Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Chem Biol ; 30(11): 1377-1389.e8, 2023 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-37586370

RESUMO

TruAB Discovery is an approach that integrates cellular immunology, high-throughput immunosequencing, bioinformatics, and computational biology in order to discover naturally occurring human antibodies for prophylactic or therapeutic use. We adapted our previously described pairSEQ technology to pair B cell receptor heavy and light chains of SARS-CoV-2 spike protein-binding antibodies derived from enriched antigen-specific memory B cells and bulk antibody-secreting cells. We identified approximately 60,000 productive, in-frame, paired antibody sequences, from which 2,093 antibodies were selected for functional evaluation based on abundance, isotype and patterns of somatic hypermutation. The exceptionally diverse antibodies included RBD-binders with broad neutralizing activity against SARS-CoV-2 variants, and S2-binders with broad specificity against betacoronaviruses and the ability to block membrane fusion. A subset of these RBD- and S2-binding antibodies demonstrated robust protection against challenge in hamster and mouse models. This high-throughput approach can accelerate discovery of diverse, multifunctional antibodies against any target of interest.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Camundongos , Humanos , Anticorpos Neutralizantes , Anticorpos Amplamente Neutralizantes , Anticorpos Antivirais
2.
J Immunol ; 210(9): 1247-1256, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36939421

RESUMO

Retinoic acid-inducible gene I (RIG-I) is essential for activating host cell innate immunity to regulate the immune response against many RNA viruses. We previously identified that a small molecule compound, KIN1148, led to the activation of IFN regulatory factor 3 (IRF3) and served to enhance protection against influenza A virus (IAV) A/California/04/2009 infection. We have now determined direct binding of KIN1148 to RIG-I to drive expression of IFN regulatory factor 3 and NF-κB target genes, including specific immunomodulatory cytokines and chemokines. Intriguingly, KIN1148 does not lead to ATPase activity or compete with ATP for binding but activates RIG-I to induce antiviral gene expression programs distinct from type I IFN treatment. When administered in combination with a vaccine against IAV, KIN1148 induces both neutralizing Ab and IAV-specific T cell responses compared with vaccination alone, which induces comparatively poor responses. This robust KIN1148-adjuvanted immune response protects mice from lethal A/California/04/2009 and H5N1 IAV challenge. Importantly, KIN1148 also augments human CD8+ T cell activation. Thus, we have identified a small molecule RIG-I agonist that serves as an effective adjuvant in inducing noncanonical RIG-I activation for induction of innate immune programs that enhance adaptive immune protection of antiviral vaccination.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana , Humanos , Animais , Camundongos , Proteína DEAD-box 58/metabolismo , Virus da Influenza A Subtipo H5N1/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Adjuvantes Imunológicos , Antivirais/farmacologia , Imunidade Inata
3.
Disabil Rehabil Assist Technol ; 16(8): 821-830, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32189537

RESUMO

PURPOSE: Rates of prosthetic device abandonment are dramatically high; however, the reasons behind abandonment are less understood. A scoping review was conducted to explore the current state of the literature on why individuals abandon upper limb prosthetic devices and consider how these reasons have evolved historically. MATERIALS AND METHODS: A systematic search of the literature identified 123 articles. After reviewing the articles using predetermined inclusion and exclusion criteria, nine relevant articles were included in the final review. The included articles covered passive, body-powered and myoelectric prosthetic devices. RESULTS: Across time, reasons for abandonment could be broadly categorized into comfort and function. Weight, temperature and perspiration were among the most common and persistent comfort-related reasons for abandonment. Regarding function, studies-reported abandonment was attributed to key concerns about control and sensory feedback, whereby participants may feel more functional without their device. CONCLUSIONS: In agreement with the previous literature, lack of comfort and function remain persistent reasons for upper limb prosthesis abandonment. Up-to-date research on reasons for abandonment of upper limb prosthetic devices is lacking, and recent prosthesis advancements have not been included in studies of device use, adoption and abandonment. Therefore, future work should explore reasons for abandonment in contemporary upper limb prosthetic devices. By understanding the reasons for prosthetic device abandonment, clinicians, therapists and researchers can use this information to proactively mitigate future upper limb prosthetic device abandonment. Findings from this review can be used to guide future prosthetic device development to improve these areas of concern and satisfy user needs.IMPLICATIONS FOR REHABILITATIONBy understanding the reasons for prosthetic device abandonment, clinicians, therapists and researchers can use this information to proactively mitigate future upper limb prosthetic device abandonment.The findings from this review can be used to guide future prosthetic device development to improve areas of concern and satisfy user needs.


Assuntos
Membros Artificiais , Humanos , Desenho de Prótese , Extremidade Superior
4.
Sci Transl Med ; 11(519)2019 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-31748228

RESUMO

Activated CD4 T cells are a major target of HIV infection. Results from the STEP HIV vaccine trial highlighted a potential role for total activated CD4 T cells in promoting HIV acquisition. However, the influence of vaccine insert-specific CD4 T cell responses on HIV acquisition is not known. Here, using the data obtained from four macaque studies, we show that the DNA prime/modified vaccinia Ankara boost vaccine induced interferon γ (IFNγ+) CD4 T cells [T helper 1 (TH1) cells] rapidly migrate to multiple tissues including colon, cervix, and vaginal mucosa. These mucosal TH1 cells persisted at higher frequencies and expressed higher density of CCR5, a viral coreceptor, compared to cells in blood. After intravaginal or intrarectal simian immunodeficiency virus (SIV)/simian-human immunodeficiency virus (SHIV) challenges, strong vaccine protection was evident only in animals that had lower frequencies of vaccine-specific TH1 cells but not in animals that had higher frequencies of TH1 cells, despite comparable vaccine-induced humoral and CD8 T cell immunity in both groups. An RNA transcriptome signature in blood at 7 days after priming immunization from one study was associated with induction of fewer TH1-type CD4 cells and enhanced protection. These results demonstrate that high and persisting frequencies of HIV vaccine-induced TH1-biased CD4 T cells in the intestinal and genital mucosa can mitigate beneficial effects of protective antibodies and CD8 T cells, highlighting a critical role of priming immunization and vaccine adjuvants in modulating HIV vaccine efficacy.


Assuntos
Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Linfócitos T CD8-Positivos/imunologia , Colo/patologia , Feminino , Perfilação da Expressão Gênica , Interferon gama/metabolismo , Contagem de Linfócitos , Macaca mulatta , Masculino , Mucosa/patologia , Receptores CCR5/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Resultado do Tratamento , Vacinação , Vagina/imunologia , Vagina/virologia
5.
Nat Commun ; 10(1): 3649, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31409781

RESUMO

RIG-I-Like Receptors (RLRs) RIG-I, MDA5, and LGP2, are vital pathogen recognition receptors in the defense against RNA viruses. West Nile Virus (WNV) infections continue to grow in the US. Here, we use a systems biology approach to define the contributions of each RLR in the innate immune response to WNV. Genome-wide RNAseq and bioinformatics analyses of macrophages from mice lacking either RLR reveal that the RLRs drive distinct immune gene activation and response polarization to mediate an M1/inflammatory signature while suppressing the M2/wound healing phenotype. While LGP2 functions to modulate inflammatory signaling, RIG-I and MDA5 together are essential for M1 macrophage polarization in vivo and the control of WNV infection through potential downstream control of ATF4 and SMAD4 to regulate target gene expression for cell polarization. These analyses reveal the RLR-driven signature of macrophage polarization, innate immune protection, and immune programming against WNV infection.


Assuntos
Proteína DEAD-box 58/imunologia , Macrófagos/imunologia , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia , Animais , Polaridade Celular , Proteína DEAD-box 58/genética , Feminino , Humanos , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/imunologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Febre do Nilo Ocidental/genética , Febre do Nilo Ocidental/fisiopatologia , Febre do Nilo Ocidental/virologia
6.
PLoS Pathog ; 15(8): e1007899, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31415679

RESUMO

West Nile Virus (WNV), an emerging and re-emerging RNA virus, is the leading source of arboviral encephalitic morbidity and mortality in the United States. WNV infections are acutely controlled by innate immunity in peripheral tissues outside of the central nervous system (CNS) but WNV can evade the actions of interferon (IFN) to facilitate CNS invasion, causing encephalitis, encephalomyelitis, and death. Recent studies indicate that STimulator of INterferon Gene (STING), canonically known for initiating a type I IFN production and innate immune response to cytosolic DNA, is required for host defense against neurotropic RNA viruses. We evaluated the role of STING in host defense to control WNV infection and pathology in a murine model of infection. When challenged with WNV, STING knock out (-/-) mice displayed increased morbidity and mortality compared to wild type (WT) mice. Virologic analysis and assessment of STING activation revealed that STING signaling was not required for control of WNV in the spleen nor was WNV sufficient to mediate canonical STING activation in vitro. However, STING-/- mice exhibited a clear trend of increased viral load and virus dissemination in the CNS. We found that STING-/- mice exhibited increased and prolonged neurological signs compared to WT mice. Pathological examination revealed increased lesions, mononuclear cellular infiltration and neuronal death in the CNS of STING-/- mice, with sustained pathology after viral clearance. We found that STING was required in bone marrow derived macrophages for early control of WNV replication and innate immune activation. In vivo, STING-/- mice developed an aberrant T cell response in both the spleen and brain during WNV infection that linked with increased and sustained CNS pathology compared to WT mice. Our findings demonstrate that STING plays a critical role in immune programming for the control of neurotropic WNV infection and CNS disease.


Assuntos
Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Imunidade Inata/imunologia , Proteínas de Membrana/fisiologia , Replicação Viral , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Animais , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Carga Viral , Febre do Nilo Ocidental/metabolismo , Febre do Nilo Ocidental/virologia
7.
J Immunol ; 201(10): 3036-3050, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30297339

RESUMO

We examined the signaling pathways and cell type-specific responses of IFN regulatory factor (IRF) 5, an immune-regulatory transcription factor. We show that the protein kinases IKKα, IKKß, IKKε, and TANK-binding kinase 1 each confer IRF5 phosphorylation/dimerization, thus extending the family of IRF5 activator kinases. Among primary human immune cell subsets, we found that IRF5 is most abundant in plasmacytoid dendritic cells (pDCs). Flow cytometric cell imaging revealed that IRF5 is specifically activated by endosomal TLR signaling. Comparative analyses revealed that IRF3 is activated in pDCs uniquely through RIG-I-like receptor (RLR) signaling. Transcriptomic analyses of pDCs show that the partitioning of TLR7/IRF5 and RLR/IRF3 pathways confers differential gene expression and immune cytokine production in pDCs, linking IRF5 with immune regulatory and proinflammatory gene expression. Thus, TLR7/IRF5 and RLR-IRF3 partitioning serves to polarize pDC response outcome. Strategies to differentially engage IRF signaling pathways should be considered in the design of immunotherapeutic approaches to modulate or polarize the immune response for specific outcome.


Assuntos
Células Dendríticas/imunologia , Fator Regulador 3 de Interferon/imunologia , Fatores Reguladores de Interferon/imunologia , Transdução de Sinais/imunologia , Células Cultivadas , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/imunologia , Humanos , Fator Regulador 3 de Interferon/metabolismo , Fatores Reguladores de Interferon/metabolismo
8.
mBio ; 9(2)2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29559569

RESUMO

Induction of interferon beta (IFN-ß), IFN-stimulated genes (ISGs), and inflammatory responses is critical for control of viral infection. We recently identified an essential linkage of stimulation of the inflammatory cytokine interleukin-1ß (IL-1ß) and induction of ISGs that function as host restriction pathways against the emerging flavivirus West Nile virus (WNV) in vivo Here we utilized ex vivo global transcriptome analysis of primary dendritic cells, known targets of WNV replication, to define gene signatures required for this IL-1ß-driven antiviral response. Dendritic cells that were deficient in IL-1 receptor signaling showed dysregulation of cell-intrinsic defense genes and loss of viral control during WNV infection. Surprisingly, we found that in wild-type cells, IL-1ß treatment, in the absence of infection, drove the transcription of IFN-ß and ISGs at late times following treatment. Expression of these antiviral innate immune genes was dependent on the transcription factor IFN regulatory factor 3 (IRF3) and appears to reflect a general shift in IL-1ß signaling from an early inflammatory response to a late IFN-mediated response. These data demonstrate that inflammatory and antiviral signals integrate to control viral infection in myeloid cells through a process of IL-1ß-to-IRF3 signaling crosstalk. Strategies to exploit these cytokines in the activation of host defense programs should be investigated as novel therapeutic approaches against individual pathogens.IMPORTANCE West Nile virus is an emerging mosquito-borne flavivirus that can result in serious illness, neuropathology, and death in infected individuals. Currently, there are no vaccines or therapies for human use against West Nile virus. Immune control of West Nile virus infection requires inflammatory and antiviral responses, though the effect that each arm of this response has on the other is unclear. The significance of our research is in defining how virus-induced inflammatory responses regulate critical antiviral immune programs for effective control of West Nile virus infection. These data identify essential mechanisms of immune control that can inform therapeutic efforts against West Nile virus, with potential efficacy against other neuroinvasive viruses.


Assuntos
Células Dendríticas/metabolismo , Interleucina-1beta/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Cricetinae , Ensaio de Imunoadsorção Enzimática , Flavivirus/patogenicidade , Imunidade Inata/fisiologia , Inflamassomos/metabolismo , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Interleucina-1beta/genética , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Células Mieloides/virologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Vírus do Nilo Ocidental/patogenicidade
9.
Methods Mol Biol ; 1656: 119-129, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28808965

RESUMO

Pathogen recognition receptors (PRR)s and their cognate pathogen-associated molecular pattern (PAMP) represent the basis of innate immune activation and immune response induction driven by the host-pathogen interaction that occurs during microbial infection in humans and other animals. For RNA virus infection such as hepatitis C virus (HCV) and others, specific motifs within viral RNA mark it as nonself and visible to the host as a PAMP through interaction with RIG-I-like receptors including retinoic inducible gene-I (RIG-I). Here, we present methods for producing and using HCV PAMP RNA as a molecular tool to study RIG-I and its signaling pathway, both in vitro and in vivo, in innate immune regulation.


Assuntos
Proteína DEAD-box 58 , Hepacivirus , Hepatite C , Imunidade Inata , RNA Viral , Receptores de Reconhecimento de Padrão , Animais , Linhagem Celular , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Hepacivirus/genética , Hepacivirus/imunologia , Hepatite C/genética , Hepatite C/imunologia , Humanos , RNA Viral/química , RNA Viral/genética , RNA Viral/imunologia , Receptores Imunológicos , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/imunologia
10.
G3 (Bethesda) ; 7(6): 1665-1682, 2017 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-28592649

RESUMO

The oligoadenylate-synthetase (Oas) gene locus provides innate immune resistance to virus infection. In mouse models, variation in the Oas1b gene influences host susceptibility to flavivirus infection. However, the impact of Oas variation on overall innate immune programming and global gene expression among tissues and in different genetic backgrounds has not been defined. We examined how Oas1b acts in spleen and brain tissue to limit West Nile virus (WNV) susceptibility and disease across a range of genetic backgrounds. The laboratory founder strains of the mouse Collaborative Cross (CC) (A/J, C57BL/6J, 129S1/SvImJ, NOD/ShiLtJ, and NZO/HlLtJ) all encode a truncated, defective Oas1b, whereas the three wild-derived inbred founder strains (CAST/EiJ, PWK/PhJ, and WSB/EiJ) encode a full-length OAS1B protein. We assessed disease profiles and transcriptional signatures of F1 hybrids derived from these founder strains. F1 hybrids included wild-type Oas1b (F/F), homozygous null Oas1b (N/N), and heterozygous offspring of both parental combinations (F/N and N/F). These mice were challenged with WNV, and brain and spleen samples were harvested for global gene expression analysis. We found that the Oas1b haplotype played a role in WNV susceptibility and disease metrics, but the presence of a functional Oas1b allele in heterozygous offspring did not absolutely predict protection against disease. Our results indicate that Oas1b status as wild-type or truncated, and overall Oas1b gene dosage, link with novel innate immune gene signatures that impact specific biological pathways for the control of flavivirus infection and immunity through both Oas1b-dependent and independent processes.


Assuntos
2',5'-Oligoadenilato Sintetase/genética , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Transcrição Gênica , Febre do Nilo Ocidental/genética , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Perfilação da Expressão Gênica , Imunidade Inata/genética , Imunomodulação/genética , Imunomodulação/imunologia , Masculino , Camundongos , Locos de Características Quantitativas , Transcriptoma , Febre do Nilo Ocidental/virologia
11.
Methods Mol Biol ; 1488: 251-263, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27933528

RESUMO

The Collaborative Cross (CC) is a large panel of inbred mouse strains currently being developed for multiple areas of research. Scientists are taking integrated omics-style approaches to collecting data in order to obtain a deeper understanding of the biological mechanisms underlying a number of diverse disease phenotypes. As the cost of the next generation sequencing (NGS) decreases, RNA-sequencing (RNA-seq) has become the favored approach to transcriptomic analyses versus microarrays due to increases in sensitivity and resolution. This is particularly the case with newly defined genomes, where experimental annotation has not caught up to the new microarray platforms. Traditional RNA-seq approaches are not ideal when working with results from collaborative cross studies, as the genomes across individual strains differ considerably. In this chapter we will provide an overview of how to effectively perform RNA-seq analysis from data obtained from the CC mice.


Assuntos
Biologia Computacional/métodos , Cruzamentos Genéticos , Sequenciamento de Nucleotídeos em Larga Escala , Análise de Sequência de RNA , Software , Animais , Mapeamento Cromossômico , Genoma , Genômica/métodos , Camundongos , Camundongos Endogâmicos , Pseudogenes , Navegador
12.
PLoS Pathog ; 12(11): e1005996, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27806117

RESUMO

Infection with West Nile virus (WNV) leads to a range of disease outcomes, including chronic infection, though lack of a robust mouse model of chronic WNV infection has precluded identification of the immune events contributing to persistent infection. Using the Collaborative Cross, a population of recombinant inbred mouse strains with high levels of standing genetic variation, we have identified a mouse model of persistent WNV disease, with persistence of viral loads within the brain. Compared to lines exhibiting no disease or marked disease, the F1 cross CC(032x013)F1 displays a strong immunoregulatory signature upon infection that correlates with restraint of the WNV-directed cytolytic response. We hypothesize that this regulatory T cell response sufficiently restrains the immune response such that a chronic infection can be maintained in the CNS. Use of this new mouse model of chronic neuroinvasive virus will be critical in developing improved strategies to prevent prolonged disease in humans.


Assuntos
Linfócitos T Reguladores/imunologia , Febre do Nilo Ocidental/imunologia , Animais , Doença Crônica , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Vírus do Nilo Ocidental/imunologia
13.
Genom Data ; 10: 114-117, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27843766

RESUMO

Flaviviruses are hematophagous arthropod-viruses that pose global challenges to human health. Like Zika virus, West Nile Virus (WNV) is a flavivirus for which no approved vaccine exists [1]. The role host genetics play in early detection and response to WNV still remains largely unexplained. In order to capture the impact of genetic variation on innate immune responses, we studied gene expression following WNV infection using the collaborative cross (CC). The CC is a mouse genetics resource composed of hundreds of independently bred, octo-parental recombinant inbred mouse lines [2]. To accurately capture the host immune gene expression signatures of West Nile infection, we used the nanostring platform to evaluate expression in spleen tissue isolated from CC mice infected with WNV over a time course of 4, 7, and 12 days' post-infection [3]. Nanostring is a non-amplification based digital method to quantitate gene expression that uses color-coded molecular barcodes to detect hundreds of transcripts in a sample. Using this approach, we identified unique gene signatures in spleen tissue at days 4, 7, and 12 following WNV infection, which delineated distinct differences between asymptomatic and symptomatic CC lines. We also identified novel immune genes. Data was deposited into the Gene Expression Omnibus under accession GSE86000.

14.
Genom Data ; 10: 137-140, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27872814

RESUMO

West Nile Virus (WNV) is a mosquito-transmitted virus from the Flaviviridae family that causes fever in 1 in 5 infected people. WNV can also become neuro-invasive and cross the blood-brain barrier leading to severe neurological symptoms in a subset of WNV infected individuals [1]. WNV neuro-invasion is believed to be influenced by a number of factors including host genetics. In order to explore these effects and recapitulate the complex immune genetic differences among individuals, we studied gene expression following WNV infection in the Collaborative Cross (CC) model. The CC is a mouse genetics resource composed of > 70 independently bred, octo-parental recombinant inbred mouse lines [2]. To identify the individual host gene expression signatures influencing protection or susceptibility to WNV disease and WNV neuroinvasion, we used the nanostring nsolver platform to quantify gene expression in brain tissue isolated from WNV-infected CC mice at days 4, 7 and 12 post-infection [3]. This nanostring technology provided a high throughput, non-amplification based mRNA quantitation method to detect immune genes involved in neuro-invasion. Data was deposited into the Gene Expression Omnibus (GEO) under accession GSE85999.

15.
J Virol ; 90(5): 2372-87, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26676770

RESUMO

UNLABELLED: The cellular response to virus infection is initiated when pathogen recognition receptors (PRR) engage viral pathogen-associated molecular patterns (PAMPs). This process results in induction of downstream signaling pathways that activate the transcription factor interferon regulatory factor 3 (IRF3). IRF3 plays a critical role in antiviral immunity to drive the expression of innate immune response genes, including those encoding antiviral factors, type 1 interferon, and immune modulatory cytokines, that act in concert to restrict virus replication. Thus, small molecule agonists that can promote IRF3 activation and induce innate immune gene expression could serve as antivirals to induce tissue-wide innate immunity for effective control of virus infection. We identified small molecule compounds that activate IRF3 to differentially induce discrete subsets of antiviral genes. We tested a lead compound and derivatives for the ability to suppress infections caused by a broad range of RNA viruses. Compound administration significantly decreased the viral RNA load in cultured cells that were infected with viruses of the family Flaviviridae, including West Nile virus, dengue virus, and hepatitis C virus, as well as viruses of the families Filoviridae (Ebola virus), Orthomyxoviridae (influenza A virus), Arenaviridae (Lassa virus), and Paramyxoviridae (respiratory syncytial virus, Nipah virus) to suppress infectious virus production. Knockdown studies mapped this response to the RIG-I-like receptor pathway. This work identifies a novel class of host-directed immune modulatory molecules that activate IRF3 to promote host antiviral responses to broadly suppress infections caused by RNA viruses of distinct genera. IMPORTANCE: Incidences of emerging and reemerging RNA viruses highlight a desperate need for broad-spectrum antiviral agents that can effectively control infections caused by viruses of distinct genera. We identified small molecule compounds that can selectively activate IRF3 for the purpose of identifying drug-like molecules that can be developed for the treatment of viral infections. Here, we report the discovery of a hydroxyquinoline family of small molecules that can activate IRF3 to promote cellular antiviral responses. These molecules can prophylactically or therapeutically control infection in cell culture by pathogenic RNA viruses, including West Nile virus, dengue virus, hepatitis C virus, influenza A virus, respiratory syncytial virus, Nipah virus, Lassa virus, and Ebola virus. Our study thus identifies a class of small molecules with a novel mechanism to enhance host immune responses for antiviral activity against a variety of RNA viruses that pose a significant health care burden and/or that are known to cause infections with high case fatality rates.


Assuntos
Antivirais/farmacologia , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Vírus de RNA/imunologia , Vírus de RNA/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/isolamento & purificação , Linhagem Celular , Perfilação da Expressão Gênica , Humanos , Fatores Imunológicos/isolamento & purificação , Carga Viral , Cultura de Vírus
16.
PLoS One ; 10(3): e0117963, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25734423

RESUMO

RIG-I pathway signaling of innate immunity against RNA virus infection is organized between the ER and mitochondria on a subdomain of the ER called the mitochondrial-associated ER membrane (MAM). The RIG-I adaptor protein MAVS transmits downstream signaling of antiviral immunity, with signaling complexes assembling on the MAM in association with mitochondria and peroxisomes. To identify components that regulate MAVS signalosome assembly on the MAM, we characterized the proteome of MAM, ER, and cytosol from cells infected with either chronic (hepatitis C) or acute (Sendai) RNA virus infections, as well as mock-infected cells. Comparative analysis of protein trafficking dynamics during both chronic and acute viral infection reveals differential protein profiles in the MAM during RIG-I pathway activation. We identified proteins and biochemical pathways recruited into and out of the MAM in both chronic and acute RNA viral infections, representing proteins that drive immunity and/or regulate viral replication. In addition, by using this comparative proteomics approach, we identified 3 new MAVS-interacting proteins, RAB1B, VTN, and LONP1, and defined LONP1 as a positive regulator of the RIG-I pathway. Our proteomic analysis also reveals a dynamic cross-talk between subcellular compartments during both acute and chronic RNA virus infection, and demonstrates the importance of the MAM as a central platform that coordinates innate immune signaling to initiate immunity against RNA virus infection.


Assuntos
Retículo Endoplasmático/metabolismo , Hepacivirus/fisiologia , Mitocôndrias/metabolismo , Proteoma/análise , Vírus Sendai/fisiologia , Espectrometria de Massas em Tandem , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Retículo Endoplasmático/virologia , Humanos , Redes e Vias Metabólicas , Microssomos/metabolismo , Microssomos/virologia , Mitocôndrias/virologia , Análise de Componente Principal , Replicação Viral
17.
Proc Natl Acad Sci U S A ; 111(5): 1909-14, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24449862

RESUMO

In response to viral infection, the host induces over 300 IFN-stimulated genes (ISGs), which are the central component of intracellular antiviral innate immunity. Inefficient induction of ISGs contributes to poor control and persistence of hepatitis C virus infection. Therefore, further understanding of the hepatocytic ISG regulation machinery will guide us to an improved management strategy against hepatitis C virus infection. In this study, comprehensive genome-wide, high-throughput cDNA screening for genes regulating ISG expression identified a tyrosine kinase nonreceptor 1 (TNK1) as a unique player in the ISG induction pathway. The immune-modulatory function of TNK1 has never been studied, and this study characterizes its significance in antiviral innate immunity. TNK1 is abundantly expressed in hepatocytes and maintains basal ISG expression. More importantly, TNK1 plays a critical role in type I IFN-mediated ISG induction. We discovered that the activated IFN receptor complex recruits TNK1 from the cytoplasm. TNK1 is then phosphorylated to enhance its kinase activity. The activated TNK1 potentiates JAK-STAT signaling through dual phosphorylation of STAT1 at tyrosine 701 and serine 727 amino acid positions. Our loss-of-function approach demonstrated that TNK1 governs a cluster of ISG expression that defines the TNK1 pathway effector genes. More importantly, TNK1 abundance is inversely correlated to viral replication efficiency and is also a determinant factor for the hepatocytic response to antiviral treatment. Taken together, our studies found a critical but unidentified integrated component of the IFN-JAK-STAT signaling cascade.


Assuntos
Antivirais/metabolismo , Proteínas Fetais/metabolismo , Interferons/metabolismo , Fosfosserina/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Animais , Linhagem Celular Tumoral , DNA Complementar/genética , Suscetibilidade a Doenças , Deleção de Genes , Regulação da Expressão Gênica , Testes Genéticos , Genoma Humano/genética , Hepacivirus/fisiologia , Hepatite C/enzimologia , Hepatite C/genética , Hepatite C/patologia , Hepatite C/virologia , Hepatócitos/enzimologia , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Imunidade Inata/genética , Janus Quinase 1/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação
18.
Immunity ; 38(1): 3-5, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23352217

RESUMO

In this issue of Immunity, studies by Blanc et al. (2013) and Liu et al. (2013) reveal how interferon induction of cholesterol-25-hydroxylase mediates innate immunity against multiple diverse viruses.

19.
PLoS Pathog ; 9(1): e1003118, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23300459

RESUMO

Although the transcription factors IRF-3 and IRF-7 are considered master regulators of type I interferon (IFN) induction and IFN stimulated gene (ISG) expression, Irf3(-/-)×Irf7(-/-) double knockout (DKO) myeloid dendritic cells (mDC) produce relatively normal levels of IFN-ß after viral infection. We generated Irf3(-/-)×Irf5(-/-)×Irf7(-/-) triple knockout (TKO) mice to test whether IRF-5 was the source of the residual induction of IFN-ß and ISGs in mDCs. In pathogenesis studies with two unrelated positive-sense RNA viruses (West Nile virus (WNV) and murine norovirus), TKO mice succumbed at rates greater than DKO mice and equal to or approaching those of mice lacking the type I IFN receptor (Ifnar(-/-)). In ex vivo studies, after WNV infection or exposure to Toll-like receptor agonists, TKO mDCs failed to produce IFN-ß or express ISGs. In contrast, this response was sustained in TKO macrophages following WNV infection. To define IRF-regulated gene signatures, we performed microarray analysis on WNV-infected mDC from wild type (WT), DKO, TKO, or Ifnar(-/-) mice, as well as from mice lacking the RIG-I like receptor adaptor protein MAVS. Whereas the gene induction pattern in DKO mDC was similar to WT cells, remarkably, almost no ISG induction was detected in TKO or Mavs(-/-) mDC. The relative equivalence of TKO and Mavs(-/-) responses suggested that MAVS dominantly regulates ISG induction in mDC. Moreover, we showed that MAVS-dependent induction of ISGs can occur through an IRF-5-dependent yet IRF-3 and IRF-7-independent pathway. Our results establish IRF-3, -5, and -7 as the key transcription factors responsible for mediating the type I IFN and ISG response in mDC during WNV infection and suggest a novel signaling link between MAVS and IRF-5.


Assuntos
Fator Regulador 3 de Interferon/imunologia , Fator Regulador 7 de Interferon/imunologia , Fatores Reguladores de Interferon/imunologia , Interferon beta/imunologia , Vírus do Nilo Ocidental/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Receptor de Interferon alfa e beta/genética , Transdução de Sinais , Receptores Toll-Like/imunologia , Carga Viral , Febre do Nilo Ocidental/genética , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/genética
20.
Hepatology ; 57(2): 461-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22996292

RESUMO

UNLABELLED: Type 1 interferon (IFN) continues to be the foundation for the current standard of care combination therapy for chronic hepatitis C virus (HCV) infection, yet the component interferon-stimulated genes (ISGs) that mediate the antiviral actions of IFN are not fully defined. Interferon-induced transmembrane protein 1 (IFITM1) is an ISG product that suppresses early stage infection by a number of viruses through an unknown mechanism of action. Moreover, the actions of IFITM1 on HCV infection are not fully elucidated. Here we identify IFITM1 as a hepatocyte tight junction protein and a potent anti-HCV effector molecule. IFITM1 expression is induced early during IFN treatment of hepatocytes and accumulates at hepatic tight junctions in HCV-infected human patient liver during IFN therapy. Additionally, we found that IFITM1 interacts with HCV coreceptors, including CD81 and occludin, to disrupt the process of viral entry. Thus, IFITM1 is an anti-HCV ISG whose actions impart control of HCV infection through interruption of viral coreceptor function. CONCLUSION: This study defines IFITM1 as an ISG effector with action against HCV entry. Design of therapy regimens to enhance IFITM1 expression should improve the virologic response among HCV patients undergoing treatment with type I IFN.


Assuntos
Antígenos de Diferenciação/uso terapêutico , Antivirais/uso terapêutico , Hepatite C/imunologia , Interferon Tipo I/uso terapêutico , Receptores Virais/efeitos dos fármacos , Proteínas de Junções Íntimas/fisiologia , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/metabolismo , Células Cultivadas , Hepacivirus/efeitos dos fármacos , Hepacivirus/fisiologia , Humanos , Tetraspanina 28/metabolismo , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...