Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 96(18): e0077622, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36069546

RESUMO

The nonstructural protein 1 (NS1) of influenza A viruses is an important virulence factor that controls host cell immune responses. In human cells, NS1 proteins inhibit the induction of type I interferon by several mechanisms, including potentially, by preventing the activation of the retinoic acid-inducible gene I (RIG-I) receptor by the ubiquitin ligase tripartite motif-containing protein 25 (TRIM25). It is unclear whether the inhibition of human TRIM25 is a universal function of all influenza A NS1 proteins or is strain dependent. It is also unclear if NS1 proteins similarly target the TRIM25 of mallard ducks, a natural reservoir host of avian influenza viruses with a long coevolutionary history and unique disease dynamics. To answer these questions, we compared the ability of five different NS1 proteins to interact with human and duck TRIM25 using coimmunoprecipitation and microscopy and assessed the consequence of this on RIG-I ubiquitination and signaling in both species. We show that NS1 proteins from low-pathogenic and highly pathogenic avian influenza viruses potently inhibit RIG-I ubiquitination and reduce interferon promoter activity and interferon-beta protein secretion in transfected human cells, while the NS1 of the mouse-adapted PR8 strain does not. However, all the NS1 proteins, when cloned into recombinant viruses, suppress interferon in infected alveolar cells. In contrast, avian NS1 proteins do not suppress duck RIG-I ubiquitination and interferon promoter activity, despite interacting with duck TRIM25. IMPORTANCE Influenza A viruses are a major cause of human and animal disease. Periodically, avian influenza viruses from wild waterfowl, such as ducks, pass through intermediate agricultural hosts and emerge into the human population as zoonotic diseases with high mortality rates and epidemic potential. Because of their coevolution with influenza A viruses, ducks are uniquely resistant to influenza disease compared to other birds, animals, and humans. Here, we investigate a mechanism of influenza A virus interference in an important antiviral signaling pathway that is orthologous in humans and ducks. We show that NS1 proteins from four avian influenza strains can block the coactivation and signaling of the human RIG-I antiviral receptor, while none block the coactivation and signaling of duck RIG-I. Understanding host-pathogen dynamics in the natural reservoir will contribute to our understanding of viral disease mechanisms, viral evolution, and the pressures that drive it, which benefits global surveillance and outbreak prevention.


Assuntos
Proteínas Aviárias , Vírus da Influenza A , Influenza Aviária , Interferon beta , Receptores do Ácido Retinoico , Transdução de Sinais , Proteínas não Estruturais Virais , Animais , Antivirais/metabolismo , Proteínas Aviárias/metabolismo , Patos , Humanos , Vírus da Influenza A/genética , Interferon Tipo I/metabolismo , Interferon beta/metabolismo , Camundongos , Receptores do Ácido Retinoico/metabolismo , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitinação , Proteínas não Estruturais Virais/metabolismo
2.
Viruses ; 12(4)2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32272772

RESUMO

RIG-I plays an essential role in the duck innate immune response to influenza infection. RIG-I engages the critical adaptor protein mitochondrial antiviral signaling (MAVS) to activate the downstream signaling pathway. The influenza A virus non-structural protein PB1-F2 interacts with MAVS in human cells to inhibit interferon production. As duck and human MAVS share only 28% amino acid similarity, it is not known whether the influenza virus can similarly inhibit MAVS signaling in avian cells. Using confocal microscopy we show that MAVS and the constitutively active N-terminal end of duck RIG-I (2CARD) co-localize in DF-1 cells, and duck MAVS is pulled down with GST-2CARD. We establish that either GST-2CARD, or duck MAVS can initiate innate signaling in chicken cells and their co-transfection augments interferon-beta promoter activity. Demonstrating the limits of cross-species interactions, duck RIG-I 2CARD initiates MAVS signaling in chicken cells, but works poorly in human cells. The D122A mutation of human 2CARD abrogates signaling by affecting MAVS engagement, and the reciprocal A120D mutation in duck 2CARD improves signaling in human cells. We show mitochondrial localization of PB1-F2 from influenza A virus strain A/Puerto Rico/8/1934 (H1N1; PR8), and its co-localization and co-immunoprecipitation with duck MAVS. PB1-F2 inhibits interferon-beta promoter activity induced by overexpression of either duck RIG-I 2CARD, full-length duck RIG-I, or duck MAVS. Finally, we show that the effect of PB1-F2 on mitochondria abrogates TRIM25-mediated ubiquitination of RIG-I CARD in both human and avian cells, while an NS1 variant from the PR8 influenza virus strain does not.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/imunologia , Imunidade Inata , Vírus da Influenza A Subtipo H1N1/imunologia , Transdução de Sinais , Proteínas Virais/imunologia , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Galinhas , Patos/imunologia , Patos/virologia , Fibroblastos , Células HEK293 , Humanos , Mitocôndrias/imunologia , Receptores do Ácido Retinoico/imunologia , Receptores do Ácido Retinoico/metabolismo , Ubiquitinação , Proteínas Virais/genética
3.
Bioessays ; 38(5): 427-39, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26990286

RESUMO

We propose a biochemical mechanism for celiac disease and non-celiac gluten sensitivity that may rationalize many of the extradigestive disorders not explained by the current immunogenetic model. Our hypothesis is based on the homology between the 33-mer gliadin peptide and a component of the NMDA glutamate receptor ion channel - the human GRINA protein - using BLASTP software. Based on this homology the 33-mer may act as a natural antagonist interfering with the normal interactions of GRINA and its partners. The theory is supported by numerous independent data from the literature, and provides a mechanistic link with otherwise unrelated disorders, such as cleft lip and palate, thyroid dysfunction, restless legs syndrome, depression, ataxia, hearing loss, fibromyalgia, dermatitis herpetiformis, schizophrenia, toxoplasmosis, anemia, osteopenia, Fabry disease, Barret's adenocarcinoma, neuroblastoma, urinary incontinence, recurrent miscarriage, cardiac anomalies, reduced risk of breast cancer, stiff person syndrome, etc. The hypothesis also anticipates better animal models, and has the potential to open new avenues of research.


Assuntos
Doença Celíaca/metabolismo , Gliadina/metabolismo , Modelos Genéticos , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Ataxia/genética , Ataxia/metabolismo , Ataxia/patologia , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/metabolismo , Doenças Ósseas Metabólicas/patologia , Doença Celíaca/induzido quimicamente , Doença Celíaca/genética , Doença Celíaca/patologia , Fenda Labial/genética , Fenda Labial/metabolismo , Fenda Labial/patologia , Fissura Palatina/genética , Fissura Palatina/metabolismo , Fissura Palatina/patologia , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo , Dermatite Herpetiforme/genética , Dermatite Herpetiforme/metabolismo , Dermatite Herpetiforme/patologia , Regulação da Expressão Gênica , Gliadina/genética , Glutens/efeitos adversos , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Ligação Proteica , Multimerização Proteica , Proteínas/genética , Proteínas/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Tireoidite/genética , Tireoidite/metabolismo , Tireoidite/patologia , Transativadores
4.
Mol Immunol ; 67(2 Pt B): 607-15, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26254985

RESUMO

Wild waterfowl, including mallard ducks, are the natural reservoir of avian influenza A virus and they are resistant to strains that would cause fatal infection in chickens. Here we investigate potential involvement of TRIM proteins in the differential response of ducks and chickens to influenza. We examine a cluster of TRIM genes located on a single scaffold in the duck genome, which is a conserved synteny group with a TRIM cluster located in the extended MHC region in chickens and turkeys. We note a TRIM27-like gene is present in ducks, and absent in chickens and turkeys. Orthologous genes are predicted in many birds and reptiles, suggesting the gene has been lost in chickens and turkeys. Using quantitative real-time PCR (qPCR) we show that TRIM27-L, and the related TRIM27.1, are upregulated 5- and 9-fold at 1 day post-infection with highly pathogenic A/Vietnam/1203/2004. To assess whether TRIM27.1 or TRIM27-L are involved in modulation of antiviral gene expression, we overexpressed them in DF1 chicken cells, and neither show any direct effect on innate immune gene expression. However, when co-transfected with duck RIG-I-N (d2CARD) to constitutively activate the MAVS pathway, TRIM27.1 weakly decreases, while TRIM27-L strongly activates innate immune signaling leading to increased transcription of antiviral genes MX1 and IFN-ß. Furthermore, when both are co-expressed, the activation of the MAVS signaling pathway by TRIM27-L over-rides the inhibition by TRIM27.1. Thus, ducks have an activating TRIM27-L to augment MAVS signaling following RIG-I detection, while chickens lack both TRIM27-L and RIG-I itself.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Galinhas/imunologia , Proteínas de Ligação a DNA/metabolismo , Patos/imunologia , Transdução de Sinais , Perus/imunologia , Animais , Proteínas de Ligação a DNA/genética , Influenza Aviária/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Família Multigênica , Filogenia , RNA Helicases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação para Cima/genética
5.
PLoS One ; 9(1): e86968, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24466302

RESUMO

Retinoic acid inducible gene I (RIG-I) is a viral RNA sensor crucial in defense against several viruses including measles, influenza A and hepatitis C. RIG-I activates type-I interferon signalling through the adaptor for mitochondrial antiviral signaling (MAVS). The E3 ubiquitin ligase, tripartite motif containing protein 25 (TRIM25), activates human RIG-I through generation of anchored K63-linked polyubiquitin chains attached to lysine 172, or alternatively, through the generation of unanchored K63-linked polyubiquitin chains that interact non-covalently with RIG-I CARD domains. Previously, we identified RIG-I of ducks, of interest because ducks are the host and natural reservoir of influenza viruses, and showed it initiates innate immune signaling leading to production of interferon-beta (IFN-ß). We noted that K172 is not conserved in RIG-I of ducks and other avian species, or mouse. Because K172 is important for both mechanisms of activation of human RIG-I, we investigated whether duck RIG-I was activated by TRIM25, and if other residues were the sites for attachment of ubiquitin. Here we show duck RIG-I CARD domains are ubiquitinated for activation, and ubiquitination depends on interaction with TRIM25, as a splice variant that cannot interact with TRIM25 is not ubiquitinated, and cannot be activated. We expressed GST-fusion proteins of duck CARD domains and characterized TRIM25 modifications of CARD domains by mass spectrometry. We identified two sites that are ubiquitinated in duck CARD domains, K167 and K193, and detected K63 linked polyubiquitin chains. Site directed mutagenesis of each site alone, does not alter the ubiquitination profile of the duck CARD domains. However, mutation of both sites resulted in loss of all attached ubiquitin and polyubiquitin chains. Remarkably, the double mutant duck RIG-I CARD still interacts with TRIM25, and can still be activated. Our results demonstrate that anchored ubiquitin chains are not necessary for TRIM25 activation of duck RIG-I.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , RNA Helicases DEAD-box/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Processamento Alternativo/genética , Sequência de Aminoácidos , Animais , Western Blotting , Proteínas Adaptadoras de Sinalização CARD/genética , Células Cultivadas , Galinhas , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Patos/virologia , Humanos , Imunidade Inata , Imunoprecipitação , Vírus da Influenza A/fisiologia , Interferon beta/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação/genética , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores Imunológicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Espectrometria de Massas em Tandem , Fatores de Transcrição/genética , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
6.
Dev Comp Immunol ; 41(3): 377-88, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23624185

RESUMO

Birds have a smaller repertoire of immune genes than mammals. In our efforts to study antiviral responses to influenza in avian hosts, we have noted key genes that appear to be missing. As a result, we speculate that birds have impaired detection of viruses and intracellular pathogens. Birds are missing TLR8, a detector for single-stranded RNA. Chickens also lack RIG-I, the intracellular detector for single-stranded viral RNA. Riplet, an activator for RIG-I, is also missing in chickens. IRF3, the nuclear activator of interferon-beta in the RIG-I pathway is missing in birds. Downstream of interferon (IFN) signaling, some of the antiviral effectors are missing, including ISG15, and ISG54 and ISG56 (IFITs). Birds have only three antibody isotypes and IgD is missing. Ducks, but not chickens, make an unusual truncated IgY antibody that is missing the Fc fragment. Chickens have an expanded family of LILR leukocyte receptor genes, called CHIR genes, with hundreds of members, including several that encode IgY Fc receptors. Intriguingly, LILR homologues appear to be missing in ducks, including these IgY Fc receptors. The truncated IgY in ducks, and the duplicated IgY receptor genes in chickens may both have resulted from selective pressure by a pathogen on IgY FcR interactions. Birds have a minimal MHC, and the TAP transport and presentation of peptides on MHC class I is constrained, limiting function. Perhaps removing some constraint, ducks appear to lack tapasin, a chaperone involved in loading peptides on MHC class I. Finally, the absence of lymphotoxin-alpha and beta may account for the observed lack of lymph nodes in birds. As illustrated by these examples, the picture that emerges is some impairment of immune response to viruses in birds, either a cause or consequence of the host-pathogen arms race and long evolutionary relationship of birds and RNA viruses.


Assuntos
Proteínas Aviárias/deficiência , Galinhas/imunologia , Imunidade Inata , Imunoglobulina D/deficiência , Fatores Reguladores de Interferon/deficiência , Receptores Imunológicos/deficiência , Ubiquitina-Proteína Ligases/deficiência , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/imunologia , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Evolução Biológica , Galinhas/microbiologia , Galinhas/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Imunoglobulina D/genética , Imunoglobulina D/imunologia , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/imunologia , Mamíferos/imunologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
7.
J Steroid Biochem Mol Biol ; 105(1-5): 1-15, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17631997

RESUMO

Aromatase (CYP19) converts adrenal and ovarian androgens into estrogens, which supports the growth of estrogen-dependent breast cancers. Anti-aromatase agents are displacing antiestrogens as the first-line treatment for estrogen receptor positive breast cancers. Androgens can act as estrogen precursors, but besides this capability they can also directly act on breast cancer cells by binding to androgen receptors, which are present in the majority of breast cancer specimens. Epidemiological and clinical evidences suggest that higher levels of circulating androgen increase the risk of developing breast cancer. Androgen receptor gene polymorphisms which render the more transcriptionally active receptors have been related to a lower risk of breast cancer. It is currently accepted that androgens act as antiproliferative agents in the presence of estrogens in some breast cancer cell lines. However, emerging evidence suggests that direct androgenic activity might also stimulate cell growth in a subset of estrogen-resistant breast tumors. Here we discuss the supporting evidence which proposes that androgens themselves are actively involved in breast carcinogenesis and its clinical behaviour.


Assuntos
Androgênios/fisiologia , Neoplasias da Mama/fisiopatologia , Receptores Androgênicos/fisiologia , Progressão da Doença , Humanos , Polimorfismo Genético , Receptores Androgênicos/genética , Medição de Risco
8.
Pharmacol Toxicol ; 93(5): 238-43, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14629736

RESUMO

The protective effect of the 21-aminosteroid U-74389G was studied in an experimental model of partial ischaemia reperfusion liver injury. Previous studies have proven the remarkable potency of 21-aminosteroids to prevent oxidant-induced cell injury in vitro and in vivo. However, the capability of these compounds to limit oxidative injury in clinical trials has been considered to be less certain. The potential protective effect exerted by U-74389G on reduced and prolonged models of ischaemia reperfusion liver injury was studied in male rats subjected to 75 min. of segmentary hepatic ischaemia followed by 1 or 24 hr of reperfusion. Liver injury was evaluated by measuring serum levels of liver enzymes and by histopathological studies. The oxidative status of liver cells was measured by evaluating the levels of liver lipid peroxidation products such as malondialdehyde and the levels of reduced glutathione. Our results lead us to think that treatment with U-74389G (6 mg/kg) does not bring about any protective effect neither in the levels of transaminases nor in the percentage of hepatocellular necrosis and cellular infiltration observed in any reperfusion-period groups. In fact and in contrast with our expectations, U-74389G seemed to increase enzyme release. Furthermore, at the dose used, this 21-aminosteroid is not capable of inhibiting the lipoperoxidation processes, although it induced an important increase of GSH levels at any time-period of reperfusion. This last finding seem to suggest that U-74389G could increase the resistance to oxidant-induced liver tissue damage. However, our results show that, at the dose used, this compound did not exert any protective effect on liver tissue, thus explaining, at least partially, the absence of beneficial effects on the part of these compounds in clinical trials carried out to limit organ injury in transplants.


Assuntos
Antioxidantes/farmacologia , Isquemia/complicações , Fígado/irrigação sanguínea , Pregnatrienos/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/etiologia
9.
J Steroid Biochem Mol Biol ; 81(3): 191-201, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12163131

RESUMO

Prostate carcinoma is the most frequently diagnosed malignancy and the second leading cause of death as a result of cancer in men in the western countries. Withdrawal of androgens or the peripheral blockage of androgen action remain the critical therapeutic options for the treatment of advanced prostate cancer. However, after initial regression, most of the prostate cancers become androgen-independent and progress further, with eventual fatal outcome. Understanding the mechanisms of transition to androgen independence and tumor progression in prostate cancer is critical to finding new ways to treat aged patients that are ineligible for conventional chemotherapy. A large number of different molecular mechanisms might be responsible for the transition to androgen-independence. Many of these involve the androgen receptor (AR) and its signalling pathways, but they might also include genetic changes that affect several genes, which results in the activation of oncogenes or the inactivation of tumor suppressor genes. Here, we discuss the most recent and relevant findings on androgen resistance in prostate cancer in order provide a comprehensive interpretation of the clinical behaviour of tumors at molecular levels.


Assuntos
Androgênios/metabolismo , Neoplasias da Próstata/metabolismo , Dimerização , Éxons , Humanos , Masculino , Modelos Biológicos , Mutação , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transdução de Sinais , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...