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.
Allergy ; 76(6): 1629-1639, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33452689

RESUMO

The first approved COVID-19 vaccines include Pfizer/BioNTech BNT162B2, Moderna mRNA-1273 and AstraZeneca recombinant adenoviral ChAdOx1-S. Soon after approval, severe allergic reactions to the mRNA-based vaccines that resolved after treatment were reported. Regulatory agencies from the European Union, Unites States and the United Kingdom agree that vaccinations are contraindicated only when there is an allergy to one of the vaccine components or if there was a severe allergic reaction to the first dose. This position paper of the European Academy of Allergy and Clinical Immunology (EAACI) agrees with these recommendations and clarifies that there is no contraindication to administer these vaccines to allergic patients who do not have a history of an allergic reaction to any of the vaccine components. Importantly, as is the case for any medication, anaphylaxis may occur after vaccination in the absence of a history of allergic disease. Therefore, we provide a simplified algorithm of prevention, diagnosis and treatment of severe allergic reactions and a list of recommended medications and equipment for vaccine centres. We also describe potentially allergenic/immunogenic components of the approved vaccines and propose a workup to identify the responsible allergen. Close collaboration between academia, regulatory agencies and vaccine producers will facilitate approaches for patients at risks, such as incremental dosing of the second injection or desensitization. Finally, we identify unmet research needs and propose a concerted international roadmap towards precision diagnosis and management to minimize the risk of allergic reactions to COVID-19 vaccines and to facilitate their broader and safer use.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Vacina BNT162 , Humanos , SARS-CoV-2 , Reino Unido
2.
Front Immunol ; 11: 605673, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33542718

RESUMO

Type I interferons (IFN-I) were first discovered as an antiviral factor by Isaacs and Lindenmann in 1957, but they are now known to also modulate innate and adaptive immunity and suppress proliferation of cancer cells. While much has been revealed about IFN-I, it remains a mystery as to why there are 16 different IFN-I gene products, including IFNß, IFNω, and 12 subtypes of IFNα. Here, we discuss shared and unique aspects of these IFN-I in the context of their evolution, expression patterns, and signaling through their shared heterodimeric receptor. We propose that rather than investigating responses to individual IFN-I, these contexts can serve as an alternative approach toward investigating roles for IFNα subtypes. Finally, we review uses of IFNα and IFNß as therapeutic agents to suppress chronic viral infections or to treat multiple sclerosis.


Assuntos
Interferon-alfa/metabolismo , Interferon beta/metabolismo , Antivirais/uso terapêutico , Evolução Molecular , Interações entre Hospedeiro e Microrganismos , Humanos , Fatores Imunológicos/uso terapêutico , Fatores Reguladores de Interferon/metabolismo , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon-alfa/uso terapêutico , Interferon beta/genética , Interferon beta/imunologia , Interferon beta/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Transdução de Sinais , Viroses/tratamento farmacológico , Viroses/imunologia , Viroses/metabolismo , Viroses/virologia
3.
Front Immunol ; 10: 1019, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31156620

RESUMO

Viral defense at mucosal sites depends on interferons (IFN) and IFN stimulated genes (ISGs), either of which may be constitutively expressed to maintain an "antiviral state" (AVS). However, the mechanisms that govern the AVS are poorly defined. Using a BEAS-2B respiratory epithelial cell line deficient in IRF1, we demonstrate higher susceptibility to infection with vesicular stomatitis virus (VSV) and influenza virus. IRF1-mediated restriction of VSV is IFN-independent, as blockade of types I and III IFNs and JAK-STAT signaling before infection did not affect VSV infection of either parent or IRF1 KO cells. Transcriptome analysis revealed that IRF1 regulates constitutive expression of ~300 genes, including antiviral ISGs: OAS2, BST2, and RNASEL and knockdown of any of these IRF1-dependent genes increased VSV infection. Additionally, IRF1 enhances rapid expression of IFNß and IFNλ after stimulation with poly I:C and also regulates ISG expression. Mechanistically, IRF1 enhances recruitment of BRD4 to promotor-enhancer regions of ISGs for rapid expression and maintains levels of histone H3K4me1 for optimal constitutive expression. Finally, IRF1 also regulates constitutive expression of TLR2 and TLR3 and promotes signaling through these pattern recognition receptors (PRR). These data reveal multiple roles for IRF1 toward effective anti-viral responses by maintaining IFN-independent constitutive expression of anti-viral ISGs and supporting early IFN-dependent responses to PRR stimulation.


Assuntos
2',5'-Oligoadenilato Sintetase/genética , Antígenos CD/genética , Endorribonucleases/genética , Influenza Humana/imunologia , Fator Regulador 1 de Interferon/genética , Orthomyxoviridae/imunologia , Infecções por Rhabdoviridae/imunologia , Vesiculovirus/imunologia , Células A549 , Células Epiteliais/metabolismo , Proteínas Ligadas por GPI/genética , Regulação da Expressão Gênica/genética , Técnicas de Inativação de Genes , Humanos , Influenza Humana/virologia , Fator Regulador 1 de Interferon/metabolismo , Interferons/metabolismo , Mucosa Respiratória/citologia , Infecções por Rhabdoviridae/virologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptores Toll-Like/metabolismo , Transfecção , Replicação Viral/genética , Replicação Viral/imunologia
4.
J Interferon Cytokine Res ; 39(5): 283-292, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30920934

RESUMO

Type I interferons (IFNs) signal by forming a high affinity IFN-IFNAR2 dimer, which subsequently recruits IFNAR1 to form a ternary complex that initiates JAK/STAT signaling. Among the 12 IFNα subtypes, IFNα1 has a uniquely low affinity for IFNAR2 (<100 × of the other IFNα subtypes) and commensurately weak antiviral activity, suggesting an undefined function distinct from suppression of viral infections. Also unique in IFNα1 is substitution of a serine for phenylalanine at position 27, a contact point that stabilizes the IFNα:IFNAR2 hydrophobic interface. To determine whether IFNα1-S27 contributes to the low affinity for IFNAR2, we created an IFNα1 mutein, IFNα1-S27F, and compared it to wild-type IFNα1 and IFNα2. Substitution of phenylalanine for serine increased affinity for IFNAR2 ∼4-fold and commensurately enhanced activation of STAT1, STAT3, and STAT5, transcription of a subset of interferon stimulated genes, and restriction of vesicular stomatitis virus infection in vitro. Structural modeling suggests that S27 of IFNα1 disrupts the IFNα:IFNAR2 hydrophobic interface that is otherwise stabilized by F27 and that replacing S27 with phenylalanine partially restores the hydrophobic surface. Disruption of the hydrophobic IFNα:IFNAR2 interface by the unique S27 of IFN α1 contributes to its low affinity and weak antiviral activity.


Assuntos
Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Serina , Vesiculovirus/imunologia , Humanos , Interações Hidrofóbicas e Hidrofílicas , Interferon-alfa/química , Testes de Sensibilidade Microbiana , Modelos Moleculares , Serina/genética , Serina/metabolismo , Células Tumorais Cultivadas
5.
J Virol ; 92(17)2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29976658

RESUMO

Host-influenza virus interplay at the transcript level has been extensively characterized in epithelial cells. Yet, there are no studies that simultaneously characterize human host and influenza A virus (IAV) genomes. We infected human bronchial epithelial BEAS-2B cells with two seasonal IAV/H3N2 strains, Brisbane/10/07 and Perth/16/09 (reference strains for past vaccine seasons) and the well-characterized laboratory strain Udorn/307/72. Strand-specific RNA sequencing (RNA-seq) of the infected BEAS-2B cells allowed for simultaneous analysis of host and viral transcriptomes, in addition to pathogen genomes, to reveal changes in mRNA expression and alternative splicing (AS). In general, patterns of global and immune gene expression induced by the three IAVs were mostly shared. However, AS of host transcripts and small nuclear RNAs differed between the seasonal and laboratory strains. Analysis of viral transcriptomes showed deletions of the polymerase components (defective interfering-like RNAs) within the genome. Surprisingly, we found that the neuraminidase gene undergoes AS and that the splicing event differs between seasonal and laboratory strains. Our findings reveal novel elements of the host-virus interaction and highlight the importance of RNA-seq in identifying molecular changes at the genome level that may contribute to shaping RNA-based innate immunity.IMPORTANCE The use of massively parallel RNA sequencing (RNA-seq) has revealed insights into human and pathogen genomes and their evolution. Dual RNA-seq allows simultaneous dissection of host and pathogen genomes and strand-specific RNA-seq provides information about the polarity of the RNA. This is important in the case of negative-strand RNA viruses like influenza virus, which generate positive (complementary and mRNA) and negative-strand RNAs (genome) that differ in their potential to trigger innate immunity. Here, we characterize interactions between human bronchial epithelial cells and three influenza A/H3N2 strains using strand-specific dual RNA-seq. We focused on this subtype because of its epidemiological importance in causing significant morbidity and mortality during influenza epidemics. We report novel elements that differ between seasonal and laboratory strains highlighting the complexity of the host-virus interplay at the RNA level.


Assuntos
Genoma Humano/genética , Genoma Viral/genética , Interações Hospedeiro-Patógeno/genética , Imunidade Inata/genética , Vírus da Influenza A Subtipo H3N2/genética , Influenza Humana/imunologia , Brônquios/citologia , Brônquios/virologia , Células Epiteliais/virologia , Perfilação da Expressão Gênica , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Vírus da Influenza A Subtipo H3N2/imunologia , Influenza Humana/virologia , Neuraminidase/genética , Splicing de RNA/genética , Estações do Ano , Análise de Sequência de RNA/métodos
6.
J Virol ; 92(15)2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29769339

RESUMO

Respiratory syncytial virus (RSV) infects small foci of respiratory epithelial cells via infected droplets. Infection induces expression of type I and III interferons (IFNs) and proinflammatory cytokines, the balance of which may restrict viral replication and affect disease severity. We explored this balance by infecting two respiratory epithelial cell lines with low doses of recombinant RSV expressing green fluorescent protein (rgRSV). A549 cells were highly permissive, whereas BEAS-2B cells restricted infection to individual cells or small foci. After infection, A549 cells expressed higher levels of IFN-ß-, IFN-λ-, and NF-κB-inducible proinflammatory cytokines. In contrast, BEAS-2B cells expressed higher levels of antiviral interferon-stimulated genes, pattern recognition receptors, and other signaling intermediaries constitutively and after infection. Transcriptome analysis revealed that constitutive expression of antiviral and proinflammatory genes predicted responses by each cell line. These two cell lines provide a model for elucidating critical mediators of local control of viral infection in respiratory epithelial cells.IMPORTANCE Airway epithelium is both the primary target of and the first defense against respiratory syncytial virus (RSV). Whether RSV replicates and spreads to adjacent epithelial cells depends on the quality of their innate immune responses. A549 and BEAS-2B are alveolar and bronchial epithelial cell lines, respectively, that are often used to study RSV infection. We show that A549 cells are permissive to RSV infection and express genes characteristic of a proinflammatory response. In contrast, BEAS-2B cells restrict infection and express genes characteristic of an antiviral response associated with expression of type I and III interferons. Transcriptome analysis of constitutive gene expression revealed patterns that may predict the response of each cell line to infection. This study suggests that restrictive and permissive cell lines may provide a model for identifying critical mediators of local control of infection and stresses the importance of the constitutive antiviral state for the response to viral challenge.


Assuntos
Citocinas/imunologia , Células Epiteliais/imunologia , Regulação da Expressão Gênica/imunologia , Mucosa Respiratória/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Células A549 , Células Epiteliais/virologia , Humanos , Mucosa Respiratória/virologia , Infecções por Vírus Respiratório Sincicial/patologia
7.
Virology ; 504: 63-72, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28157546

RESUMO

Whether respiratory syncytial virus (RSV) induces severe infantile pulmonary disease may depend on viral strain and expression of types I and III interferons (IFNs). These IFNs impact disease severity by inducing expression of many anti-viral IFN-stimulated genes (ISGs). To investigate the impact of RSV strain on IFN and ISG expression, we stimulated human monocyte-derived DCs (MDDCs) with either RSV A2 or Line 19 and measured expression of types I and III IFNs and ISGs. At 24h, A2 elicited higher ISG expression than Line 19. Both strains induced MDDCs to express genes for IFN-ß, IFN-α1, IFN-α8, and IFN-λ1-3, but only A2 induced IFN-α2, -α14 and -α21. We then show that IFN-α8 and IFN-α14 most potently induced MDDCs and bronchial epithelial cells (BECs) to express ISGs. Our findings demonstrate that RSV strain may impact patterns of types I and III IFN expression and the magnitude of the ISG response by DCs and BECs.


Assuntos
Células Dendríticas/imunologia , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Adulto , Brônquios/citologia , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/virologia , Células Epiteliais/citologia , Humanos , Inflamação/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Mucosa Respiratória/citologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/metabolismo
8.
J Virol ; 89(15): 7567-83, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25972536

RESUMO

UNLABELLED: Ebola virus (EBOV) causes a severe hemorrhagic fever with a deficient immune response, lymphopenia, and lymphocyte apoptosis. Dendritic cells (DC), which trigger the adaptive response, do not mature despite EBOV infection. We recently demonstrated that DC maturation is unblocked by disabling the innate response antagonizing domains (IRADs) in EBOV VP35 and VP24 by the mutations R312A and K142A, respectively. Here we analyzed the effects of VP35 and VP24 with the IRADs disabled on global gene expression in human DC. Human monocyte-derived DC were infected by wild-type (wt) EBOV or EBOVs carrying the mutation in VP35 (EBOV/VP35m), VP24 (EBOV/VP24m), or both (EBOV/VP35m/VP24m). Global gene expression at 8 and 24 h was analyzed by deep sequencing, and the expression of interferon (IFN) subtypes up to 5 days postinfection was analyzed by quantitative reverse transcription-PCR (qRT-PCR). wt EBOV induced a weak global gene expression response, including markers of DC maturation, cytokines, chemokines, chemokine receptors, and multiple IFNs. The VP35 mutation unblocked the expression, resulting in a dramatic increase in expression of these transcripts at 8 and 24 h. Surprisingly, DC infected with EBOV/VP24m expressed lower levels of many of these transcripts at 8 h after infection, compared to wt EBOV. In contrast, at 24 h, expression of the transcripts increased in DC infected with any of the three mutants, compared to wt EBOV. Moreover, sets of genes affected by the two mutations only partially overlapped. Pathway analysis demonstrated that the VP35 mutation unblocked pathways involved in antigen processing and presentation and IFN signaling. These data suggest that EBOV IRADs have profound effects on the host adaptive immune response through massive transcriptional downregulation of DC. IMPORTANCE: This study shows that infection of DC with EBOV, but not its mutant forms with the VP35 IRAD and/or VP24 IRAD disabled, causes a global block in expression of host genes. The temporal effects of mutations disrupting the two IRADs differ, and the lists of affected genes only partially overlap such that VP35 and VP24 IRADs each have profound effects on antigen presentation by exposed DC. The global modulation of DC gene expression and the resulting lack of their maturation represent a major mechanism by which EBOV disables the T cell response and suggests that these suppressive pathways are a therapeutic target that may unleash the T cell responses during EBOV infection.


Assuntos
Células Dendríticas/metabolismo , Ebolavirus/metabolismo , Expressão Gênica , Doença pelo Vírus Ebola/genética , Interferons/genética , Nucleoproteínas/metabolismo , Proteínas do Core Viral/metabolismo , Proteínas Virais/metabolismo , Células Cultivadas , Células Dendríticas/virologia , Ebolavirus/química , Ebolavirus/genética , Doença pelo Vírus Ebola/metabolismo , Doença pelo Vírus Ebola/virologia , Humanos , Interferons/metabolismo , Proteínas do Nucleocapsídeo , Nucleoproteínas/química , Nucleoproteínas/genética , Estrutura Terciária de Proteína , Transdução de Sinais , Proteínas do Core Viral/química , Proteínas do Core Viral/genética , Proteínas Virais/química , Proteínas Virais/genética
9.
J Virol ; 88(11): 6453-69, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24672038

RESUMO

UNLABELLED: Human metapneumovirus (HMPV) is a major etiologic agent of respiratory disease worldwide. HMPV reinfections are common in healthy adults and children, suggesting that the protective immune response to HMPV is incomplete and short-lived. We used gene-deletion viruses to evaluate the role of the attachment G and small hydrophobic SH glycoproteins on virus uptake by primary human monocyte-derived dendritic cells (MDDC) in vitro and on subsequent MDDC maturation and activation of autologous T cells. HMPV with deletion of G and SH (ΔSHG) exhibited increased infectivity but had little effect on MDDC maturation. However, MDDC stimulated with ΔSHG induced increased proliferation of autologous Th1-polarized CD4(+) T cells. This effect was independent of virus replication. Increased T cell proliferation was strictly dependent on contact between virus-stimulated MDDC and CD4(+) T cells. Confocal microscopy revealed that deletion of SH and G was associated with an increased number of immunological synapses between memory CD4(+) T cells and virus-stimulated MDDC. Uptake of HMPV by MDDC was found to be primarily by macropinocytosis. Uptake of wild-type (WT) virus was reduced compared to that of ΔSHG, indicative of inhibition by the SH and G glycoproteins. In addition, DC-SIGN-mediated endocytosis provided a minor alternative pathway that depended on SH and/or G and thus operated only for WT. Altogether, our results show that SH and G glycoproteins reduce the ability of HMPV to be internalized by MDDC, resulting in a reduced ability of the HMPV-stimulated MDDC to activate CD4(+) T cells. This study describes a previously unknown mechanism of virus immune evasion. IMPORTANCE: Human metapneumovirus (HMPV) is a major etiologic agent of respiratory disease worldwide. HMPV reinfections are common in healthy adults and children, suggesting that the protective immune response to HMPV is incomplete and short-lived. We found that HMPV attachment G and small hydrophobic SH glycoproteins reduce the ability of HMPV to be internalized by macropinocytosis into human dendritic cells (DC). This results in a reduced ability of the HMPV-stimulated DC to activate Th1-polarized CD4(+) T cells. These results contribute to a better understanding of the nature of incomplete protection against this important human respiratory virus, provide new information on the entry of HMPV into human cells, and describe a new mechanism of virus immune evasion.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/virologia , Glicoproteínas/imunologia , Evasão da Resposta Imune/imunologia , Metapneumovirus/imunologia , Pinocitose/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Proteínas Virais/imunologia , Análise de Variância , Moléculas de Adesão Celular/imunologia , Células Dendríticas/imunologia , Citometria de Fluxo , Humanos , Sinapses Imunológicas/imunologia , Lectinas Tipo C/imunologia , Ativação Linfocitária/imunologia , Metapneumovirus/genética , Microscopia Confocal , Receptores de Superfície Celular/imunologia , Internalização do Vírus
10.
Eur J Immunol ; 43(12): 3197-208, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24030809

RESUMO

Human type I interferons (IFNs) include IFN-ß and 12 subtypes of IFN-α. During viral infection, infiltrating memory CD4(+) T cells are exposed to IFNs, but their impact on memory T-cell function is poorly understood. To address this, we pretreated PBMCs with different IFNs for 16 h before stimulation with Staphylococcus aureus enterotoxin B and measured cytokine expression by flow cytometry. IFN-α8 and -α10 most potently enhanced expression of IFN-γ, IL-2, and IL-4. Potency among the subtypes differed most at doses between 10 and 100 U/mL. While enhancement of IL-2 and IL-4 correlated with the time of preincubation with type I IFN, IFN-γ production was enhanced best when IFN-α was added immediately preceding or simultaneously with T-cell stimulation. Comparison of T-cell responses to multiple doses of Staphylococcus aureus enterotoxin B and to peptide libraries from RSV or CMV demonstrated that IFN-α best enhanced cytokine expression when CD4(+) T cells were suboptimally stimulated. We conclude that type I IFNs enhance Th1 and Th2 function with dose dependency and subtype specificity, and best when T-cell stimulation is suboptimal. While type I IFNs may beneficially enhance CD4(+) T-cell memory responses to vaccines or viral pathogens, they may also enhance the function of resident Th2 cells and exacerbate allergic inflammation.


Assuntos
Interferon-alfa/imunologia , Células Th1/imunologia , Células Th2/imunologia , Proteínas de Bactérias/química , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/farmacologia , Citomegalovirus/química , Citomegalovirus/imunologia , Enterotoxinas/química , Enterotoxinas/imunologia , Enterotoxinas/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Memória Imunológica/efeitos dos fármacos , Memória Imunológica/imunologia , Interferon gama/imunologia , Interleucina-2/imunologia , Interleucina-4/imunologia , Masculino , Peptídeos/química , Peptídeos/imunologia , Peptídeos/farmacologia , Vírus Sinciciais Respiratórios/química , Vírus Sinciciais Respiratórios/imunologia , Staphylococcus aureus/química , Staphylococcus aureus/imunologia , Células Th1/citologia , Células Th2/citologia , Proteínas Virais/química , Proteínas Virais/imunologia , Proteínas Virais/farmacologia
11.
Hum Immunol ; 74(12): 1524-30, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23993990

RESUMO

Macrophages can be polarized into classically (CAM) or alternatively (AAM) activated macrophages with IFN-γ or IL-4, respectively. CAM are associated with type 1 immune responses and are implicated in autoimmunity; AAM are associated with type 2 responses and are implicated in allergic diseases. An impediment in investigating macrophage biology using primary human monocyte derived macrophages is the wide inter-donor heterogeneity and the limited quantity of cells that survive in vitro polarization. To overcome this impediment, we established a protocol to generate CAM and AAM cultures derived from the THP-1 human promonocytic cell line. In this report, we demonstrate that THP-CAM and -AAM express gene and protein markers that define their primary human monocyte derived counterparts, such as IL-1ß, CXCL10, and CXCL11 for CAM, and MRC1, IL-4 and CCL22 for AAM. In addition, we demonstrate that STAT6 is selectively activated in THP-AAM which, upon LPS stimulation, have an attenuated or delayed expression of IFN-ß, IFN-λ1, and IFN α/ß pathway genes compared to their CAM counterparts. Taken together, these findings may help further investigate human diseases associated with the alternatively activated macrophage phenotype using this reproducible in vitro macrophage model.


Assuntos
Interferon beta/metabolismo , Interferon gama/metabolismo , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Interferon beta/genética , Interferon gama/genética , Ativação de Macrófagos/genética , Monócitos/citologia , Monócitos/imunologia , Monócitos/metabolismo , Transdução de Sinais
12.
J Interferon Cytokine Res ; 32(9): 407-15, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22817480

RESUMO

Rhesus macaques provide a valuable research and preclinical model for cancer and infectious diseases, as nonhuman primates share immune pathways with humans. Interferons (IFNs) are key cytokines in both innate and adaptive immunity, so a detailed analysis of gene expression in peripheral blood and tissues may shed insight into immune responses. Macaques have 18 IFN genes, of which 14 encode for 13 distinct IFN-α subtypes, and one for IFN-ß. Here, we developed a high-throughput array to evaluate each of the IFN-α subtypes, as well as IFN-ß, IFN-γ and 2 subtypes of IFN-λ. With this array, expression of each IFN species may be quantified as relative to a reference (housekeeping) gene (ΔCq) or fitted to its own 4-point standard curve for absolute quantification (copy number per mass unit RNA). After validating the assay with IFN complementary DNA, we determined the IFN expression profile of peripheral blood mononuclear cells from 3 rhesus macaques in response to TLR agonists, and demonstrated that the profiles are consistent among animals. Furthermore, because the IFN expression profiles differ depending on the TLR stimuli, they suggest different biological functions for many of the IFN species measured, including individual subtypes of IFN-α.


Assuntos
Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Interferons/biossíntese , Reação em Cadeia da Polimerase em Tempo Real/métodos , Animais , DNA Complementar/genética , Interferons/genética , Macaca mulatta
13.
Immunol Cell Biol ; 90(8): 774-83, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22249201

RESUMO

Recent genome-wide association studies suggest distinct roles for 12 human interferon-alpha (IFN-α) and 3 IFN-λ subtypes that may be elucidated by defining the expression patterns of these sets of genes. To overcome the impediment of high homology among each of the sets, we designed a quantitative real-time PCR assay that incorporates the use of molecular beacon and locked nucleic acid (LNA) probes, and in some instances, LNA oligonucleotide inhibitors. We then measured IFN subtype expression by human peripheral blood mononuclear cells and by purified monocytes, myeloid dendritic cells (mDC), plasmacytoid dendritic cells (pDC), and monocyte-derived macrophages (MDM), and -dendritic cells (MDDC) in response to poly I:C, lipopolysaccharide (LPS), imiquimod and CpG oligonucleotides. We found that in response to poly I:C and LPS, monocytes, MDM and MDDC express a subtype pattern restricted primarily to IFN-ß and IFN-λ1. In addition, while CpG elicited expression of all type I IFN subtypes by pDC, imiquimod did not. Furthermore, MDM and mDC highly express IFN-λ, and the subtypes of IFN-λ are expressed hierarchically in the order IFN-λ1 followed by IFN-λ2, and then IFN-λ3. These data support a model of coordinated cell- and ligand-specific expression of types I and III IFN. Defining IFN subtype expression profiles in a variety of contexts may elucidate specific roles for IFN subtypes as protective, therapeutic or pathogenic mediators.


Assuntos
Perfilação da Expressão Gênica , Interferon-alfa/genética , Interleucinas/genética , Animais , Sondas de DNA/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon-alfa/metabolismo , Interferons , Interleucinas/metabolismo , Ligantes , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , Poli I-C/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reprodutibilidade dos Testes , Receptores Toll-Like/metabolismo
14.
J Immunol ; 187(5): 2540-7, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21784976

RESUMO

Mycobacterium tuberculosis is a virulent intracellular pathogen that survives in macrophages even in the presence of an intact adaptive immune response. Type I IFNs have been shown to exacerbate tuberculosis in mice and to be associated with disease progression in infected humans. Nevertheless, the mechanisms by which type I IFNs regulate the host response to M. tuberculosis infection are poorly understood. In this study, we show that M. tuberculosis induces an IFN-related gene expression signature in infected primary human macrophages, which is dependent on host type I IFN signaling as well as the mycobacterial virulence factor, region of difference-1. We further demonstrate that type I IFNs selectively limit the production of IL-1ß, a critical mediator of immunity to M. tuberculosis. This regulation occurs at the level of IL1B mRNA expression, rather than caspase-1 activation or autocrine IL-1 amplification and appears to be preferentially used by virulent mycobacteria since avirulent M. bovis bacillus Calmette-Guérin (BCG) fails to trigger significant expression of type I IFNs or release of mature IL-1ß protein. The latter property is associated with decreased caspase-1-dependent IL-1ß maturation in the BCG-infected macrophages. Interestingly, human monocytes in contrast to macrophages produce comparable levels of IL-1ß in response to either M. tuberculosis or BCG. Taken together, these findings demonstrate that virulent and avirulent mycobacteria employ distinct pathways for regulating IL-1ß production in human macrophages and reveal that in the case of M. tuberculosis infection the induction of type I IFNs is a major mechanism used for this purpose.


Assuntos
Interferon Tipo I/imunologia , Interleucina-1beta/biossíntese , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Transdução de Sinais/imunologia , Western Blotting , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Regulação da Expressão Gênica/imunologia , Humanos , Interferon Tipo I/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
PLoS One ; 5(11): e15017, 2010 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-21124776

RESUMO

BACKGROUND: Human respiratory syncytial virus (HRSV), and to a lesser extent human metapneumovirus (HMPV) and human parainfluenza virus type 3 (HPIV3), re-infect symptomatically throughout life without antigenic change, suggestive of incomplete immunity. One causative factor is thought to be viral interference with dendritic cell (DC)-mediated stimulation of CD4+ T cells. METHODOLOGY, PRINCIPAL FINDINGS: We infected human monocyte-derived DC with purified HRSV, HMPV, HPIV3, or influenza A virus (IAV) and compared their ability to induce activation and proliferation of autologous CD4+ T cells in vitro. IAV was included because symptomatic re-infection without antigenic change is less frequent, suggesting that immune protection is more complete and durable. We examined virus-specific memory responses and superantigen-induced responses by multiparameter flow cytometry. Live virus was more stimulatory than inactivated virus in inducing DC-mediated proliferation of virus-specific memory CD4+ T cells, suggesting a lack of strong suppression by live virus. There were trends of increasing proliferation in the order: HMPV

Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Metapneumovirus/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Proliferação de Células , Células Cultivadas , Chlorocebus aethiops , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/virologia , Citometria de Fluxo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Vírus da Influenza A Subtipo H3N2/fisiologia , Interferon gama/metabolismo , Ativação Linfocitária/imunologia , Metapneumovirus/fisiologia , Monócitos/citologia , Vírus da Parainfluenza 3 Humana/fisiologia , Vírus Sincicial Respiratório Humano/fisiologia , Especificidade da Espécie , Fator de Necrose Tumoral alfa/metabolismo , Células Vero
16.
Ann Pharmacother ; 43(5): 967-72, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19417117

RESUMO

OBJECTIVE: To report a case of probable anaphylaxis due to anakinra in a patient with rheumatoid arthritis and multiple drug allergies. CASE SUMMARY: A 46-year-old Indian female with rheumatoid arthritis demonstrated distinct adverse reactions to all commercially available anti-tumor necrosis factor therapies, sulfasalazine, and hydroxychloroquine. Over a 4-year period her disease remained active during therapy with methotrexate and prednisone. Biologics were added sequentially, with development of intolerable reactions, first to infliximab (urticarial rash, infusion reactions) after 3 doses, and then to etanercept (autoantibodies, worsening Raynaud's phenomenon, digital microinfarcts) after 1 year. Following 2 months of daily injections of anakinra, she experienced an immediate immunoglobulin E-mediated anaphylactic reaction within 20 minutes of an injection, as evidenced by positive testing to both anakinra and histamine with the skin prick method. The patient subsequently started adalimumab therapy, which was discontinued after the fourth dose due to the development of generalized hives. DISCUSSION: The Naranjo probability scale demonstrated a probable relationship between anaphylaxis and anakinra in this patient. Although cases of anakinra-related hypersensitivity have been reported in patients in which therapy was interrupted and then reintroduced, to our knowledge, this is the first report of anaphylaxis with continuous therapy. CONCLUSIONS: This unusual case of a patient with multiple drug allergies presents a difficult clinical scenario, which was unsuccessfully managed with multiple biologic therapies on a trial-and-error basis. In the future, pharmacogenetics may help to better identify individuals at risk for multiple drug reactions and preclude unnecessary exposure to potentially harmful therapeutic options in similar patients.


Assuntos
Anafilaxia/induzido quimicamente , Antirreumáticos/efeitos adversos , Artrite Reumatoide/tratamento farmacológico , Hipersensibilidade a Drogas/etiologia , Proteína Antagonista do Receptor de Interleucina 1/efeitos adversos , Artrite Reumatoide/imunologia , Feminino , Humanos , Pessoa de Meia-Idade
17.
Virology ; 385(1): 169-82, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19128816

RESUMO

Human respiratory syncytial virus (HRSV), human metapneumovirus (HMPV), and human parainfluenza virus type 3 (HPIV3) are common, important respiratory pathogens, but HRSV has a substantially greater impact with regard to acute disease, long-term effects on airway function, and frequency of re-infection. It has been reported to strongly interfere with the functioning of dendritic cells (DC). We compared HRSV to HMPV and HPIV3 with regard to their effects on human monocyte-derived immature DC (IDC). Side-by-side analysis distinguished between common effects versus those specific to individual viruses. The use of GFP-expressing viruses yielded clear identification of robustly infected cells and provided the means to distinguish between direct effects of robust viral gene expression versus bystander effects. All three viruses infected inefficiently based on GFP expression, with considerable donor-to donor-variability. The GFP-negative cells exhibited low, abortive levels of viral RNA synthesis. The three viruses induced low-to-moderate levels of DC maturation and cytokine/chemokine responses, increasing slightly in the order HRSV, HMPV, and HPIV3. Infection at the individual cell level was relatively benign, such that in general GFP-positive cells were neither more nor less able to mature compared to GFP-negative bystanders, and cells were responsive to a secondary treatment with lipopolysaccharide, indicating that the ability to mature was not impaired. However, there was a single exception, namely that HPIV3 down-regulated CD38 expression at the RNA level. Maturation by these viruses was anti-apoptotic. Inefficient infection of IDC and sub-optimal maturation might result in reduced immune responses, but these effects would be common to all three viruses rather than specific to HRSV.


Assuntos
Células Dendríticas/virologia , Metapneumovirus/fisiologia , Vírus da Parainfluenza 3 Humana/fisiologia , Infecções por Paramyxoviridae/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/fisiologia , Infecções por Respirovirus/virologia , ADP-Ribosil Ciclase 1/metabolismo , Adulto , Animais , Apoptose , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Citocinas/metabolismo , Células Dendríticas/citologia , Regulação da Expressão Gênica , Humanos , Monócitos/imunologia , Monócitos/virologia , RNA Viral/metabolismo , Células Vero , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Proteínas Virais de Fusão/metabolismo
18.
J Leukoc Biol ; 79(6): 1328-38, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16614257

RESUMO

We report a novel mechanism, involving up-regulation of the interleukin (IL)-7 cytokine receptor, by which human immunodeficiency virus (HIV) enhances its own production in monocyte-derived macrophages (MDM) in vitro. HIV-1 infection or treatment of MDM cultures with exogenous HIV-1 Tat(86) protein up-regulates the IL-7 receptor (IL-7R) alpha-chain at the levels of steady-state RNA, protein, and functional IL-7R on the cell surface (as measured by ligand-induced receptor signaling). This IL-7R up-regulation is associated with increased amounts of HIV-1 virions in the supernatants of infected MDM cultures treated with exogenous IL-7 cytokine. The overall effect of IL-7 stimulation on HIV replication in MDM culture supernatants is typically in the range of one log and greater. The results are consistent with a model in which HIV infection produces the Tat protein, which in turn up-regulates IL-7R in a paracrine manner. This results in increased IL-7R signaling in response to the IL-7 cytokine, which ultimately promotes early events in HIV replication, including binding/entry and possibly other steps prior to reverse transcription. The results suggest that the effects of IL-7 on HIV replication in MDM should be considered when analyzing and designing clinical trials involving treatment of patients with IL-7 or Tat vaccines.


Assuntos
Produtos do Gene tat/fisiologia , HIV-1/fisiologia , Interleucina-7/fisiologia , Macrófagos/virologia , Modelos Biológicos , Replicação Viral/fisiologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Células Cultivadas/virologia , Genes tat , Transcriptase Reversa do HIV/metabolismo , Humanos , Interleucina-7/efeitos adversos , Interleucina-7/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Comunicação Parácrina , Fator de Transcrição STAT3/metabolismo , Vírion , Replicação Viral/efeitos dos fármacos , Produtos do Gene tat do Vírus da Imunodeficiência Humana
19.
Int Immunol ; 18(3): 485-93, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16481346

RESUMO

Multidrug resistance protein-1 (MRP1) belongs to subfamily C of the ATP-binding cassette transporters, and exports leukotriene C(4) and organic anions including the fluorescent calcium indicator indo-1. The observation that leukocytes from patients with an autoimmune disease exported indo-1 at a higher rate than controls prompted the hypothesis that MRP1 contributes to the function of activated cells. To test this, we defined the expression of MRP1 on resting and activated human T cells, and determined whether T cell activation is dependent upon MRP1 function. MRP1 is expressed on resting memory but not on naive CD4 and CD8 T cells. After activation through the TCR, cord blood CD4 T cells express high levels of MRP1. Blockade of MRP1 with the specific inhibitor MK-571 abrogated superantigen-induced expression of IFN-gamma, tumor necrosis factor-alpha, IL-10, IL-2, IL-4 and CD69 by T cells without affecting their viability, and was reversible upon removal of MK-571 from the culture media. Electrophoretic mobility shift assays demonstrate that MRP1 blockade with MK-571 induces activation of the transcriptional repressor peroxisome proliferator-activated receptor-gamma in CD4 T cells, thus providing insight into the potential mechanism by which their responses are abrogated.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/imunologia , Citocinas/metabolismo , Linfócitos T/imunologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Sobrevivência Celular , Humanos , Interferon gama/metabolismo , Ativação Linfocitária , Propionatos/farmacologia , Quinolinas/farmacologia , Linfócitos T/efeitos dos fármacos , Regulação para Cima
20.
Virology ; 332(2): 491-7, 2005 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-15680414

RESUMO

Natural killer (NK) cells play an important role in both innate and adaptive antiviral immune responses. The adaptive response typically requires that virus-specific antibodies decorate infected cells which then direct NK cell lysis through a CD16 mediated process termed antibody-dependent cellular cytotoxicity (ADCC). In this report, we employ a highly polymerized chimeric IgG1/IgA immunoglobulin (Ig) fusion protein that, by virtue of its capacity to extensively crosslink CD16, activates NK cells while directing the lysis of infected target cells. We employ HIV as a model system, and demonstrate that freshly isolated NK cells preloaded with an HIV gp120-specific chimeric IgG1/IgA fusion protein efficiently lyse HIV-infected target cells at picomolar concentrations. NK cells pre-armed in this manner retain the capacity to kill targets over an extended period of time. This strategy may have application to other disease states including various viral infections and cancers.


Assuntos
Infecções por HIV/imunologia , Células Matadoras Naturais/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Antígenos CD/imunologia , Sinalização do Cálcio/fisiologia , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Imunoglobulina G/imunologia , Imunoterapia/métodos , Receptores de IgG/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA