Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Annu Rev Immunol ; 35: 177-198, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28125358

RESUMO

The discovery of long noncoding RNAs (lncRNA) has provided a new perspective on gene regulation in diverse biological contexts. lncRNAs are remarkably versatile molecules that interact with RNA, DNA, or proteins to promote or restrain the expression of protein-coding genes. Activation of immune cells is associated with dynamic changes in expression of genes, the products of which combat infectious microorganisms, initiate repair, and resolve inflammatory responses in cells and tissues. Recent evidence indicates that lncRNAs play important roles in directing the development of diverse immune cells and controlling the dynamic transcriptional programs that are a hallmark of immune cell activation. The importance of these molecules is underscored by their newly recognized roles in inflammatory diseases. In this review, we discuss the contribution of lncRNAs in the development and activation of immune cells and their roles in immune-related diseases. We also discuss challenges faced in identifying biological functions for this large and complex class of genes.


Assuntos
Doenças do Sistema Imunitário/genética , Imunidade/genética , RNA Longo não Codificante/imunologia , Animais , Regulação da Expressão Gênica , Humanos
2.
Immunity ; 57(3): 446-461.e7, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38423012

RESUMO

In response to viral infection, how cells balance translational shutdown to limit viral replication and the induction of antiviral components like interferons (IFNs) is not well understood. Moreover, how distinct isoforms of IFN-induced oligoadenylate synthetase 1 (OAS1) contribute to this antiviral response also requires further elucidation. Here, we show that human, but not mouse, OAS1 inhibits SARS-CoV-2 replication through its canonical enzyme activity via RNase L. In contrast, both mouse and human OAS1 protect against West Nile virus infection by a mechanism distinct from canonical RNase L activation. OAS1 binds AU-rich elements (AREs) of specific mRNAs, including IFNß. This binding leads to the sequestration of IFNß mRNA to the endomembrane regions, resulting in prolonged half-life and continued translation. Thus, OAS1 is an ARE-binding protein with two mechanisms of antiviral activity: driving inhibition of translation but also a broader, non-canonical function of protecting IFN expression from translational shutdown.


Assuntos
Interferons , Oligorribonucleotídeos , Viroses , Animais , Humanos , Camundongos , Nucleotídeos de Adenina , Antivirais/farmacologia , 2',5'-Oligoadenilato Sintetase/genética , 2',5'-Oligoadenilato Sintetase/metabolismo
3.
Cell ; 165(7): 1672-1685, 2016 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-27315481

RESUMO

Long intergenic noncoding RNAs (lincRNAs) are important regulators of gene expression. Although lincRNAs are expressed in immune cells, their functions in immunity are largely unexplored. Here, we identify an immunoregulatory lincRNA, lincRNA-EPS, that is precisely regulated in macrophages to control the expression of immune response genes (IRGs). Transcriptome analysis of macrophages from lincRNA-EPS-deficient mice, combined with gain-of-function and rescue experiments, revealed a specific role for this lincRNA in restraining IRG expression. Consistently, lincRNA-EPS-deficient mice manifest enhanced inflammation and lethality following endotoxin challenge in vivo. lincRNA-EPS localizes at regulatory regions of IRGs to control nucleosome positioning and repress transcription. Further, lincRNA-EPS mediates these effects by interacting with heterogeneous nuclear ribonucleoprotein L via a CANACA motif located in its 3' end. Together, these findings identify lincRNA-EPS as a repressor of inflammatory responses, highlighting the importance of lincRNAs in the immune system.


Assuntos
Regulação da Expressão Gênica , Inflamação/genética , Macrófagos/imunologia , RNA Longo não Codificante/metabolismo , Animais , Cromátides/metabolismo , Deleção de Genes , Humanos , Listeria monocytogenes/fisiologia , Listeriose/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Longo não Codificante/genética , Infecções por Respirovirus/imunologia , Vírus Sendai/fisiologia , Receptores Toll-Like/metabolismo , Transcriptoma
4.
Cell ; 156(6): 1193-1206, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24630722

RESUMO

Inflammasomes elicit host defense inside cells by activating caspase-1 for cytokine maturation and cell death. AIM2 and NLRP3 are representative sensor proteins in two major families of inflammasomes. The adaptor protein ASC bridges the sensor proteins and caspase-1 to form ternary inflammasome complexes, achieved through pyrin domain (PYD) interactions between sensors and ASC and through caspase activation and recruitment domain (CARD) interactions between ASC and caspase-1. We found that PYD and CARD both form filaments. Activated AIM2 and NLRP3 nucleate PYD filaments of ASC, which, in turn, cluster the CARD of ASC. ASC thus nucleates CARD filaments of caspase-1, leading to proximity-induced activation. Endogenous NLRP3 inflammasome is also filamentous. The cryoelectron microscopy structure of ASC(PYD) filament at near-atomic resolution provides a template for homo- and hetero-PYD/PYD associations, as confirmed by structure-guided mutagenesis. We propose that ASC-dependent inflammasomes in both families share a unified assembly mechanism that involves two successive steps of nucleation-induced polymerization. PAPERFLICK:


Assuntos
Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/metabolismo , Inflamassomos/química , Sequência de Aminoácidos , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Transporte/metabolismo , Microscopia Crioeletrônica , Proteínas de Ligação a DNA , Humanos , Inflamassomos/metabolismo , Inflamassomos/ultraestrutura , Interleucina-1beta/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas Nucleares/metabolismo , Polimerização , Estrutura Terciária de Proteína
6.
Nat Methods ; 16(6): 489-492, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31133759

RESUMO

Modular domains of long non-coding RNAs can serve as scaffolds to bring distant regions of the linear genome into spatial proximity. Here, we present HiChIRP, a method leveraging bio-orthogonal chemistry and optimized chromosome conformation capture conditions, which enables interrogation of chromatin architecture focused around a specific RNA of interest down to approximately ten copies per cell. HiChIRP of three nuclear RNAs reveals insights into promoter interactions (7SK), telomere biology (telomerase RNA component) and inflammatory gene regulation (lincRNA-EPS).


Assuntos
Cromatina/química , Cromatina/genética , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica , RNA Longo não Codificante/genética , RNA/química , Telomerase/química , Animais , Células Cultivadas , Cromossomos , Células-Tronco Embrionárias/citologia , Genoma , Camundongos , Regiões Promotoras Genéticas , RNA/genética , Telomerase/genética
7.
J Immunol ; 204(2): 428-437, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31836654

RESUMO

Functional peptides encoded by short open reading frames are emerging as important mediators of fundamental biological processes. In this study, we identified a micropeptide produced from a putative long noncoding RNA (lncRNAs) that is important in controlling innate immunity. By studying lncRNAs in mice macrophages, we identified lncRNA 1810058I24Rik, which was downregulated in both human and murine myeloid cells exposed to LPS as well as other TLR ligands and inflammatory cytokines. Analysis of lncRNA 1810058I24Rik subcellular localization revealed that this transcript was localized in the cytosol, prompting us to evaluate its coding potential. In vitro translation with 35S-labeled methionine resulted in translation of a 47 aa micropeptide. Microscopy and subcellular fractionation studies in macrophages demonstrated endogenous expression of this peptide on the mitochondrion. We thus named this gene mitochondrial micropeptide-47 (Mm47). Crispr-Cas9-mediated deletion of Mm47, as well as small interfering RNA studies in mice primary macrophages, showed that the transcriptional response downstream of TLR4 was intact in cells lacking Mm47. In contrast, Mm47-deficient or knockdown cells were compromised for Nlrp3 inflammasome responses. Activation of Nlrc4 or Aim2 inflammasomes were intact in cells lacking Mm47. This study therefore identifies, to our knowledge, a novel mitochondrial micropeptide Mm47 that is required for the activation of the Nlrp3 inflammasome. This work further highlights the functional activity of short open reading frame-encoded peptides and underscores their importance in innate immunity.


Assuntos
Citosol/metabolismo , Inflamassomos/metabolismo , Macrófagos/fisiologia , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fragmentos de Peptídeos/metabolismo , RNA Longo não Codificante/genética , Animais , Sistemas CRISPR-Cas , Células Cultivadas , Humanos , Imunidade Inata/genética , Lipopolissacarídeos/imunologia , Camundongos , Mitocôndrias/genética , Fragmentos de Peptídeos/genética , RNA Interferente Pequeno/genética
8.
Nucleic Acids Res ; 48(21): 12055-12073, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33179750

RESUMO

Proper cytokine gene expression is essential in development, homeostasis and immune responses. Studies on the transcriptional control of cytokine genes have mostly focused on highly researched transcription factors (TFs) and cytokines, resulting in an incomplete portrait of cytokine gene regulation. Here, we used enhanced yeast one-hybrid (eY1H) assays to derive a comprehensive network comprising 1380 interactions between 265 TFs and 108 cytokine gene promoters. Our eY1H-derived network greatly expands the known repertoire of TF-cytokine gene interactions and the set of TFs known to regulate cytokine genes. We found an enrichment of nuclear receptors and confirmed their role in cytokine regulation in primary macrophages. Additionally, we used the eY1H-derived network as a framework to identify pairs of TFs that can be targeted with commercially-available drugs to synergistically modulate cytokine production. Finally, we integrated the eY1H data with single cell RNA-seq and phenotypic datasets to identify novel TF-cytokine regulatory axes in immune diseases and immune cell lineage development. Overall, the eY1H data provides a rich resource to study cytokine regulation in a variety of physiological and disease contexts.


Assuntos
Linhagem da Célula/imunologia , Citocinas/genética , Redes Reguladoras de Genes/imunologia , Linfócitos/imunologia , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Linhagem da Célula/genética , Citocinas/classificação , Citocinas/imunologia , Conjuntos de Dados como Assunto , Células Dendríticas/citologia , Células Dendríticas/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Ontologia Genética , Células HEK293 , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Linfócitos/classificação , Linfócitos/citologia , Macrófagos/citologia , Macrófagos/imunologia , Anotação de Sequência Molecular , Monócitos/citologia , Monócitos/imunologia , Cultura Primária de Células , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/imunologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Análise de Célula Única , Células THP-1 , Fatores de Transcrição/classificação , Fatores de Transcrição/imunologia , Transcrição Gênica , Técnicas do Sistema de Duplo-Híbrido
9.
J Immunol ; 196(2): 547-52, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26685207

RESUMO

With the stimulator of IFN genes (STING) C terminus being extensively studied, the role of the N-terminal domain (NTD) of STING remains an important subject of investigation. In this article, we identify novel mutations in NTD of Sting of the MOLF strain in response to HSV and Listeria monocytogenes both in vitro and in vivo. These mutations are responsible for low levels of IFN-ß caused by failure of MOLF STING to translocate from the endoplasmic reticulum. These data provide evidence that the NTD of STING affects DNA responses via control of trafficking. They also show that the genetic diversity of wild-derived mice resembles the diversity observed in humans. Several human alleles of STING confer attenuated IFN-I production similar to what we observe with the MOLF Sting allele, a crucial functional difference not apparent in classical inbred mice. Thus, understanding the functional significance of polymorphisms in MOLF STING can provide basic mechanistic insights relevant to humans.


Assuntos
Interferon Tipo I/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Alelos , Animais , DNA/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Microscopia Confocal , Mutação , Transporte Proteico/fisiologia
10.
J Immunol ; 195(4): 1359-63, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26179904

RESUMO

Natural antisense transcripts (NATs) are a class of long noncoding RNAs (lncRNAs) that are complementary to other protein-coding genes. Although thousands of NATs are encoded by mammalian genomes, their functions in innate immunity are unknown. In this study, we identified and characterized a novel NAT, AS-IL1α, which is partially complementary to IL-1α. Similar to IL-1α, AS-IL1α is expressed at low levels in resting macrophages and is induced following infection with Listeria monocytogenes or stimulation with TLR ligands (Pam3CSK4, LPS, polyinosinic-polycytidylic acid). Inducible expression of IL-1α mRNA and protein were significantly reduced in macrophages expressing shRNA that target AS-IL1α. AS-IL1α is located in the nucleus and did not alter the stability of IL-1α mRNA. Instead, AS-IL1α was required for the recruitment of RNA polymerase II to the IL-1α promoter. In summary, our studies identify AS-IL1α as an important regulator of IL-1α transcription during the innate immune response.


Assuntos
Regulação da Expressão Gênica , Mediadores da Inflamação , Interleucina-1alfa/genética , RNA Antissenso/genética , RNA não Traduzido/genética , Transcrição Gênica , Animais , Linhagem Celular , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Loci Gênicos , Ligantes , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Interferência de RNA , Receptores Toll-Like/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(21): 7765-70, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24828532

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) is an extracellular pathogen that causes hemorrhagic colitis and hemolytic uremic syndrome. The proinflammatory cytokine, interleukin-1ß, has been linked to hemolytic uremic syndrome. Here we identify the nucleotide-binding domain and leucine rich repeat containing family, pyrin domain containing 3 (NLRP3) inflammasome as an essential mediator of EHEC-induced IL-1ß. Whereas EHEC-specific virulence factors were dispensable for NLRP3 activation, bacterial nucleic acids such as RNA:DNA hybrids and RNA gained cytosolic access and mediated inflammasome-dependent responses. Consistent with a direct role for RNA:DNA hybrids in inflammasome activation, delivery of synthetic EHEC RNA:DNA hybrids into the cytosol triggered NLRP3-dependent responses, and introduction of RNase H, which degrades such hybrids, into infected cells specifically inhibited inflammasome activation. Notably, an E. coli rnhA mutant, which is incapable of producing RNase H and thus harbors increased levels of RNA:DNA hybrid, induced elevated levels of NLRP3-dependent caspase-1 activation and IL-1ß maturation. Collectively, these findings identify RNA:DNA hybrids of bacterial origin as a unique microbial trigger of the NLRP3 inflammasome.


Assuntos
Proteínas de Transporte/metabolismo , DNA de Cadeia Simples/metabolismo , Escherichia coli Êntero-Hemorrágica/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Inflamassomos/imunologia , Interleucina-1beta/imunologia , RNA/metabolismo , Animais , Sequência de Bases , Proteínas de Transporte/imunologia , Caspase 1/imunologia , DNA de Cadeia Simples/genética , Ensaio de Imunoadsorção Enzimática , Proteínas de Escherichia coli/genética , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Dados de Sequência Molecular , Proteína 3 que Contém Domínio de Pirina da Família NLR , RNA/genética , Proteínas Ribossômicas/genética
12.
J Biol Chem ; 289(34): 23568-81, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-25002588

RESUMO

The interferon γ-inducible protein 16 (IFI16) has recently been linked to the detection of nuclear and cytosolic DNA during infection with herpes simplex virus-1 and HIV. IFI16 binds dsDNA via HIN200 domains and activates stimulator of interferon genes (STING), leading to TANK (TRAF family member-associated NF-κB activator)-binding kinase-1 (TBK1)-dependent phosphorylation of interferon regulatory factor (IRF) 3 and transcription of type I interferons (IFNs) and related genes. To better understand the role of IFI16 in coordinating type I IFN gene regulation, we generated cell lines with stable knockdown of IFI16 and examined responses to DNA and RNA viruses as well as cyclic dinucleotides. As expected, stable knockdown of IFI16 led to a severely attenuated type I IFN response to DNA ligands and viruses. In contrast, expression of the NF-κB-regulated cytokines IL-6 and IL-1ß was unaffected in IFI16 knockdown cells, suggesting that the role of IFI16 in sensing these triggers was unique to the type I IFN pathway. Surprisingly, we also found that knockdown of IFI16 led to a severe attenuation of IFN-α and the IFN-stimulated gene retinoic acid-inducible gene I (RIG-I) in response to cyclic GMP-AMP, a second messenger produced by cyclic GMP-AMP synthase (cGAS) as well as RNA ligands and viruses. Analysis of IFI16 knockdown cells revealed compromised occupancy of RNA polymerase II on the IFN-α promoter in these cells, suggesting that transcription of IFN-stimulated genes is dependent on IFI16. These results indicate a broader role for IFI16 in the regulation of the type I IFN response to RNA and DNA viruses in antiviral immunity.


Assuntos
Vírus de DNA/imunologia , Interferon Tipo I/fisiologia , Proteínas Nucleares/fisiologia , Fosfoproteínas/fisiologia , Vírus de RNA/imunologia , Transcrição Gênica , Sequência de Bases , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Técnicas de Silenciamento de Genes , Inativação Gênica , Células HEK293 , Humanos , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Reação em Cadeia da Polimerase
13.
J Immunol ; 190(5): 1911-8, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23417527

RESUMO

Recognition of microbial nucleic acids is one strategy by which mammalian hosts respond to infectious agents. Intracellular DNA that is introduced into cells during infection elicits potent inflammatory responses by triggering the induction of antiviral type I IFNs and the maturation and secretion of inflammatory cytokines, such as TNF-α, IL-1ß, and IL-18. In addition, if nucleases, such as DNase II or DNase III (Trex1), fail to clear self-DNA, accumulated DNA gains access to intracellular compartments where it drives inflammatory responses leading to autoimmune disease. In this review, we discuss a rapidly evolving view of how cytosolic DNA-sensing machineries coordinate antimicrobial immunity and, if unchecked, lead to autoimmune disease.


Assuntos
Infecções Bacterianas/imunologia , DNA Bacteriano/imunologia , Sistema Imunitário/microbiologia , Imunidade Inata , Inflamação/imunologia , Animais , Autoimunidade , Bactérias/imunologia , Infecções Bacterianas/microbiologia , Citocinas/biossíntese , Citocinas/imunologia , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/imunologia , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno , Humanos , Inflamassomos/imunologia , Inflamação/microbiologia , Transdução de Sinais/imunologia
14.
J Biol Chem ; 287(30): 25216-29, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22654100

RESUMO

Francisella tularensis, the causative agent of tularemia, is one of the deadliest agents of biological warfare and bioterrorism. Extremely high virulence of this bacterium is associated with its ability to dampen or subvert host innate immune response. The objectives of this study were to identify factors and understand the mechanisms of host innate immune evasion by F. tularensis. We identified and explored the pathogenic role of a mutant interrupted at gene locus FTL_0325, which encodes an OmpA-like protein. Our results establish a pathogenic role of FTL_0325 and its ortholog FTT0831c in the virulent F. tularensis SchuS4 strain in intramacrophage survival and suppression of proinflammatory cytokine responses. This study provides mechanistic evidence that the suppressive effects on innate immune responses are due specifically to these proteins and that FTL_0325 and FTT0831c mediate immune subversion by interfering with NF-κB signaling. Furthermore, FTT0831c inhibits NF-κB activity primarily by preventing the nuclear translocation of p65 subunit. Collectively, this study reports a novel F. tularensis factor that is required for innate immune subversion caused by this deadly bacterium.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Francisella tularensis/imunologia , Francisella tularensis/patogenicidade , Imunidade Inata , Macrófagos/imunologia , Tularemia/imunologia , Fatores de Virulência/imunologia , Animais , Proteínas da Membrana Bacteriana Externa/genética , Linhagem Celular , Francisella tularensis/genética , Loci Gênicos/imunologia , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/imunologia , Tularemia/genética , Fatores de Virulência/genética
15.
J Biol Chem ; 286(47): 40536-47, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21976665

RESUMO

Activation of transcription factor NF-κB and inflammasome-directed caspase-1 cleavage of IL-1ß are key processes in the inflammatory response to pathogen or host-derived signals. Pyrin-only proteins (POPs) are restricted to Old World monkeys, apes, and humans and have previously been shown to impair inflammasome assembly and/or NF-κB p65 transcriptional activity in transfected epithelial cells. However, the biological role of POP2 and the molecular basis for its observed functions are not well understood. In this report we demonstrate that POP2 regulates TNFα and IL-1ß responses in human monocytic THP-1 cells and in stable transfectants of mouse J774A.1 macrophages. Deletion analysis of POP2 revealed that the first α-helix (residues 1-19) is necessary and sufficient for both inflammasome and NF-κB inhibitory functions. Further, key acidic residues Glu(6), Asp(8), and Glu(16), believed critical for Pyrin/Pyrin domain interaction, are important for inflammasome inhibition. Moreover, these mutations did not reduce the effect of POP2 upon NF-κB, indicating that the inflammasome and NF-κB inhibitory properties of POP2 can be uncoupled mechanistically. Collectively, these data demonstrate that POP2 acts as a regulator of inflammatory signals and exerts its two known functions through distinct modalities employed by its first α-helix.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Inflamassomos/metabolismo , Fator de Transcrição RelA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto/metabolismo , Francisella/fisiologia , Células HEK293 , Humanos , Interleucina-1beta/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Dados de Sequência Molecular , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Mutagênese , Mutação , Proteína 3 que Contém Domínio de Pirina da Família NLR , Estrutura Secundária de Proteína , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/genética , Ativação Transcricional/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
16.
J Biol Chem ; 286(45): 39033-42, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21930705

RESUMO

Pathogen-triggered activation of the inflammasome complex leading to caspase-1 activation and IL-1ß production involves similar sensor proteins between mouse and human. However, the specific sensors used may differ between infectious agents and host species. In mice, Francisella infection leads to seemingly exclusive activation of the Aim2 inflammasome with no apparent role for Nlrp3. Here we examine the IL-1ß response of human cells to Francisella infection. Francisella strains exhibit differences in IL-1ß production by influencing induction of IL-1ß and ASC transcripts. Unexpectedly, our results demonstrate that Francisella activates the NLRP3 inflammasome in human cells. Francisella infection of THP-1 cells elicits IL-1ß production, which is reduced by siRNA targeting of NLRP3. Moreover, in reconstituted 293T cells, Francisella triggers assembly of the NLRP3 inflammasome complex. In addition, inhibitors of reactive oxygen species, cathepsin B, and K(+) efflux pathways, known to specifically influence NLRP3, substantially but not completely impair the Francisella-elicited IL-1ß response, suggesting the involvement of another inflammasome pathway. Finally, shRNA targeting of NLRP3 and AIM2 reveals that both pathways contribute to the inflammasome response. Together these results establish NLRP3 as a cytosolic sensor for Francisella in human cells, a role not observed in mouse.


Assuntos
Proteínas de Transporte/metabolismo , Francisella tularensis/metabolismo , Inflamassomos/metabolismo , Tularemia/metabolismo , Animais , Proteínas de Transporte/genética , Catepsina B/genética , Catepsina B/metabolismo , Proteínas de Ligação a DNA , Células HEK293 , Humanos , Inflamassomos/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Transporte de Íons/genética , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Potássio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Especificidade da Espécie , Tularemia/genética
17.
BMC Evol Biol ; 11: 56, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21362197

RESUMO

BACKGROUND: Pyrin-only protein 2 (POP2) is a small human protein comprised solely of a pyrin domain that inhibits NF-κB p65/RelA and blocks the formation of functional IL-1ß processing inflammasomes. Pyrin proteins are abundant in mammals and several, like POP2, have been linked to activation or regulation of inflammatory processes. Because POP2 knockout mice would help probe the biological role of inflammatory regulation, we thus considered whether POP2 is common in the mammalian lineage. RESULTS: BLAST searches revealed that POP2 is absent from the available genomes of not only mice and rats, but those of other domestic mammals and New World monkeys as well. POP2 is however present in the genome of the primate species most closely related to humans including Pan troglodytes (chimpanzees), Macaca mulatta (rhesus macaques) and others. Interestingly, chimpanzee POP2 is identical to human POP2 (huPOP2) at both the DNA and protein level. Macaque POP2 (mqPOP2), although highly conserved is not identical to the human sequence; however, both functions of the human protein are retained. Further, POP2 appears to have arisen in the mammalian genome relatively recently (~25 mya) and likely derived from retrogene insertion of NLRP2. CONCLUSION: Our findings support the hypothesis that the NLR loci of mammals, encoding proteins involved in innate and adaptive immunity as well as mammalian development, have been subject to recent and strong selective pressures. Since POP2 is capable of regulating signaling events and processes linked to innate immunity and inflammation, its presence in the genomes of hominids and Old World primates further suggests that additional regulation of these signals is important in these species.


Assuntos
Evolução Molecular , Inflamassomos/metabolismo , Primatas/genética , Proteínas/genética , Fator de Transcrição RelA/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Clonagem Molecular , Bases de Dados Genéticas , Humanos , Imunidade Inata , Camundongos , Dados de Sequência Molecular , Filogenia , Proteínas/metabolismo , Pseudogenes , Ratos , Alinhamento de Sequência , Análise de Sequência de Proteína
18.
Nat Commun ; 8: 15564, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28580947

RESUMO

Pyrin domain-only proteins (POPs) are recently evolved, primate-specific proteins demonstrated in vitro as negative regulators of inflammatory responses. However, their in vivo function is not understood. Of the four known POPs, only POP2 is reported to regulate NF-κB-dependent transcription and multiple inflammasomes. Here we use a transgenic mouse-expressing POP2 controlled by its endogenous human promotor to study the immunological functions of POP2. Despite having significantly reduced inflammatory cytokine responses to LPS and bacterial infection, POP2 transgenic mice are more resistant to bacterial infection than wild-type mice. In a pulmonary tularaemia model, POP2 enhances IFN-γ production, modulates neutrophil numbers, improves macrophage functions, increases bacterial control and diminishes lung pathology. Thus, unlike other POPs thought to diminish innate protection, POP2 reduces detrimental inflammation while preserving and enhancing protective immunity. Our findings suggest that POP2 acts as a high-order regulator balancing cellular function and inflammation with broad implications for inflammation-associated diseases and therapeutic intervention.


Assuntos
Francisella/imunologia , Infecções por Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/prevenção & controle , Inflamação/imunologia , Tularemia/imunologia , Tularemia/prevenção & controle , Animais , Linhagem Celular , Feminino , Células HEK293 , Células HeLa , Humanos , Inflamassomos/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Listeria monocytogenes/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína 3 que Contém Domínio de Pirina da Família NLR/biossíntese , Neutrófilos/imunologia , Regiões Promotoras Genéticas/genética , Salmonella typhimurium/imunologia , Fator de Transcrição RelA/biossíntese , Tularemia/microbiologia , Células U937
19.
Trends Mol Med ; 20(11): 623-31, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25262537

RESUMO

All cells of the immune system rely on a highly integrated and dynamic gene expression program that is controlled by both transcriptional and post-transcriptional mechanisms. Recently, non-coding RNAs, including long non-coding RNAs (lncRNAs), have emerged as important regulators of gene expression in diverse biological contexts. lncRNAs control gene expression in the nucleus by modulating transcription or via post-transcriptional mechanisms targeting the splicing, stability, or translation of mRNAs. Our knowledge of lncRNA biogenesis, their cell type-specific expression, and their versatile molecular functions is rapidly progressing in all areas of biology. We discuss here these exciting new regulators and highlight an emerging paradigm of lncRNA-mediated control of gene expression in the immune system.


Assuntos
Regulação da Expressão Gênica , Sistema Imunitário/fisiologia , Imunidade/genética , RNA Longo não Codificante , Animais , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos
20.
Science ; 341(6147): 789-92, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23907535

RESUMO

An inducible program of inflammatory gene expression is central to antimicrobial defenses. This response is controlled by a collaboration involving signal-dependent activation of transcription factors, transcriptional co-regulators, and chromatin-modifying factors. We have identified a long noncoding RNA (lncRNA) that acts as a key regulator of this inflammatory response. Pattern recognition receptors such as the Toll-like receptors induce the expression of numerous lncRNAs. One of these, lincRNA-Cox2, mediates both the activation and repression of distinct classes of immune genes. Transcriptional repression of target genes is dependent on interactions of lincRNA-Cox2 with heterogeneous nuclear ribonucleoprotein A/B and A2/B1. Collectively, these studies unveil a central role of lincRNA-Cox2 as a broad-acting regulatory component of the circuit that controls the inflammatory response.


Assuntos
Regulação da Expressão Gênica , Imunidade Inata/genética , Inflamação/genética , Macrófagos/imunologia , Macrófagos/metabolismo , RNA Longo não Codificante/genética , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Ciclo-Oxigenase 2/genética , Citocinas/genética , Citocinas/metabolismo , Citosol/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Ativação de Macrófagos , Camundongos , Modelos Imunológicos , Interferência de RNA , RNA Longo não Codificante/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa