyte-specific vitamin D3 receptor (Vdr) gene expression was required for these responses through induction of the thymic stromal lymphopoietin (Tslp) gene. Experiments involving administration of recombinant TSLP or, conversely, anti-TSLP antibody demonstrated that TSLP plays a key role in the GC reactions. Furthermore, cell-type-specific Tslpr gene deletion or diphtheria toxin-mediated deletion of specific cell types revealed that CD11c+ cells excluding Langerhans cells were responsible for the Oxarol-mediated GC reactions. These results indicate that active vitamin D3 is able to enhance the humoral response via Tslp induction in the skin and serves as a new vaccine adjuvant.


Assuntos
Calcitriol/análogos & derivados , Fármacos Dermatológicos/uso terapêutico , Vacinas contra Influenza/imunologia , Pomadas/uso terapêutico , Psoríase/terapia , Animais , Calcitriol/uso terapêutico , Reposicionamento de Medicamentos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Psoríase/imunologia
12.
BMC Bioinformatics ; 20(1): 297, 2019 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-31159726

RESUMO

BACKGROUND: Host factors of influenza virus replication are often found in key topological positions within protein-protein interaction networks. This work explores how protein states can be manipulated through controllability analysis: the determination of the minimum manipulation needed to drive the cell system to any desired state. Here, we complete a two-part controllability analysis of two protein networks: a host network representing the healthy cell state and an influenza A virus-host network representing the infected cell state. In this context, controllability analyses aim to identify key regulating host factors of the infected cell's progression. This knowledge can be utilized in further biological analysis to understand disease dynamics and isolate proteins for study as drug target candidates. RESULTS: Both topological and controllability analyses provide evidence of wide-reaching network effects stemming from the addition of viral-host protein interactions. Virus interacting and driver host proteins are significant both topologically and in controllability, therefore playing important roles in cell behavior during infection. Functional analysis finds overlap of results with previous siRNA studies of host factors involved in influenza replication, NF-kB pathway and infection relevance, and roles as interferon regulating genes. 24 proteins are identified as holding regulatory roles specific to the infected cell by measures of topology, controllability, and functional role. These proteins are recommended for further study as potential antiviral drug targets. CONCLUSIONS: Seasonal outbreaks of influenza A virus are a major cause of illness and death around the world each year with a constant threat of pandemic infection. This research aims to increase the efficiency of antiviral drug target discovery using existing protein-protein interaction data and network analysis methods. These results are beneficial to future studies of influenza virus, both experimental and computational, and provide evidence that the combination of topology and controllability analyses may be valuable for future efforts in drug target discovery.


Assuntos
Antivirais/farmacologia , Sistemas de Liberação de Medicamentos , Descoberta de Drogas , Interações Hospedeiro-Patógeno , Mapas de Interação de Proteínas , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/metabolismo , RNA Interferente Pequeno/metabolismo , Reprodutibilidade dos Testes , Replicação Viral/efeitos dos fármacos
13.
J Theor Biol ; 462: 148-157, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30395807

RESUMO

Cyclic GMP-AMP synthase (cGAS) has recently been identified as the primary protein that detects cytosolic double stranded DNA to invoke a type I interferon response. The cGAS pathway is vital in the recognition of DNA encoded viruses as well as self-DNA leaked from the nucleus of damaged cells. Currently, the dynamics regulating the cGAS pathway are poorly understood; limiting our knowledge of how DNA-induced immune responses are regulated. Using systems biology approaches, we formulated a mathematical model to describe the dynamics of this pathway and examine the resulting system-level emergent properties. Unknown model parameters were fit to data compiled from literature using a Parallel Tempering Markov Chain Monte Carlo (PT-MCMC) approach, resulting in an ensemble of parameterized models. A local sensitivity analysis demonstrated that parameter sensitivity trends across model ensembles were independent of the select parameterization. An in-silico knock-down of TREX1 found that the interferon response is highly robust, showing that complete inhibition is necessary to induce chemical conditions consistent with chronic inflammation. Lastly, we demonstrate that the model recapitulates interferon expression data resulting from small molecule inhibition of cGAS. Overall, the importance of this model is exhibited in its capacity to identify sensitive components of the cGAS pathway, generate testable hypotheses, and confirm experimental observations.


Assuntos
DNA/imunologia , Exodesoxirribonucleases/metabolismo , Modelos Teóricos , Nucleotidiltransferases/metabolismo , Fosfoproteínas/metabolismo , Animais , DNA Viral/imunologia , Retroalimentação , Humanos , Inflamação , Interferon Tipo I/metabolismo , Cadeias de Markov , Método de Monte Carlo , Biologia de Sistemas/métodos
14.
ALTEX ; 36(1): 91-102, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30332685

RESUMO

Current efforts in chemical safety are focused on utilizing human in vitro or alternative animal data in biological pathway context. However, it remains unclear how biological pathways, and toxicology data developed in that context, can be used to quantitatively facilitate decision-making.  The objective of this work is to determine if hypothesis testing using Adverse Outcome Pathways (AOPs) can provide quantitative chemical hazard predictions.  Current methods for predicting hazards of chemicals in a biological pathway context were extensively reviewed, specific case studies examined and computational modeling used to demonstrate quantitative hazard prediction based on an AOP. Since AOPs are chemically agnostic, we propose that AOPs function as hypotheses for how specific chemicals may cause adverse effects via specific pathways. Three broad approaches were identified for testing the hypothesis with AOPs, semi-quantitative weight of evidence, probabilistic, and mechanistic modeling. We then demonstrate how these approaches could be used to test hypotheses using high throughput in vitro data and alternative animal data. Finally, we discuss standards in development and documentation that would facilitate use in a regulatory context. We conclude that quantitative AOPs provide a flexible hypothesis framework for predicting chemical hazards. It accommodates a wide range of approaches that are useful at many stages and build upon one another to become increasingly quantitative.


Assuntos
Rotas de Resultados Adversos , Alternativas aos Testes com Animais , Simulação por Computador , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Substâncias Perigosas/toxicidade , Animais , Tomada de Decisões , Humanos , Projetos de Pesquisa , Medição de Risco
15.
mBio ; 9(6)2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30563907

RESUMO

The positions of host factors required for viral replication within a human protein-protein interaction (PPI) network can be exploited to identify drug targets that are robust to drug-mediated selective pressure. Host factors can physically interact with viral proteins, be a component of virus-regulated pathways (where proteins do not interact with viral proteins), or be required for viral replication but unregulated by viruses. Here, we demonstrate a method of combining human PPI networks with virus-host PPI data to improve antiviral drug discovery for influenza viruses by identifying target host proteins. Analysis shows that influenza virus proteins physically interact with host proteins in network positions significant for information flow, even after the removal of known abundance-degree bias within PPI data. We have isolated a subnetwork of the human PPI network that connects virus-interacting host proteins to host factors that are important for influenza virus replication without physically interacting with viral proteins. The subnetwork is enriched for signaling and immune processes distinct from those associated with virus-interacting proteins. Selecting proteins based on subnetwork topology, we performed an siRNA screen to determine whether the subnetwork was enriched for virus replication host factors and whether network position within the subnetwork offers an advantage in prioritization of drug targets to control influenza virus replication. We found that the subnetwork is highly enriched for target host proteins-more so than the set of host factors that physically interact with viral proteins. Our findings demonstrate that network positions are a powerful predictor to guide antiviral drug candidate prioritization.IMPORTANCE Integrating virus-host interactions with host protein-protein interactions, we have created a method using these established network practices to identify host factors (i.e., proteins) that are likely candidates for antiviral drug targeting. We demonstrate that interaction cascades between host proteins that directly interact with viral proteins and host factors that are important to influenza virus replication are enriched for signaling and immune processes. Additionally, we show that host proteins that interact with viral proteins are in network locations of power. Finally, we demonstrate a new network methodology to predict novel host factors and validate predictions with an siRNA screen. Our results show that integrating virus-host proteins interactions is useful in the identification of antiviral drug target candidates.


Assuntos
Interações Hospedeiro-Patógeno/genética , Orthomyxoviridae/fisiologia , Mapas de Interação de Proteínas , Replicação Viral , Linhagem Celular , Sistemas de Liberação de Medicamentos , Descoberta de Drogas , Humanos , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Ligação Proteica , Transporte Proteico , RNA Interferente Pequeno , Proteínas Virais/metabolismo
16.
Nucleic Acids Res ; 46(6): e34, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29325153

RESUMO

Genome-wide transcriptional profiling provides a global view of cellular state and how this state changes under different treatments (e.g. drugs) or conditions (e.g. healthy and diseased). Here, we present ProTINA (Protein Target Inference by Network Analysis), a network perturbation analysis method for inferring protein targets of compounds from gene transcriptional profiles. ProTINA uses a dynamic model of the cell-type specific protein-gene transcriptional regulation to infer network perturbations from steady state and time-series differential gene expression profiles. A candidate protein target is scored based on the gene network's dysregulation, including enhancement and attenuation of transcriptional regulatory activity of the protein on its downstream genes, caused by drug treatments. For benchmark datasets from three drug treatment studies, ProTINA was able to provide highly accurate protein target predictions and to reveal the mechanism of action of compounds with high sensitivity and specificity. Further, an application of ProTINA to gene expression profiles of influenza A viral infection led to new insights of the early events in the infection.


Assuntos
Algoritmos , Biologia Computacional/métodos , Redes Reguladoras de Genes , Influenza Humana/genética , Mapas de Interação de Proteínas/genética , Transcriptoma , Antivirais/farmacologia , Linhagem Celular Tumoral , Perfilação da Expressão Gênica/métodos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/fisiologia , Influenza Humana/virologia
17.
Antimicrob Agents Chemother ; 60(3): 1902-6, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26711748

RESUMO

New strategies to develop novel broad-spectrum antiviral drugs against influenza virus infections are needed due to the emergence of antigenic variants and drug-resistant viruses. Here, we evaluated C646, a novel p300/CREB-binding protein-specific inhibitor of histone acetyltransferase (HAT), as an anti-influenza virus agent in vitro and in vivo and explored how C646 affects the viral life cycle and host response. Our studies highlight the value of targeting HAT activity for anti-influenza drug development.


Assuntos
Antivirais/farmacologia , Benzoatos/farmacologia , Proteína de Ligação a CREB/antagonistas & inibidores , Proteína p300 Associada a E1A/antagonistas & inibidores , Histona Acetiltransferases/antagonistas & inibidores , Vírus da Influenza A/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , Pirazóis/farmacologia , Animais , Proteína de Ligação a CREB/metabolismo , Linhagem Celular , Cães , Proteína p300 Associada a E1A/metabolismo , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Nitrobenzenos , Pirazolonas
18.
Front Pharmacol ; 6: 186, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26388775

RESUMO

Identifying promising compounds during the early stages of drug development is a major challenge for both academia and the pharmaceutical industry. The difficulties are even more pronounced when we consider multi-target pharmacology, where the compounds often target more than one protein, or multiple compounds are used together. Here, we address this problem by using machine learning and network analysis to process sequence and interaction data from human proteins to identify promising compounds. We used this strategy to identify properties that make certain proteins more likely to cause harmful effects when targeted; such proteins usually have domains commonly found throughout the human proteome. Additionally, since currently marketed drugs hit multiple targets simultaneously, we combined the information from individual proteins to devise a score that quantifies the likelihood of a compound being harmful to humans. This approach enabled us to distinguish between approved and problematic drugs with an accuracy of 60-70%. Moreover, our approach can be applied as soon as candidate drugs are available, as demonstrated with predictions for more than 5000 experimental drugs. These resources are available at http://sourceforge.net/projects/psin/.

19.
PLoS Pathog ; 11(6): e1004856, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26046528

RESUMO

Influenza viruses present major challenges to public health, evident by the 2009 influenza pandemic. Highly pathogenic influenza virus infections generally coincide with early, high levels of inflammatory cytokines that some studies have suggested may be regulated in a strain-dependent manner. However, a comprehensive characterization of the complex dynamics of the inflammatory response induced by virulent influenza strains is lacking. Here, we applied gene co-expression and nonlinear regression analysis to time-course, microarray data developed from influenza-infected mouse lung to create mathematical models of the host inflammatory response. We found that the dynamics of inflammation-associated gene expression are regulated by an ultrasensitive-like mechanism in which low levels of virus induce minimal gene expression but expression is strongly induced once a threshold virus titer is exceeded. Cytokine assays confirmed that the production of several key inflammatory cytokines, such as interleukin 6 and monocyte chemotactic protein 1, exhibit ultrasensitive behavior. A systematic exploration of the pathways regulating the inflammatory-associated gene response suggests that the molecular origins of this ultrasensitive response mechanism lie within the branch of the Toll-like receptor pathway that regulates STAT1 phosphorylation. This study provides the first evidence of an ultrasensitive mechanism regulating influenza virus-induced inflammation in whole lungs and provides insight into how different virus strains can induce distinct temporal inflammation response profiles. The approach developed here should facilitate the construction of gene regulatory models of other infectious diseases.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Infecções por Orthomyxoviridae/imunologia , Animais , Western Blotting , Feminino , Citometria de Fluxo , Inflamação/genética , Inflamação/imunologia , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Infecções por Orthomyxoviridae/genética , Transcriptoma , Virulência
20.
Nat Commun ; 6: 6600, 2015 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-25807527

RESUMO

Seasonal influenza A viruses cause annual epidemics of respiratory disease; highly pathogenic avian H5N1 and the recently emerged H7N9 viruses cause severe infections in humans, often with fatal outcomes. Although numerous studies have addressed the pathogenicity of influenza viruses, influenza pathogenesis remains incompletely understood. Here we generate influenza viruses expressing fluorescent proteins of different colours ('Color-flu' viruses) to facilitate the study of viral infection in in vivo models. On adaptation to mice, stable expression of the fluorescent proteins in infected animals allows their detection by different types of microscopy and by flow cytometry. We use this system to analyse the progression of viral spread in mouse lungs, for live imaging of virus-infected cells, and for differential gene expression studies in virus antigen-positive and virus antigen-negative live cells in the lungs of Color-flu-infected mice. Collectively, Color-flu viruses are powerful tools to analyse virus infections at the cellular level in vivo to better understand influenza pathogenesis.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Fluorescência Verde/genética , Vírus da Influenza A Subtipo H1N1/genética , Proteínas Luminescentes/genética , Pulmão/virologia , Infecções por Orthomyxoviridae , Proteínas não Estruturais Virais/genética , Animais , Fusão Gênica Artificial , Genes Reporter , Camundongos , Camundongos Endogâmicos C57BL , Replicação Viral , Proteína Vermelha Fluorescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA