Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 221(2)2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38095631

RESUMO

Toll-like receptors 7 (TLR7) and 8 (TLR8) each sense single-stranded RNA (ssRNA), but their activation results in different immune activation profiles. Attempts to selectively target either TLR7 or TLR8 have been hindered by their high degree of homology. However, recent studies revealed that TLR7 and TLR8 bind different ligands resulting from the processing of ssRNA by endolysosomal RNases. We demonstrate that by introducing precise 2' sugar-modified bases into oligoribonucleotides (ORNs) containing known TLR7 and TLR8 binding motifs, we could prevent RNase-mediated degradation into the monomeric uridine required for TLR8 activation while preserving TLR7 activation. Furthermore, a novel, optimized protocol for CRISPR-Cas9 knockout in primary human plasmacytoid dendritic cells showed that TLR7 activation is dependent on RNase processing of ORNs and revealed a previously undescribed role for RNase 6 in degrading ORNs into TLR ligands. Finally, 2' sugar-modified ORNs demonstrated robust innate immune activation in mice. Altogether, we identified a strategy for creating tunable TLR7-selective agonists.


Assuntos
Ribonucleases , Receptor 7 Toll-Like , Humanos , Camundongos , Animais , Receptor 7 Toll-Like/genética , Nucleotídeos , Receptor 8 Toll-Like/genética , Ligantes , RNA , Adjuvantes Imunológicos , Açúcares
3.
J Exp Med ; 218(9)2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34297039

RESUMO

Cytosolic double-stranded RNA (dsRNA) initiates type I IFN responses. Endogenous retroelements, notably Alu elements, constitute a source of dsRNA. Adenosine-to-inosine (A-to-I) editing by ADAR induces mismatches in dsRNA and prevents recognition by MDA5 and autoinflammation. To identify additional endogenous dsRNA checkpoints, we conducted a candidate screen in THP-1 monocytes and found that hnRNPC and ADAR deficiency resulted in synergistic induction of MDA5-dependent IFN responses. RNA-seq analysis demonstrated dysregulation of Alu-containing introns in hnRNPC-deficient cells via utilization of unmasked cryptic splice sites, including introns containing ADAR-dependent A-to-I editing clusters. These putative MDA5 ligands showed reduced editing in the absence of ADAR, providing a plausible mechanism for the combined effects of hnRNPC and ADAR. This study contributes to our understanding of the control of repetitive element-induced autoinflammation and suggests that patients with hnRNPC-mutated tumors might maximally benefit from ADAR inhibition-based immunotherapy.


Assuntos
Adenosina Desaminase/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo C/genética , Interferon Tipo I/genética , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/genética , Adenosina Desaminase/metabolismo , Elementos Alu , Sistemas CRISPR-Cas , Citosol/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo C/metabolismo , Humanos , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/metabolismo , Íntrons , Células MCF-7 , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Edição de RNA , Proteínas de Ligação a RNA/metabolismo , Células THP-1
4.
J Cell Biol ; 216(11): 3535-3549, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-28887438

RESUMO

A propensity for rewiring genetic and epigenetic regulatory networks, thus enabling sustained cell proliferation, suppression of apoptosis, and the ability to evade the immune system, is vital to cancer cell propagation. An increased understanding of how this is achieved is critical for identifying or improving therapeutic interventions. In this study, using acute myeloid leukemia (AML) human cell lines and a custom CRISPR/Cas9 screening platform, we identify the H3K9 methyltransferase SETDB1 as a novel, negative regulator of innate immunity. SETDB1 is overexpressed in many cancers, and loss of this gene in AML cells triggers desilencing of retrotransposable elements that leads to the production of double-stranded RNAs (dsRNAs). This is coincident with induction of a type I interferon response and apoptosis through the dsRNA-sensing pathway. Collectively, our findings establish a unique gene regulatory axis that cancer cells can exploit to circumvent the immune system.


Assuntos
Inativação Gênica , Interferon Tipo I/metabolismo , Leucemia Mieloide Aguda/enzimologia , Proteínas Metiltransferases/metabolismo , Retroelementos , Apoptose , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Sobrevivência Celular , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase , Humanos , Imunidade Inata , Interferon Tipo I/imunologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/patologia , Proteínas Metiltransferases/genética , Interferência de RNA , RNA de Cadeia Dupla/biossíntese , RNA de Cadeia Dupla/genética , Transdução de Sinais , Fatores de Tempo , Transfecção , Evasão Tumoral
5.
Cancer Immunol Res ; 5(6): 455-467, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28468914

RESUMO

A hypoxic tumor microenvironment is linked to poor prognosis. It promotes tumor cell dedifferentiation and metastasis and desensitizes tumor cells to type-I IFN, chemotherapy, and irradiation. The cytoplasmic immunoreceptor retinoic acid-inducible gene-I (RIG-I) is ubiquitously expressed in tumor cells and upon activation by 5'-triphosphate RNA (3pRNA) drives the induction of type I IFN and immunogenic cell death. Here, we analyzed the impact of hypoxia on the expression of RIG-I in various human and murine tumor and nonmalignant cell types and further investigated its function in hypoxic murine melanoma. 3pRNA-inducible RIG-I-expression was reduced in hypoxic melanoma cells compared with normoxic controls, a phenomenon that depended on the hypoxia-associated transcription factor HIF1α. Still, RIG-I functionality was conserved in hypoxic melanoma cells, whereas responsiveness to recombinant type-I IFN was abolished, due to hypoxia-induced loss of type I IFN receptor expression. Likewise, RIG-I activation in hypoxic melanoma cells, but not exposure to recombinant IFNα, provoked melanocyte antigen-specific CD8+ T-cell and NK-cell attack. Scavenging of hypoxia-induced reactive oxygen species by vitamin C restored the inducible expression of RIG-I under hypoxia in vitro, boosted in vitro anti-melanoma NK- and CD8+ T-cell attack, and augmented 3pRNA antitumor efficacy in vivo These results demonstrate that RIG-I remains operational under hypoxia and that RIG-I function is largely insensitive to lower cell surface expression of the IFNα receptor. RIG-I function could be fortified under hypoxia by the combined use of 3pRNA with antioxidants. Cancer Immunol Res; 5(6); 455-67. ©2017 AACR.


Assuntos
Hipóxia/metabolismo , Tolerância Imunológica , Melanoma/metabolismo , Receptores do Ácido Retinoico/metabolismo , Animais , Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Técnicas de Inativação de Genes , Humanos , Camundongos Endogâmicos C57BL , RNA/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptores do Ácido Retinoico/genética , Baço/citologia
6.
Sci Rep ; 6: 38405, 2016 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-27941826

RESUMO

Excessive inflammation can cause damage to host cells and tissues. Thus, the secretion of inflammatory cytokines is tightly regulated at transcriptional, post-transcriptional and post-translational levels and influenced by cellular stress responses, such as endoplasmic reticulum (ER) stress or apoptosis. Here, we describe a novel type of post-transcriptional regulation of the type-I-IFN response that was induced in monocytes by cytosolic transfection of a short immunomodulatory DNA (imDNA), a G-tetrad forming CpG-free derivative of the TLR9 agonist ODN2216. When co-transfected with cytosolic nucleic acid stimuli (DNA or 3P-dsRNA), imDNA induced caspase-3 activation, translational shutdown and upregulation of stress-induced genes. This stress response inhibited the type-I-IFN induction at the translational level. By contrast, the induction of most type-I-IFN-associated chemokines, including Chemokine (C-X-C Motif) Ligand (CXCL)10 was not affected, suggesting a differential translational regulation of chemokines and type-I-IFN. Pan-caspase inhibitors could restore IFN-ß secretion, yet, strikingly, caspase inhibition did not restore global translation but instead induced a compensatory increase in the transcription of IFN-ß but not CXCL10. Altogether, our data provide evidence for a differential regulation of cytokine release at both transcriptional and post-transcriptional levels which suppresses type-I-IFN induction yet allows for CXCL10 secretion during imDNA-induced cellular stress.


Assuntos
Quimiocina CXCL10/metabolismo , Interferon-alfa/metabolismo , Leucócitos Mononucleares/metabolismo , Células Cultivadas , Quimiocina CXCL10/genética , Sequência Rica em GC , Regulação da Expressão Gênica/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Interferon beta/genética , Interferon beta/metabolismo , Oligonucleotídeos/farmacologia , Biossíntese de Proteínas , Estresse Fisiológico
7.
ChemMedChem ; 11(20): 2272-2286, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27531666

RESUMO

2-Amino[1,2,4]triazolo[1,5-c]quinazolines were identified as potent adenosine receptor (AR) antagonists. Synthetic strategies were devised to gain access to a broad range of derivatives including novel polyheterocyclic compounds. Potent and selective A3 AR antagonists were discovered, including 3,5-diphenyl[1,2,4]triazolo[4,3-c]quinazoline (17, Ki human A3 AR 1.16 nm) and 5'-phenyl-1,2-dihydro-3'H-spiro[indole-3,2'-[1,2,4]triazolo[1,5-c]quinazolin]-2-one (20, Ki human A3 AR 6.94 nm). In addition, multitarget antagonists were obtained, such as the dual A1 /A3 antagonist 2,5-diphenyl[1,2,4]triazolo[1,5-c]quinazoline (13 b, Ki human A1 AR 51.6 nm, human A3 AR 11.1 nm), and the balanced pan-AR antagonists 5-(2-thienyl)[1,2,4]triazolo[1,5-c]quinazolin-2-amine (11 c, Ki human A1 AR 131 nm, A2A AR 32.7 nm, A2B AR 150 nm, A3 AR 47.5 nm) and 9-bromo-5-phenyl[1,2,4]triazolo[1,5-c]quinazolin-2-amine (11 q, Ki human A1 AR 67.7 nm, A2A AR 13.6 nm, A2B AR 75.0 nm, A3 AR 703 nm). In many cases, significantly different affinities for human and rat receptors were observed, which emphasizes the need for caution in extrapolating conclusions between different species.


Assuntos
Compostos Heterocíclicos/farmacologia , Antagonistas de Receptores Purinérgicos P1/síntese química , Antagonistas de Receptores Purinérgicos P1/farmacologia , Quinazolinas/farmacologia , Receptores Purinérgicos P1/metabolismo , Triazóis/farmacologia , Animais , Relação Dose-Resposta a Droga , Compostos Heterocíclicos/síntese química , Compostos Heterocíclicos/química , Humanos , Estrutura Molecular , Antagonistas de Receptores Purinérgicos P1/química , Quinazolinas/síntese química , Quinazolinas/química , Ratos , Relação Estrutura-Atividade , Triazóis/síntese química , Triazóis/química
8.
Nat Immunol ; 16(10): 1025-33, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26343537

RESUMO

Cytosolic DNA that emerges during infection with a retrovirus or DNA virus triggers antiviral type I interferon responses. So far, only double-stranded DNA (dsDNA) over 40 base pairs (bp) in length has been considered immunostimulatory. Here we found that unpaired DNA nucleotides flanking short base-paired DNA stretches, as in stem-loop structures of single-stranded DNA (ssDNA) derived from human immunodeficiency virus type 1 (HIV-1), activated the type I interferon-inducing DNA sensor cGAS in a sequence-dependent manner. DNA structures containing unpaired guanosines flanking short (12- to 20-bp) dsDNA (Y-form DNA) were highly stimulatory and specifically enhanced the enzymatic activity of cGAS. Furthermore, we found that primary HIV-1 reverse transcripts represented the predominant viral cytosolic DNA species during early infection of macrophages and that these ssDNAs were highly immunostimulatory. Collectively, our study identifies unpaired guanosines in Y-form DNA as a highly active, minimal cGAS recognition motif that enables detection of HIV-1 ssDNA.


Assuntos
DNA Complementar/química , DNA Viral/química , DNA Viral/imunologia , HIV-1/genética , HIV-1/imunologia , Interferon-alfa/imunologia , Nucleotidiltransferases/genética , Animais , Linhagem Celular , Células Cultivadas , DNA Complementar/genética , DNA Complementar/imunologia , DNA Viral/genética , Células HEK293 , Humanos , Imunização , Camundongos
9.
Immunity ; 43(1): 41-51, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26187414

RESUMO

The cytosolic helicase retinoic acid-inducible gene-I (RIG-I) initiates immune responses to most RNA viruses by detecting viral 5'-triphosphorylated RNA (pppRNA). Although endogenous mRNA is also 5'-triphosphorylated, backbone modifications and the 5'-ppp-linked methylguanosine ((m7)G) cap prevent immunorecognition. Here we show that the methylation status of endogenous capped mRNA at the 5'-terminal nucleotide (N1) was crucial to prevent RIG-I activation. Moreover, we identified a single conserved amino acid (H830) in the RIG-I RNA binding pocket as the mediator of steric exclusion of N1-2'O-methylated RNA. H830A alteration (RIG-I(H830A)) restored binding of N1-2'O-methylated pppRNA. Consequently, endogenous mRNA activated the RIG-I(H830A) mutant but not wild-type RIG-I. Similarly, knockdown of the endogenous N1-2'O-methyltransferase led to considerable RIG-I stimulation in the absence of exogenous stimuli. Studies involving yellow-fever-virus-encoded 2'O-methyltransferase and RIG-I(H830A) revealed that viruses exploit this mechanism to escape RIG-I. Our data reveal a new role for cap N1-2'O-methylation in RIG-I tolerance of self-RNA.


Assuntos
RNA Helicases DEAD-box/genética , Tolerância Imunológica/genética , Processamento Pós-Transcricional do RNA/genética , RNA/genética , Vírus da Febre Amarela/enzimologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Proteína DEAD-box 58 , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Histidina/genética , Humanos , Metilação , Metiltransferases/genética , Camundongos , Estrutura Terciária de Proteína , RNA/química , RNA/imunologia , RNA Viral/imunologia , Receptores Imunológicos , Vírus da Febre Amarela/genética
10.
PLoS One ; 8(4): e62872, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23653683

RESUMO

The innate immune system senses pathogens by pattern recognition receptors in different cell compartments. In the endosome, bacteria are generally recognized by TLRs; facultative intracellular bacteria such as Listeria, however, can escape the endosome. Once in the cytosol, they become accessible to cytosolic pattern recognition receptors, which recognize components of the bacterial cell wall, metabolites or bacterial nucleic acids and initiate an immune response in the host cell. Current knowledge has been focused on the type I IFN response to Listeria DNA or Listeria-derived second messenger c-di-AMP via the signaling adaptor STING. Our study focused on the recognition of Listeria RNA in the cytosol. With the aid of a novel labeling technique, we have been able to visualize immediate cytosolic delivery of Listeria RNA upon infection. Infection with Listeria as well as transfection of bacterial RNA induced a type-I-IFN response in human monocytes, epithelial cells or hepatocytes. However, in contrast to monocytes, the type-I-IFN response of epithelial cells and hepatocytes was not triggered by bacterial DNA, indicating a STING-independent Listeria recognition pathway. RIG-I and MAVS knock-down resulted in abolishment of the IFN response in epithelial cells, but the IFN response in monocytic cells remained unaffected. By contrast, knockdown of STING in monocytic cells reduced cytosolic Listeria-mediated type-I-IFN induction. Our results show that detection of Listeria RNA by RIG-I represents a non-redundant cytosolic immunorecognition pathway in non-immune cells lacking a functional STING dependent signaling pathway.


Assuntos
Células Epiteliais/metabolismo , Hepatócitos/metabolismo , Listeria monocytogenes/metabolismo , Monócitos/metabolismo , RNA Bacteriano/genética , Receptores do Ácido Retinoico/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Cultivadas , Citosol/metabolismo , Citosol/microbiologia , Células Epiteliais/citologia , Células Epiteliais/microbiologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Hepatócitos/citologia , Hepatócitos/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Interferon Tipo I/biossíntese , Interferon Tipo I/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Monócitos/citologia , Monócitos/microbiologia , Fosforilação , RNA Bacteriano/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores do Ácido Retinoico/antagonistas & inibidores , Receptores do Ácido Retinoico/genética , Transdução de Sinais
11.
PLoS One ; 6(7): e22389, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21811596

RESUMO

UNLABELLED: Lantibiotics are small peptide antibiotics that contain the characteristic thioether amino acids lanthionine and methyllanthionine. As ribosomally synthesized peptides, lantibiotics possess biosynthetic gene clusters which contain the structural gene (lanA) as well as the other genes which are involved in lantibiotic modification (lanM, lanB, lanC, lanP), regulation (lanR, lanK), export (lanT(P)) and immunity (lanEFG). The lantibiotic mersacidin is produced by Bacillus sp. HIL Y-85,54728, which is not naturally competent. METHODOLOGY/PRINCIPAL FINDINGS: The aim of these studies was to test if the production of mersacidin could be transferred to a naturally competent Bacillus strain employing genomic DNA of the producer strain. Bacillus amyloliquefaciens FZB42 was chosen for these experiments because it already harbors the mersacidin immunity genes. After transfer of the biosynthetic part of the gene cluster by competence transformation, production of active mersacidin was obtained from a plasmid in trans. Furthermore, comparison of several DNA sequences and biochemical testing of B. amyloliquefaciens FZB42 and B. sp. HIL Y-85,54728 showed that the producer strain of mersacidin is a member of the species B. amyloliquefaciens. CONCLUSIONS/SIGNIFICANCE: The lantibiotic mersacidin can be produced in B. amyloliquefaciens FZB42, which is closely related to the wild type producer strain of mersacidin. The new mersacidin producer strain enables us to use the full potential of the biosynthetic gene cluster for genetic manipulation and downstream modification approaches.


Assuntos
Bacillus/metabolismo , Bacteriocinas/metabolismo , Peptídeos/metabolismo , Bacillus/enzimologia , Bacillus/genética , Bacillus/imunologia , Bacteriocinas/biossíntese , Bacteriocinas/química , Bacteriocinas/isolamento & purificação , Sequência de Bases , DNA Girase/genética , Dados de Sequência Molecular , Família Multigênica/genética , Peptídeos/química , Peptídeos/isolamento & purificação , Filogenia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...