Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Hematol Oncol ; 13(1): 100, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32703253

RESUMO

Both inflammasomes and autophagy have important roles in the intracellular homeostasis, inflammation, and pathology; the dysregulation of these processes is often associated with the pathogenesis of numerous cancers. In addition, they can crosstalk with each other in multifaceted ways to influence various physiological and pathological responses, including cancer. Multiple molecular mechanisms connect the autophagy pathway to inflammasome activation and, through this, may influence the outcome of pro-tumor or anti-tumor responses depending on the cancer types, microenvironment, and the disease stage. In this review, we highlight the rapidly growing literature on the various mechanisms by which autophagy interacts with the inflammasome pathway, to encourage additional applications in the context of tumors. In addition, we provide insight into the mechanisms by which pathogen modulates the autophagy-inflammasome pathway to favor the infection-induced carcinogenesis. We also explore the challenges and opportunities of using multiple small molecules/agents to target the autophagy/inflammasome axis and their effects upon cancer treatment. Finally, we discuss the emerging clinical efforts assessing the potential usefulness of targeting approaches for either autophagy or inflammasome as anti-cancer strategies, although it remains underexplored in terms of their crosstalks.


Assuntos
Autofagia/fisiologia , Inflamassomos/fisiologia , Neoplasias/imunologia , Animais , Autofagia/efeitos dos fármacos , Ensaios Clínicos como Assunto , Homeostase , Humanos , Inflamassomos/efeitos dos fármacos , Membranas Intracelulares/fisiologia , Mitocôndrias/metabolismo , Mitofagia/efeitos dos fármacos , Mitofagia/fisiologia , Modelos Biológicos , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias/fisiopatologia , RNA de Cadeia Dupla/fisiologia , RNA Neoplásico/fisiologia , Espécies Reativas de Oxigênio/metabolismo
2.
J Immunol ; 203(9): 2508-2519, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31548332

RESUMO

IFN responses to viral infection are necessary to establish intrinsic antiviral state, but if unchecked can lead to heightened inflammation. Recently, we showed that TLR2 activation contributes to limitation of rhinovirus (RV)-induced IFN response in the airway epithelial cells. We also demonstrated that compared with normal airway epithelial cells, those from patients with chronic obstructive pulmonary disease (COPD) show higher IFN responses to RV, but the underlying mechanisms are not known. Initially, RV-induced IFN responses depend on dsRNA receptor activation and then are amplified via IFN-stimulated activation of JAK/STAT signaling. In this study, we show that in normal cells, TLR2 limits RV-induced IFN responses by attenuating STAT1 and STAT2 phosphorylation and this was associated with TLR2-dependent SIRT-1 expression. Further, inhibition of SIRT-1 enhanced RV-induced IFN responses, and this was accompanied by increased STAT1/STAT2 phosphorylation, indicating that TLR2 may limit RV-induced IFN responses via SIRT-1. COPD airway epithelial cells showed attenuated IL-8 responses to TLR2 agonist despite expressing TLR2 similar to normal, indicating dysregulation in TLR2 signaling pathway. Unlike normal, COPD cells failed to show RV-induced TLR2-dependent SIRT-1 expression. Pretreatment with quercetin, which increases SIRT-1 expression, normalized RV-induced IFN levels in COPD airway epithelial cells. Inhibition of SIRT-1 in quercetin-pretreated COPD cells abolished the normalizing effects of quercetin on RV-induced IFN expression in these cells, confirming that quercetin exerts its effect via SIRT-1. In summary, we show that TLR2 is required for limiting RV-induced IFNs, and this pathway is dysregulated in COPD airway epithelial cells, leading to exaggerated IFN production.


Assuntos
Brônquios/imunologia , Interferons/biossíntese , Doença Pulmonar Obstrutiva Crônica/etiologia , Rhinovirus/patogenicidade , Sirtuína 1/fisiologia , Receptor 2 Toll-Like/fisiologia , Células Cultivadas , Células Epiteliais , Humanos , Helicase IFIH1 Induzida por Interferon/fisiologia , Doença Pulmonar Obstrutiva Crônica/imunologia , RNA de Cadeia Dupla/fisiologia , Fatores de Transcrição STAT/fisiologia , Transdução de Sinais/fisiologia , Sirtuína 1/genética , Proteína 1 Supressora da Sinalização de Citocina/fisiologia
3.
Nat Commun ; 10(1): 2811, 2019 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-31243280

RESUMO

How developmental programs reactivate in regeneration is a fundamental question in biology. We addressed this question through the study of Wound Induced Hair follicle Neogenesis (WIHN), an adult organogenesis model where stem cells regenerate de novo hair follicles following deep wounding. The exact mechanism is uncertain. Here we show that self-noncoding dsRNA activates the anti-viral receptor toll like receptor 3 (TLR3) to induce intrinsic retinoic acid (RA) synthesis in a pattern that predicts new hair follicle formation after wounding in mice. Additionally, in humans, rejuvenation lasers induce gene expression signatures for dsRNA and RA, with measurable increases in intrinsic RA synthesis. These results demonstrate a potent stimulus for RA synthesis by non-coding dsRNA, relevant to their broad functions in development and immunity.


Assuntos
Folículo Piloso/fisiologia , RNA de Cadeia Dupla/fisiologia , Regeneração/fisiologia , Receptor 3 Toll-Like/metabolismo , Tretinoína/metabolismo , Animais , Benzoatos/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Cabelo/crescimento & desenvolvimento , Humanos , Interleucina-6/administração & dosagem , Interleucina-6/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Interferência de RNA , RNA Interferente Pequeno , Estilbenos/farmacologia , Cicatrização
4.
PLoS Biol ; 16(11): e2006577, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30496178

RESUMO

The interferon (IFN)-mediated innate immune response is the first line of defense against viruses. However, an IFN-stimulated gene, the adenosine deaminase acting on RNA 1 (ADAR1), favors the replication of several viruses. ADAR1 binds double-stranded RNA and converts adenosine to inosine by deamination. This form of editing makes duplex RNA unstable, thereby preventing IFN induction. To better understand how ADAR1 works at the cellular level, we generated cell lines that express exclusively either the IFN-inducible, cytoplasmic isoform ADAR1p150, the constitutively expressed nuclear isoform ADAR1p110, or no isoform. By comparing the transcriptome of these cell lines, we identified more than 150 polymerase II transcripts that are extensively edited, and we attributed most editing events to ADAR1p150. Editing is focused on inverted transposable elements, located mainly within introns and untranslated regions, and predicted to form duplex RNA structures. Editing of these elements occurs also in primary human samples, and there is evidence for cross-species evolutionary conservation of editing patterns in primates and, to a lesser extent, in rodents. Whereas ADAR1p150 rarely edits tightly encapsidated standard measles virus (MeV) genomes, it efficiently edits genomes with inverted repeats accidentally generated by a mutant MeV. We also show that immune activation occurs in fully ADAR1-deficient (ADAR1KO) cells, restricting virus growth, and that complementation of these cells with ADAR1p150 rescues virus growth and suppresses innate immunity activation. Finally, by knocking out either protein kinase R (PKR) or mitochondrial antiviral signaling protein (MAVS)-another protein controlling the response to duplex RNA-in ADAR1KO cells, we show that PKR activation elicits a stronger antiviral response. Thus, ADAR1 prevents innate immunity activation by cellular transcripts that include extensive duplex RNA structures. The trade-off is that viruses take advantage of ADAR1 to elude innate immunity control.


Assuntos
Adenosina Desaminase/fisiologia , Vírus de RNA/genética , Proteínas de Ligação a RNA/fisiologia , Adenosina/metabolismo , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Células HeLa , Humanos , Imunidade Inata/fisiologia , Interferons/metabolismo , Isoformas de Proteínas , Provírus/genética , Provírus/imunologia , Vírus de RNA/metabolismo , RNA de Cadeia Dupla/fisiologia , RNA Viral/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Transcriptoma/genética , Vírion/genética
5.
J Gen Virol ; 99(9): 1199-1209, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30041712

RESUMO

Replication of the positive-strand RNA viruses generates double-stranded RNAs (dsRNAs) that are recognized by host pattern recognition receptors (PRRs) to trigger innate immune responses. Formation of the viral replication complex (RC) has been thought to shield dsRNA from being recognized by innate sensors. To elucidate the RC-mediated evasion of innate recognition, we selected poliovirus (PV) as a model. We first found that RNAs generated during PV replication were potent interferon (IFN) inducers upon transfection, while there was no obvious IFN production detected in PV-replicating cells. PV replication did not interfere with IFN production when IFN agonists were synchronously introduced with the replicating PV RNAs, and in PV-infected cells, IFN agonist-induced IFN production was only moderately impaired but not completely abolished. When PV-infected cells were in situ permeabilized by digitonin, viral dsRNAs were readily detected by an anti-dsRNA antibody and were resistant to RNase III digestion. When digitonin-permeabilized cells were further solubilized by 1 % triton X-100, the dsRNAs of PV became sensitive to RNase III digestion. A co-localization study showed that PV dsRNA did not co-localize with MDA5 in virally infected cells. Given that the PV replication complex is protruding single-membrane and tubular in form, viral replicative dsRNAs are probably shielded by the replication complex or the viral replicase to avoid being accessed by RNase III and MDA5. We propose that the replication complex- or replicase-mediated shielding of dsRNA may act as a means for innate evasion.


Assuntos
Helicase IFIH1 Induzida por Interferon/metabolismo , Poliovirus/fisiologia , RNA de Cadeia Dupla/fisiologia , RNA Polimerase Dependente de RNA/metabolismo , Replicação Viral/fisiologia , Linhagem Celular , Regulação Viral da Expressão Gênica , Humanos , Poliovirus/genética , Transporte Proteico , RNA Viral
6.
Mol Plant Pathol ; 19(4): 883-895, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28621835

RESUMO

Zucchini yellow mosaic virus (ZYMV) causes serious damage in a large number of cucurbits, and control measures are necessary. Transgenic cucurbits expressing parts of the ZYMV genome have been shown to be resistant to the cognate virus. A non-transgenic approach involving the exogenous application of double-stranded RNA (dsRNA) has also been shown to induce resistance in tobacco against Cucumber mosaic virus (CMV) and Tobacco mosaic virus (TMV). In the present study, dsRNA molecules derived from the helper component-proteinase (HC-Pro) and coat protein (CP) genes of the ZYMV_DE_2014 isolate were produced in vitro. On exogenous dsRNA application in cucumber, watermelon and squash plants, dsRNA HC-Pro conferred resistance of 82%, 50% and 18%, and dsRNA CP molecules of 70%, 43% and 16%, respectively. On deep sequencing analysis of ZYMV-infected watermelon, hot-spot regions for viral small interfering RNAs (vsiRNAs) in the genome of ZYMV were identified. Stem-loop reverse transcription-polymerase chain reaction (RT-PCR) detection of selected 21-nucleotide-long vsiRNAs in plants that received only dsRNA molecules suggested that the dsRNAs exogenously applied onto plants were successfully diced, thus initiating RNA silencing. dsRNA molecules were found to be progressively degraded in planta, and strongly detected by semi-quantitative RT-PCR for at least 9 days after exogenous application. Moreover, dsRNA molecules were detected in systemic tissue of watermelon and squash, showing that dsRNA is transported long distances in these plants.


Assuntos
Cucumovirus/genética , Genoma Viral/genética , Doenças das Plantas/virologia , Potyvirus/genética , Potyvirus/patogenicidade , RNA de Cadeia Dupla/genética , Citrullus/virologia , Cucumis sativus/virologia , Cucurbita/virologia , RNA de Cadeia Dupla/fisiologia
7.
J Exp Clin Cancer Res ; 35: 53, 2016 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-27012825

RESUMO

BACKGROUND: Previous study showed that dsP53-285 has the capacity to induce tumor suppressor gene p53 expression by targeting promoter in non-human primates' cells. And it is well known that TP53 gene is frequently mutant or inactivated in human bladder cancer. Hereby, whether this small RNA can activate the expression of wild-type p53 and inhibit human bladder cancer cells remains to be elucidated. METHODS: Oligonucleotide and lentivirus were used to overexpress dsP53-285 and dsControl. Real-time PCR and western blot were used to detect genes' mRNA and protein expression, respectively. Cell proliferation assay, colony formation, flow cytometry, transwell assay and wound healing assay were performed to determine the effects on bladder cancer cells proliferation and migration/invasion in vitro. Animal models were carried out to analyze the effects on cells growth and metastasis in vivo. RESULTS: Transfection of dsP53-285 into human bladder cancer cell lines T24 and EJ readily activate wild-type p53 expression by targeting promoter. Moreover, dsP53-285 exhibited robust capacity to inhibit cells proliferation and colony formation, induce cells G0/G1 arrest, suppress migration and invasion. Besides, the Cyclin-CDK genes (Cyclin D1 and CDK4/6) were down-regulated and the EMT-associated genes (E-cadherin, ß-catenin, ZEB1 and Vimentin) were also expressed inversely after dsP53-285 treatment. In addition, dsP53-285 could also significantly suppress the growth of bladder cancer xenografts and metastasis in nude mice. Most importantly, the anti-tumor effects mediated by dsP53-285 were mainly achieved by manipulating wild-type p53 expression. CONCLUSION: Our findings indicate that the dsP53-285 can upregulate wild-type p53 expression in human bladder cancer cells through RNA activation, and suppresses cells proliferation and metastasis in vitro and in vivo.


Assuntos
Regiões Promotoras Genéticas/efeitos dos fármacos , RNA de Cadeia Dupla/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Neoplasias da Bexiga Urinária/terapia , Animais , Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Lentivirus/genética , Camundongos , Camundongos Nus , Metástase Neoplásica , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
RNA ; 20(1): 61-75, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24249224

RESUMO

Functional microRNAs (miRNAs) are produced from both arms of their precursors (pre-miRNAs). Their abundances vary in context-dependent fashion spatiotemporarily and there is mounting evidence of regulatory interplay between them. Here, we introduce chemically synthesized pre-miRNAs (syn-pre-miRNAs) as a general class of accessible, easily transfectable mimics of pre-miRNAs. These are RNA hairpins, identical in sequence to natural pre-miRNAs. They differ from commercially available miRNA mimics through their complete hairpin structure, including any regulatory elements in their terminal-loop regions and their potential to introduce both strands into RISC. They are distinguished from transcribed pre-miRNAs by their terminal 5' hydroxyl groups and their precisely defined terminal nucleotides. We demonstrate with several examples how they fully recapitulate the properties of pre-miRNAs, including their processing by Dicer into functionally active 5p; and 3p-derived mature miRNAs. We use syn-pre-miRNAs to show that miR-34a uses its 5p and 3p miRNAs in two pathways: apoptosis during TGF-ß signaling, where SIRT1 and SP4 are suppressed by miR-34a-5p and miR-34a-3p, respectively; and the lipopolysaccharide (LPS)-activation of primary human monocyte-derived macrophages, where TNF (TNFα) is suppressed by miR-34a-5p indirectly and miR-34a-3p directly. Our results add to growing evidence that the use of both arms of a miRNA may be a widely used mechanism. We further suggest that syn-pre-miRNAs are ideal and affordable tools to investigate these mechanisms.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/fisiologia , RNA de Cadeia Dupla/fisiologia , Fator de Necrose Tumoral alfa/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Células Cultivadas , Células HeLa , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , MicroRNAs/síntese química , Precursores de RNA/síntese química , Precursores de RNA/fisiologia , RNA de Cadeia Dupla/síntese química , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/farmacologia
10.
J Sci Food Agric ; 93(3): 646-51, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23015382

RESUMO

BACKGROUND: Propolis is a bee product with various biological properties, including an antiviral activity when taken orally. However, its mechanisms at the cellular and molecular level are not well understood. RESULTS: We investigated the effect of propolis on antiviral signaling in A549 cells transfected with double-stranded RNA (dsRNA), a model for viral infection. Pretreatment of the cells with propolis inhibited poly I:C (synthetic dsRNA)-induced interferon (IFN)-ß expression. Propolis had no effect on the dsRNA-induced expression of RIG-I-like receptors (RLRs), which are known as intracellular viral RNA sensors. As to the effect on antiviral executor genes, propolis enhanced myxovirus resistance 1 (MX1) expression, whereas interferon-inducible gene 6-16 (G1P3) and 2'-5'-oligoadenylate synthetase (OAS) were unaffected. All of these genes belong to the IFN-inducible genes, suggesting that the effect of propolis on antiviral signaling is not necessarily mediated by the autocrine regulation by IFN-ß. Propolis pretreatment inhibited dsRNA-induced interleukin-8 (IL8) and CCL5 expression, and consequently lowered polymorphonuclear leukocyte (PMN) chemotactic activity in the cell-conditioned medium. CONCLUSION: Taken together, these results suggest that propolis may suppress excess inflammatory responses without affecting the innate immunity during viral infection.


Assuntos
Antivirais/farmacologia , Interferon beta/genética , Neutrófilos/efeitos dos fármacos , Própole/farmacologia , RNA de Cadeia Dupla/antagonistas & inibidores , Adenocarcinoma , Brasil , Linhagem Celular Tumoral , Quimiotaxia de Leucócito/efeitos dos fármacos , Meios de Cultivo Condicionados , Expressão Gênica/efeitos dos fármacos , Humanos , Interferon beta/antagonistas & inibidores , Neoplasias Pulmonares , Neutrófilos/fisiologia , Poli I-C/antagonistas & inibidores , Poli I-C/farmacologia , RNA de Cadeia Dupla/fisiologia , Transfecção
11.
Nucleic Acid Ther ; 22(3): 196-204, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22509928

RESUMO

Small interfering RNAs (siRNA) are able to activate the mammalian innate immune system depending on their structure, sequence, and method of delivery. The immunostimulatory activity of double-stranded RNA can be applied to antiviral and antitumor therapy. Here we identified a set of 19-bp RNA duplexes with 3-nucleotid overhangs in the 3' ends that display immunostimulating activity (here and after immunostimulating RNA, or isRNA) and studied their sequence/activity relationships. It was found that the introduction of substitutions in the middle part of the isRNA sequence (10-16 positions counting from the 5' end of strand 1) does not alter the antiproliferative activity, while substitutions in the 3' end region of isRNA substantially reduce it. isRNAs efficiently inhibit the proliferation of human oral epidermoid carcinoma cells [half-maximal inhibitory concentration (IC(50)) values varied from 10 to 100 nM]. Our research demonstrated that antiproliferative effects of isRNAs are related to cell growth arrest, rather than the induction of apoptosis. These isRNAs strongly stimulate the synthesis of interferon-α (IFN-α), and to a lesser extent the synthesis of tumor necrosis factor (TNF-α) and interleukin-6 (IL-6), in adherent peripheral blood mononuclear cells. An intravenous injection of isRNA/Lipofectamine complexes into C57BL mice increases IFN-α and IL-6 levels in the blood serum up to 15-fold and 3-fold, respectively, compared to the control mice. The results obtained clearly demonstrate the pronounced immunostimulatory and antiproliferative properties of the isRNAs under study. Hence, these short double-stranded RNAs can be considered as potential agents for the therapy of oncological and viral diseases.


Assuntos
Adjuvantes Imunológicos/farmacologia , RNA de Cadeia Dupla/fisiologia , Animais , Sequência de Bases , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/sangue , Citometria de Fluxo , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Bucais/patologia , RNA Interferente Pequeno/genética
12.
FEBS Lett ; 586(2): 196-203, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22209981

RESUMO

Viral double-stranded RNA (dsRNA) and its synthetic analog polyI:C are recognized via multiple pathways and induce the expression of genes related to inflammation. In the present study, we demonstrated the polyI:C-induced gene expression of the damage associated molecular pattern (DAMP) molecules S100A8 and S100A9, while other S100 genes were not affected. Cycloheximide and Brefeldin A treatment revealed both the expression of S100A8 and S100A9 as secondary response genes and the involvement of polyI:C-induced cytokines herein. Several type I and type III interferons such as IFNß, IL-20, IL-24, and IFNλ/IL-29 were expressed in response to polyI:C, however, they failed to induce S100A8 and S100A9 gene expression. These data indicate the involvement of the danger molecule S100A8/A9 in the resistance against viruses.


Assuntos
Cicloeximida/farmacologia , Expressão Gênica , RNA de Cadeia Dupla/fisiologia , Proteínas S100/genética , Fatores de Transcrição/fisiologia , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/genética , Imunidade Adaptativa/fisiologia , Brefeldina A/farmacologia , Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Herpes Simples/genética , Herpes Simples/metabolismo , Herpes Simples/patologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/patologia , Poli I-C/farmacologia , RNA de Cadeia Dupla/farmacologia , RNA Viral/genética , RNA Viral/fisiologia , Proteínas S100/metabolismo , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos
13.
Am J Respir Cell Mol Biol ; 46(6): 740-7, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21799119

RESUMO

Activation of innate immunity against viruses in the respiratory tracts affects the development of asthma. Most respiratory viruses generate double-stranded (ds)RNA during their replication. We recently showed that a low-dose administration of polyinosinic polycytidylic acid (poly IC), a mimetic of viral dsRNA, during allergen sensitization augments airway eosinophilia and hyperresponsiveness in mice via enhanced production of IL-13 from T cells. However, a phenotype of asthma under severer load of dsRNA remains unknown. d-galactosamine (d-GalN) is known as a strong sensitizer of poly IC. Mice were treated with poly IC plus d-GalN during allergen sensitization. A sublethal dose of poly IC/d-GalN augmented airway eosinophilia and CD4(+) T-cell accumulation in the lungs but not airway hyperresponsiveness. The augmented inflammation was associated with decreased IL-10 in the bronchoalveolar lavage fluid and decreased Foxp3(+) regulatory T cells in the lungs. Serum IL-6 was prominently higher in the mice treated with poly IC/d-GalN than in that with poly IC alone or d-GalN alone. Poly IC/d-GalN did not affect IL-17-producing T cells in the lungs. Poly IC/d-GalN failed to augment airway eosinophilia after anti-IL-10 receptor monoclonal antibody treatment during allergen challenge. Finally, anti-IL-6 receptor monoclonal antibody treatment before poly IC/d-GalN completely prevented the decrease of IL-10 and Foxp3(+) regulatory T cells and the augmentation of airway inflammation. These results indicate that enhanced production of IL-6 by poly IC/d-GalN induces the augmentation of allergic inflammation via suppression of Foxp3(+) regulatory T-cell/IL-10 axis. IL-6 may be a target for preventing asthma augmentation related to severe virus infection.


Assuntos
Fatores de Transcrição Forkhead/imunologia , Hipersensibilidade/imunologia , Inflamação/imunologia , Interleucina-10/imunologia , Interleucina-6/biossíntese , RNA de Cadeia Dupla/fisiologia , Linfócitos T/imunologia , Animais , Asma/imunologia , Citometria de Fluxo , Camundongos
14.
J Biol Chem ; 286(31): 27278-87, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21659521

RESUMO

RIG-I is a major innate immune sensor for viral infection, triggering an interferon (IFN)-mediated antiviral response upon cytosolic detection of viral RNA. Double-strandedness and 5'-terminal triphosphates were identified as motifs required to elicit optimal immunological signaling. However, very little is known about the response dynamics of the RIG-I pathway, which is crucial for the ability of the cell to react to diverse classes of viral RNA while maintaining self-tolerance. In the present study, we addressed the molecular mechanism of RIG-I signal detection and its translation into pathway activation. By employing highly quantitative methods, we could establish the length of the double-stranded RNA (dsRNA) to be the most critical determinant of response strength. Size exclusion chromatography and direct visualization in scanning force microscopy suggested that this was due to cooperative oligomerization of RIG-I along dsRNA. The initiation efficiency of this oligomerization process critically depended on the presence of high affinity motifs, like a 5'-triphosphate. It is noteworthy that for dsRNA longer than 200 bp, internal initiation could effectively compensate for a lack of terminal triphosphates. In summary, our data demonstrate a very flexible response behavior of the RIG-I pathway, in which sensing and integration of at least two distinct signals, initiation efficiency and double strand length, allow the host cell to mount an antiviral response that is tightly adjusted to the type of the detected signal, such as viral genomes, replication intermediates, or small by-products.


Assuntos
RNA Helicases DEAD-box/fisiologia , Imunidade Inata , Animais , Sequência de Bases , Linhagem Celular , Proteína DEAD-box 58 , Primers do DNA , Humanos , Camundongos , Microscopia de Força Atômica , Fosforilação , RNA de Cadeia Dupla/fisiologia , Receptores Imunológicos , Transdução de Sinais
15.
Sci China Life Sci ; 54(12): 1143-50, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22227907

RESUMO

In many eukaryotic organisms, Cdc14 phosphatase regulates multiple biological events during anaphase and is essential for mitosis. It has been shown that Cdc14 is required for sporulation in the potato blight pathogen Phytophthora infestans; however, the role that the Cdc14 homolog (PsCdc14) plays in the soil-borne soybean root rot pathogen P. sojae remains ambiguous. PsCdc14 is highly expressed in sporulation, zoospore, and cyst life stages, but not in vegetative mycelia and infection stages, suggesting that it contributes to asexual reproduction and thus the spread of the disease. Double-stranded RNA (dsRNA) mediates gene silencing, a post-transcriptional and highly conserved process in eukaryotes, involving specific gene silencing through degradation of target mRNA. We combined in vitro dsRNA synthesis and a polyethylene glycol-mediated transformation system to construct a dsRNA-mediated transient gene silencing system; and then performed a functional analysis of PsCdc14 in P. sojae. PsCdc14 mRNA was dramatically reduced in transformants after protoplasts were exposed in in vitro synthesized PsCdc14 dsRNA, resulting in low sporangial production and abnormal development in P. sojae silencing lines. Furthermore, dsRNA-mediated transient gene silencing could enable elucidation of P. sojae rapid gene function, facilitating our understanding of the development and pathogenicity mechanisms of this oomycete fungus.


Assuntos
Inativação Gênica , Glycine max/microbiologia , Monoéster Fosfórico Hidrolases/metabolismo , Phytophthora/fisiologia , Raízes de Plantas/microbiologia , RNA de Cadeia Dupla/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Primers do DNA , Humanos , Dados de Sequência Molecular , Monoéster Fosfórico Hidrolases/química , Reação em Cadeia da Polimerase , Homologia de Sequência de Aminoácidos
16.
J Parasitol ; 96(4): 815-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20476805

RESUMO

In many eukaryotes, the introduction of double-stranded RNA (dsRNA) into cells triggers the degradation of mRNAs through a post-transcriptional gene-silencing mechanism called RNA interference or RNAi. In the present study, we found that endogenous long-dsRNA was substantially more effective at producing interference than endogenous, or exogenous, short-dsRNA expression in Giardia lamblia . The effects of this interference were not evident in the highly expressed protein tubulin or the stage-specific cyst wall protein 2. However, long-dsRNA caused potent and specific interference in the medium subunits of adaptins, the RNA-dependent RNA polymerase, and the exogenous green fluorescence protein. Our results suggest that the ability of dsRNA antisense to inhibit the expression of these specific types of proteins is indicative of a gene-specific mechanism.


Assuntos
Regulação para Baixo/genética , Giardia lamblia/genética , Proteínas de Protozoários/metabolismo , Interferência de RNA/fisiologia , RNA de Cadeia Dupla/fisiologia , Subunidades do Complexo de Proteínas Adaptadoras/genética , Subunidades do Complexo de Proteínas Adaptadoras/metabolismo , Chaperona BiP do Retículo Endoplasmático , Técnica Direta de Fluorescência para Anticorpo , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica/genética , Giardia lamblia/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Microscopia Confocal , Proteínas de Protozoários/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
17.
Autophagy ; 6(1): 148-50, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19923903

RESUMO

Patients with metastatic melanoma have a poor prognosis, primarily due to a generalized inefficacy of current anticancer treatments. Therefore, the identification of novel death inducers with good bioavailability and safety profiles is a main priority in this disease. Here we summarize recent work from our group uncovering an unexpected ability of the dsRNA mimic polyinosine-polycytidylic acid (pIC) to engage the endo/lysosomal machinery of melanoma cells and induce their self degradation by autophagy and apoptosis, without noticeable secondary effects in vivo. However the antimelanoma activity of pIC strictly required conjugation with carriers (e.g., polyethyleneimine, PEI) for cytosolic delivery. Combining transcriptome analyses with RNA interference, we found RNA helicase MDA-5 as a main driver of the pIC-PEI complex. MDA-5 in turn, favored NOXA-dependent activation of apoptotic caspases. These results demonstrate new therapeutically tractable links between autophagy and apoptosis that can be coordinately engaged in tumor cells by dsRNA mimics.


Assuntos
Apoptose/genética , Autofagia/genética , Endossomos/metabolismo , Imunidade Inata/genética , Melanoma/imunologia , Fagossomos/metabolismo , RNA de Cadeia Dupla/fisiologia , Animais , Autofagia/imunologia , Autofagia/fisiologia , Citosol/metabolismo , Citosol/patologia , Endossomos/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/terapia , Modelos Biológicos , Fagossomos/genética , RNA de Cadeia Dupla/genética
18.
Nephrol Dial Transplant ; 24(11): 3312-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19608629

RESUMO

BACKGROUND: The molecular pathomechanisms by which viral infections trigger glomerulonephritis remain elusive. In the glomerulus, glomerular endothelial cells (GEnC) first interact with circulating viral particles; hence, we hypothesized that viral RNA, a known inducer of type I interferons and cytokines in dendritic cells, would also elicit proinflammatory antiviral reponses in GEnC. METHODS: Cultured murine GEnC were stimulated with poly I:C RNA and phenotype changes were assessed. Specific antagonists or s.i.RNA were used to determine the mechanisms of RNA uptake and the functional role of putative RNA receptors. RESULTS: Poly I:C RNA activated GEnC to produce IL-6, CCL2, CCL5, CXCL10, IFN-alpha and IFN-beta. This was independent of endosomal acidification or MyD88 but required complex formation with cationic lipids to be taken up into GEnC via clathrin-dependent endocytosis. RIG-1- but not MDA5-specific s.i.RNA prevented GEnC activation. Type I interferon production did not activate GEnC in an autocrine-paracrine manner. Complexed RNA also activated GEnC to express ICAM-1 and increased the albumin permeability of GEnC monolayers. CONCLUSIONS: Complexed dsRNA enters GEnC via clathrin endocytosis and activates GEnC via RIG-1 in the cytosol to produce inflammatory cytokines, chemokines and type I interferons. Furthermore, RNA induces ICAM-1 expression and increases GEnC permeability. All of these mechanisms may contribute to the onset or aggravation of glomerulonephritis associated with RNA virus infections.


Assuntos
RNA Helicases DEAD-box/fisiologia , Células Endoteliais/imunologia , Glomerulonefrite/etiologia , Interferon Tipo I/biossíntese , Glomérulos Renais/imunologia , Infecções por Vírus de RNA/complicações , RNA de Cadeia Dupla/fisiologia , Albuminas/metabolismo , Animais , Células Cultivadas , Proteína DEAD-box 58 , Endocitose , Molécula 1 de Adesão Intercelular/análise , Helicase IFIH1 Induzida por Interferon , Camundongos , Poli I-C/farmacologia , Receptores Toll-Like/fisiologia , Molécula 1 de Adesão de Célula Vascular/análise
19.
Int J Cancer ; 125(2): 446-52, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19384944

RESUMO

Small double stranded RNAs (dsRNA) are a new class of molecules which regulate gene expression. Accumulating data suggest that some dsRNA can function as tumor suppressors. Here, we report further evidence on the potential of dsRNA mediated p21 induction. Using the human renal cell carcinoma cell line A498, we found that dsRNA targeting the p21 promoter significantly induced the expression of p21 mRNA and protein levels. As a result, dsP21 transfected cells had a significant decrease in cell viability with a concomitant G1 arrest. We also observed a significant increase in apoptosis. These findings were associated with a significant decrease in survivin mRNA and protein levels. This is the first report that demonstrates dsRNA mediated gene activation in renal cell carcinoma and suggests that forced over-expression of p21 may lead to an increase in apoptosis through a survivin dependent mechanism.


Assuntos
Apoptose/genética , Carcinoma de Células Renais/patologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Fase G1/genética , Neoplasias Renais/patologia , RNA de Cadeia Dupla/fisiologia , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Primers do DNA , Citometria de Fluxo , Humanos , Proteínas Inibidoras de Apoptose , Proteínas Associadas aos Microtúbulos/genética , RNA Mensageiro/genética , Survivina
20.
Mol Cell Biol ; 29(6): 1487-97, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19124606

RESUMO

Double-stranded RNA (dsRNA)-binding proteins interact with substrate RNAs via dsRNA-binding domains (dsRBDs). Several proteins harboring these domains exhibit nucleocytoplasmic shuttling and possibly remain associated with their substrate RNAs bound in the nucleus during nuclear export. In the human RNA-editing enzyme ADAR1-c, the nuclear localization signal overlaps the third dsRBD, while the corresponding import factor is unknown. The protein also lacks a clear nuclear export signal but shuttles between the nucleus and the cytoplasm. Here we identify transportin-1 as the import receptor for ADAR1. Interestingly, dsRNA binding interferes with transportin-1 binding. At the same time, each of the dsRBDs in ADAR1 interacts with the export factor exportin-5. RNA binding stimulates this interaction but is not a prerequisite. Thus, our data demonstrate a role for some dsRBDs as RNA-sensitive nucleocytoplasmic transport signals. dsRBD3 in ADAR1 can mediate nuclear import, while interaction of all dsRBDs might control nuclear export. This finding may have implications for other proteins containing dsRBDs and suggests a selective nuclear export mechanism for substrates interacting with these proteins.


Assuntos
Adenosina Desaminase/metabolismo , Núcleo Celular/metabolismo , Carioferinas/metabolismo , RNA de Cadeia Dupla/fisiologia , Proteínas de Ligação a RNA/fisiologia , beta Carioferinas/metabolismo , Proteína ran de Ligação ao GTP/fisiologia , Transporte Ativo do Núcleo Celular/fisiologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Citoplasma/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Sinais de Localização Nuclear/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA